Thursday 12 May
10:00

"Thursday 12 May"

Added to your list of favorites
Deleted from your list of favorites
HOS
10:00 - 12:00

Hands on session & Gastruloids Workshop

Keynote Speakers: Lucille HOUYEL (Pediatric cardiologist) (Keynote Speaker, Le Plessis Robinson, France), Monique JONGBLOED (Keynote Speaker, Leiden, The Netherlands), Fabienne LESCROART (PI) (Keynote Speaker, Marseille, France)
Auditorium
13:00

"Thursday 12 May"

Added to your list of favorites
Deleted from your list of favorites
WOR
13:00 - 13:15

Welcome and opening remarks

Auditorium
13:15

"Thursday 12 May"

Added to your list of favorites
Deleted from your list of favorites
PS1
13:15 - 15:00

Session I
Early heart development

Moderators: Sigolene MEILHAC (PI) (PARIS, France), Miguel TORRES (Researcher) (Madrid, Spain)
13:15 - 15:00 #29326 - 001. Spatiotemporal sequence of endocardium and myocardium specification in the mammalian primitive heart tube.
001. Spatiotemporal sequence of endocardium and myocardium specification in the mammalian primitive heart tube.

The primitive heart tube is formed by two layers: the contractile myocardium and the endocardium that lines the inside of the chamber. Previous studies show that, at the onset of gastrulation in mouse, chicken and zebra fish, these two populations have already segregated. Nevertheless, when and how these lineages divert during embryogenesis remains unknown. Here, we use prospective and retrospective clonal analysis in mouse embryos to define the temporal sequence of cell fate specification. Our results confirm that endocardium and myocardium precursors have already segregated just after they undergo gastrulation (embryonic day E6.75). Besides, we observe that multipotent clones induced at E6.25-E.6.5 give rise to other mesoderm lineages apart from endocardium and myocardium. This shows segregation must take place just before or at gastrulation and that a bipotent progenitor exclusive to myocardium and endocardium does not exist, or it does only very transiently. To learn about the location and dynamics of these progenitors, we used long-term live imaging to track cardiovascular progenitors from the differentiated hear tube back to their positions at the primitive streak. We found that the original positions of both precursor populations are intermingled, suggesting they ingress the primitive streak simultaneously. Finding how this key fate decision takes place will help to understand cardiac congenital diseases as well as provide a tool to ambition new tissue engineering and regenerative strategies.


Miquel SENDRA (Madrid, Spain), Morena RAIOLA, Katie MCDOLE, Léo GUIGNARD, Jorge N. DOMÍNGUEZ, Miguel TORRES
13:15 - 15:00 #30489 - 002. A Mesp1-dependent developmental breakpoint in transcriptional and epigenomic specification of early cardiac precursors.
002. A Mesp1-dependent developmental breakpoint in transcriptional and epigenomic specification of early cardiac precursors.

Precise regulation of transcriptional networks governs emergence of early cardiac precursor cells (CPCs) within developing mesoderm during gastrulation. We leveraged detection of early cardiac lineage transgenes within a single cell RNA sequencing time course of whole mouse embryos to identify emerging CPCs and describe their transcriptional profiles during gastrulation prior to organogenesis.  Mesp1, a transcription factor (TF) transiently expressed in CPCs emerging from the primitive streak, has been described as an early regulator of cardiac specification. We observed a perdurance of cardiac transgene-expressing cells in Mesp1 mutants, albeit posterior-laterally located, prompting us to investigate how genetic programs for cardiogenesis and mesoderm specification potentially progress independently of Mesp1. Although mutant CPCs fail to robustly activate markers of cardiomyocyte maturity and TFs critical for heart morphogenesis, mutant CPCs express some structural myocyte genes and exhibit transcriptional profiles resembling those of cardiac mesoderm cells progressing towards cardiomyocyte fates. These results reveal Mesp1-independent aspects of early CPC specification and underscore Mesp1’s role in insuring subsequent progress through cardiogenesis. Single cell chromatin accessibility analysis of embryos at this developmental breakpoint identifies a required shift from mesendoderm transcriptional networks to cardiogenic networks in the designation of early-stage cardiac gene programs. Our investigation indicates that early CPCs are being specified distinctly from the mesoderm up until the Mesp1-deficient regulatory landscape inhibits further cardiac lineage maturation.   


Alexis Leigh KRUP (San Francisco, USA), Sarah A. B. WINCHESTER, Sanjeev S. RANADE, W. Patrick DEVINE, Deepak SRIVASTAVA, Benoit G. BRUNEAU
13:15 - 15:00 #30530 - 003. Precardiac mesoderm auto-regulates second heart field cell fate via Wnt secretion.
003. Precardiac mesoderm auto-regulates second heart field cell fate via Wnt secretion.

Congenital heart defects are often restricted to derivatives of two distinct groups of cells — the first and second heart fields (FHF & SHF). Thus, it is crucial to understand the mechanisms controlling their development. Wnt signaling has been implicated in SHF proliferation, however, the source of such Wnts remains unknown. Previously, we found upregulation of Wnts and Wnt receptor/target genes in the FHF and SHF, respectively, suggesting that cardiac progenitors may regulate SHF cell fate. To test this, we deleted Wntless (Wls), a gene required for Wnt secretion, in precardiac mesoderm. Deletion of Wls in Mesp1+ cells resulted in formation of a single-chambered heart with compromised SHF development. This phenotype was recapitulated by deleting Wls in pan-cardiac progenitors. However, no defects were observed when deleting Wls in SHF progenitors. To gain mechanistic insights, we isolated Mesp1-lineage cells and performed single-cell RNA-sequencing. We found that Wls deletion dysregulates developmental trajectories of SHF cells, marked by impaired proliferation. These results demonstrate a critical role of precardiac mesodermal Wnts in SHF fate decisions, identifying the crucial role of heart field coordinated development in chamber formation.


Matthew MIYAMOTO, Suraj KANNAN, Xihe LIU, David SUH, Myo HTET, Tejasvi KAKANI, Sean MURPHY, Emmanouil TAMPAKAKIS, Peter ANDERSEN, Hideki UOSAKI, Chulan KWON, Biyi LI (Baltimore, USA)
13:15 - 15:00 #30510 - 008. Tbx5-sensitive cues guide early progenitors for cardiac septation.
008. Tbx5-sensitive cues guide early progenitors for cardiac septation.

Many congenital heart defects (CHDs) involve incomplete septation of the atria or ventricles, in isolation or as part of more complex lesions, such as atrioventricular canal (AVC) defects. Early cardiac progenitor populations have been identified that contribute later to specific anatomical structures that are affected in CHDs. In the developing mouse heart, we found that a Tbx5+/Mef2cAHF+ progenitor lineage is intricately arranged into a band of cells at the interface of the first and second heart fields. This cohesive, coordinated population is found at a morphogenetic nexus, forming a compartment boundary bisecting the interventricular septum (IVS) and extending to the interatrial septum (IAS) via the cardiac cushions. Conditional ablation of these septal progenitors caused IVS disorganization, AVC defects and right ventricular chamber hypoplasia. Reduced dosage of the cardiac transcription factor TBX5 disrupted the integrity of the boundary, leading to lineage mixing, disrupted cell alignment and patterning defects, resulting in ventricular septal defects (VSDs), AVC defects and atrial septal defects. In the setting of reduced TBX5 dosage, we found that genes encoding guidance cues, best known for axonal guidance, were dysregulated. We further observed that loss of either guidance cue caused VSDs, suggesting that these TBX5-dependent signals are necessary for orchestrating cardiac septation. Thus, we identify essential morphogenetic cues that guide early progenitors for cardiac septation, revealing insights into the developmental origins of genetically-susceptible CHDs.


Irfan KATHIRIYA (San Francisco, USA), Martin DOMINGUEZ, W. Patrick DEVINE, Kavitha RAO, Kevin HU, Jonathon MUNCIE, Swetansu HOTA, Bayardo GARAY, Diego QUINTERO, Piyush GOYAL, Sarah WINCHESTER, Benoit BRUNEAU
13:15 - 15:00 #30629 - 005. Nkx2.7 is a novel regulator of pharyngeal arch development.
005. Nkx2.7 is a novel regulator of pharyngeal arch development.

The clinical phenotypes associated with DiGeorge Syndrome illustrate a developmental link between cardiovascular and craniofacial morphogenesis. Fate mapping studies in mice and zebrafish support this notion given the identification of a multipotent progenitor in the cardiopharyngeal field that gives rise to the heart, branchiomeric muscles, and pharyngeal arch (PA) arteries. NKX2-5 is a key cardiac transcription factor associated with human congenital heart disease and mouse models of Nkx2-5 deficiency highlight critical roles in cardiac development. nkx2.5 and nkx2.7 are two NKX2-5 homologs expressed in zebrafish cardiomyocytes and PAs. We demonstrate that Nkx2.7 serves as a previously unappreciated, crucial regulator of the first and second PA derivatives. Our previous studies show that nkx2.7 is required in the late-differentiating cardiac progenitors to maintain ventricular identity. While expression of nkx2.7 can function redundantly in this role, nkx2.5 is unable to compensate for nkx2.7 in developing PA1- and PA2-derived anterior and posterior mandibular and interhyoid muscles and cartilage elements. Taken together, these results underscore the unique function of Nkx2.7 in regulating pharyngeal arch morphogenesis. To identify targets that are exclusively regulated by Nkx2.7, we employed cardiac-specific bulk RNA-seq in wild-type, nkx2.5-/-, and nkx2.5-/-;nkx2.7-/-  embryos and single cell RNA-seq data from microdissected PA tissues in wild-type and nkx2.7-/- embryos. Our complex computational analysis uncovers barx1 and nkx3.2 as downstream effectors of Nkx2.7 in mediating muscle and cartilage development. Through examination of the tissue-specific functions and targets of Nkx2.7, we expect to reveal exciting new pathways responsible for branchiomeric muscle and craniofacial abnormalities in patients.


Caitlin FORD (New York, USA), Carmen DE SEMA TOMÁS, Cynthia GAO, Uday RANGASWAMY, Michael SEE, Hieu NIM, Remo SANGES, Mirana RAMIALISON, Kimara TARGOFF
Auditorium
15:00 Break
15:30

"Thursday 12 May"

Added to your list of favorites
Deleted from your list of favorites
PS2
15:30 - 17:15

Session II
CHD Mechanisms I

Moderators: Antonio BALDINI (Professor) (Napoli, Italy), Didier STAINIER (Director) (Bad Nauheim, Germany)
15:30 - 17:15 #30484 - 006. Nuclear envelope integrity is essential for proper cardiac development.
006. Nuclear envelope integrity is essential for proper cardiac development.

Nuclear envelope integrity is essential for compartmentalisation of nucleus and cytoplasm. Importantly, mutations in nuclear envelope-encoding genes are the second-highest cause of familial dilated cardiomyopathy. One such nuclear envelope protein that causes cardiomyopathy in humans and affects mouse heart development is Lem2. However, its role in heart remains poorly understood.

We generated mice in which Lem2 was specifically ablated either in embryonic cardiomyocytes (Lem2 cKO) or adult cardiomyocytes (Lem2 iCKO) and carried out detailed physiological, tissue and cellular analyses. High resolution episcopic microscopy was used for 3D reconstructions and detailed morphological analyses. RNA-sequencing and immunofluorescence identified altered pathways and cellular phenotypes, and cardiomyocytes were isolated to interrogate nuclear integrity in more detail. In addition, echocardiography provided physiological assessment of Lem2 iCKO adult mice. 

We found that Lem2 was essential for cardiac development, and hearts from Lem2 cKO mice were morphologically and transcriptionally underdeveloped. Lem2 cKO hearts displayed high levels of DNA damage, nuclear rupture, and apoptosis. Crucially, we found that these defects were driven by muscle contraction as they were ameliorated by inhibiting myosin contraction and L-type calcium channels.

Our data suggest that Lem2 is critical for integrity at the nascent nuclear envelope in fetal hearts, and protects the nucleus from the mechanical forces of muscle contraction. In contrast, the adult heart is not detectably affected by Lem2 loss, perhaps owing to a more established nuclear envelope and increased adaptation to mechanical stress. Taken together, these data provide insights into mechanisms underlying cardiomyopathy in patients with mutations in Lem2 and cardio-laminopathies in general.


Jacob ROSS, Matthew STROUD (London, United Kingdom)
15:30 - 17:15 #29417 - 007. Dissecting the cell type-specific roles of Hand2 during cardiac development in zebrafish.
007. Dissecting the cell type-specific roles of Hand2 during cardiac development in zebrafish.

Cardiogenesis requires the integration of diverse cardiac cell types including the myocardial and endocardial cells that form the initial layers of the heart. Differentiation, patterning, and morphogenesis of cardiomyocytes depend on the transcription factor Hand2. However, it is unclear how Hand2 regulates these early developmental events. To investigate this question, we first performed single-cell RNA sequencing on hand2 expressing cells from the cardiac cone and linear heart tube stages, and found increased expression of immature cardiomyocyte markers and concomitantly decreased expression of mature cardiomyocyte markers in hand2 mutants. In addition to these expected phenotypes, we observed an increased percentage of endothelial cells in hand2 mutants. To investigate the roles of hand2 in the myocardial and endothelial cells, we generated a floxed allele of hand2 for conditional inactivation. Knocking out hand2 in myl7 expressing cells (i.e., cardiomyocytes) led to a late phenotype whereby the trabeculae mostly fail to form. Interestingly, knocking out hand2 in kdrl expressing cells (i.e., endothelial cells) resulted in cardia bifida, recapitulating the global hand2 mutant phenotype. Transcriptional analysis of the endothelial cells revealed that the loss of Hand2 leads to a dysregulation of PDGF signaling. Notably, overexpressing pdgfra in endothelial cells could partially rescue the cardia bifida phenotype in hand2 mutants.  Together, these data indicate that during cardiac development, Hand2 serves critical functions not only in cardiomyocytes but also in endothelial cells to drive cardiomyocyte specification and also direct the migration of myocardial progenitors towards the midline. 


Yanli XU (Bad Nauheim, Germany), Stefan GUENTHER, Guilherme VALENTE, Mario LOOSO, Didier STAINIER
15:30 - 17:15 #29481 - 009. Multimodal Single Cell Analysis Reveals Regulatory Mechanisms Underlying Cell Signaling Defects in DiGeorge Syndrome.
009. Multimodal Single Cell Analysis Reveals Regulatory Mechanisms Underlying Cell Signaling Defects in DiGeorge Syndrome.

Communication between myriad cell types during development underlies proper organ morphogenesis. During cardiac development, reciprocal signaling between diverse cells is essential and disruption leads to congenital heart malformations; however, mechanistic interrogation of temporally dynamic gene networks and cis regulatory elements in signaling and receiving cells has been limited. Here, we integrated single cell chromatin accessibility (scATAC-seq) and transcriptomics (scRNA-seq) over multiple stages to provide a comprehensive epigenomic landscape of diverse cell types including mesodermal progenitors and neural crest cells in the developing mouse heart. Integrated multiomics and machine learning methods defined cis regulatory elements as enhancers for genes initiating or responding to intercellular signaling. Disruption of TBX1, a transcription factor that functions non-cell autonomously in mesodermal cardiac progenitors to affect neighboring neural crest-derived cells, causes morphogenetic defects of the cardiac outflow tract in humans and offered an opportunity to determine consequences of dysregulated signaling at single cell resolution. In mice lacking Tbx1, broad closure of chromatin regions enriched in Tbx and multiple cardiac progenitor TF motifs within a narrow subset of mesodermal progenitors correlated with diminished transcription of multiple signaling factors such as FGF, retinoic acid, Wnt, Tgfb and Semaphorins. In response, we identified subsets of cardiac and craniofacial neural crest cells containing differentially accessible chromatin regions that suggested a failure of differentiation. Lastly, we found aberrant persistence of neural crest cells with markers of multipotency within the pharyngeal region. This study provides an atlas of spatiotemporally dynamic regulatory elements in cardiogenesis and a mechanistic framework for how disruptions in cell-cell communication affect morphogenetic decisions at single cell resolution.


Sanjeev RANADE (San Francisco, USA), Sean WHALEN, Ivana ZLATANOVA, Lin YE, Benjamin VAN SOLDT, Tomohiro NISHINO, Angelo PELONERO, Langley Grace WALLACE, Pawel PRZYTYCKI, Casey GIFFORD, Brian BLACK, Katie POLLARD, Deepak SRIVASTAVA
15:30 - 17:15 #30554 - 010. Single-cell transcriptomics uncovers a Tbx1-dependent genetic program controlling cardiac neural crest cell deployment and progression.
010. Single-cell transcriptomics uncovers a Tbx1-dependent genetic program controlling cardiac neural crest cell deployment and progression.

Cardiac neural crest cells (cNCCs) are required for outflow tract (OFT) septation and in pharyngeal arch arteries (PAAs) development. Inactivation of Tbx1, the gene for 22q11.2 deletion syndrome, results in failure of cNCCs deployment leading to congenital heart disease. We aim to elucidate the cell fate transitions of the cNCCs during their deployment to the OFT and PAAs in normal development and that depends on Tbx1. We performed single-cell RNA-sequencing (scRNA-seq) of NCCs from control and Tbx1 null mouse embryos. We discovered the transcriptional signature that defines cNCC populations and investigated the gene expression dynamics that regulates cNCC fate progression into smooth muscle cells of the OFT and PAAs. We discovered three distinct cNCCs populations that emerge between embryonic day (E)9.5 and E10.5, reflecting transcriptional heterogeneity of cNCCs. Our trajectory analysis indicates that cells expressing Tbx2 and Tbx3 transition to a population that expresses Isl1 and Gata3 that then differentiates into smooth muscle of the OFT. Tbx2/3 cNCCs also form smooth muscle cells of the PAAs and we found that both Tbx2 and Tbx3 in cNCCs are necessary for aortic arch branching development. In the absence of Tbx1, multiple genes are dysregulated in the Tbx2/3 population and the Isl1/Gata3 population is strongly reduced in size, reflecting a lineage progression failure. Comparative analysis of our scRNA-seq data from control and Tbx1 null embryos indicates an abnormal upregulation of BMP signaling in Tbx1 null embryos. BMP signaling overactivation could, in part, explain failed cNCC deployment and progression in Tbx1 null embryos.


Christopher DE BONO (New York, USA), Yang LIU, Alexander FERRENA, Deyou ZHENG, Bernice MORROW
15:30 - 17:15 #30609 - 004. Dissecting Mechanisms of Chamber-Specific Cardiac Differentiation and its Perturbation Following Retinoic Acid Exposure.
004. Dissecting Mechanisms of Chamber-Specific Cardiac Differentiation and its Perturbation Following Retinoic Acid Exposure.

Proper heart development requires specification and differentiation of multiple progenitor populations, and dysregulation of these processes can lead to congenital heart defects (CHDs). Furthermore, different forms of CHDs may be driven by defects in distinct progenitor subtypes, who’s heterogeneity remains incompletely understood. To understand the transcriptomic landscape of the developing heart we performed single-cell RNA sequencing (scRNASeq) at the cardiac crescent (E8.25), primitive heart tube (E8.75) and late heart tube (E9.25) stages using Foxa2-Cre;mTmG embryos, allowing us to label atrial/ventricular fated cells prior to and during chamber morphogenesis. Through RNA velocity and lineage trajectory tools we identify heart field progenitors in multiple differentiation states, and uncover the top dynamically regulated genes for each cell type, which represent putative drivers of cell-state transitions during differentiation. We find that clustering of myocardial cell types occurs primarily based on heart field progenitor origin, and that different progenitor populations contribute to ventricular or atrial identity through separate differentiation mechanisms. Furthermore, we find that differentiation of anterior or posterior second heart field (SHF) cells occurs through deployment of separate components of the cardiac gene-regulatory network. Lastly, we show that in utero exposure to exogenous retinoic acid (RA), which plays a role in atrial chamber specification and acts as a teratogen during development, causes defects in ventricular chamber size. scRNASeq of RA-exposed embryos demonstrated dysregulation in FGF signaling in anterior SHF cells and a shunt in differentiation towards formation of head mesenchyme, and defects in cell-cycle exit in myocardial progenitors. These data demonstrate the utility of comparative scRNAseq studies for understanding lineage relationships during development and revealing cell-specific sensitivity to perturbations.


David GONZALEZ (New York, USA), Nadine SCHRODE, Tasneem A.m EBRAMIN, Nicolas BROGUIERE, Guiliana ROSSI, Lika DRAKHLIS, Robert ZWEIGERDT, Matthias P. LUTOLF, Kristin G. BEAUMONT, Robert SEBRA, Nicole DUBOIS
Auditorium
17:15

"Thursday 12 May"

Added to your list of favorites
Deleted from your list of favorites
MAG
17:15 - 17:45

Memorial Adriana Gittenberger-de Groot

Chairperson: Robert KELLY (PI) (Chairperson, Marseille, France)
Speaker: Robert POELMANN (Speaker, Leiden, The Netherlands)
Auditorium
17:45

"Thursday 12 May"

Added to your list of favorites
Deleted from your list of favorites
PO1
17:45 - 20:00

Poster session I (even number posters)

17:45 - 20:00 #29336 - 048. Conserved transcription factors promote cell fate stability and restrict reprogramming potential in differentiated cells.
048. Conserved transcription factors promote cell fate stability and restrict reprogramming potential in differentiated cells.

The characterization of novel mechanisms safeguarding cell fate identity in differentiated cells is crucial to improve our understanding of 1) how differentiation is maintained in healthy tissues or altered in a disease state and 2) the basic mechanisms governing cell fate reprogramming and our ability to use this technology for regenerative purposes. Here, we report on the first identification of a generic and transcription factor-mediated mechanism controlling cell fate stability in differentiated cells via the concomitant regulation of chromatin accessibility and transcription of genes required for large-scale phenotypic changes. Importantly, resulting knowledge could be translated in vivo, as the targeted inhibition of these new fate-stabilizers could significantly improve direct cardiac reprogramming-mediated heart repair post-myocardial infarction in adult mice.


Maria MISSINATO, Sean MURPHY, Michaela LYNOTT, Yu-Ling CHANG, Pier Lorenzo PURI, Chulan KWON, Peter ADAMS, Li QIAN, Alessandra SACCO, Peter ANDERSEN, Alexandre COLAS (LA JOLLA, USA)
17:45 - 20:00 #29443 - 050. Comparison of the regenerative capacity of seven wild-type zebrafish strains reveals inter-strain variations in the wound healing process, cardiomyocyte proliferation and apoptosis levels following ventricular cryoinjury.
050. Comparison of the regenerative capacity of seven wild-type zebrafish strains reveals inter-strain variations in the wound healing process, cardiomyocyte proliferation and apoptosis levels following ventricular cryoinjury.

Teleost fish maintain the capacity to regenerate lost cardiac tissue throughout adulthood. However, recent reports suggest that both inter- and intra-species variations exist. Based on these studies, we hypothesised that there is strain-dependent variability in zebrafish heart regeneration. To test this, we characterised the regenerative response of seven wild-type zebrafish strains (AB, NA, SAT, TL, TU, WIK, KCL). Analysing morphology, wound size and composition, apoptosis, proliferation and gene expression, we identified significantly different characteristics between the NA, TL, TU and WIK strains during regeneration. The WIK strain showed increased proliferation in cardiomyocytes bordering the wound at 7dpci . Accordingly, RNA-seq showed upregulation of cell cycle and DNA replication-related genes at 7dpci in the WIK compared to the other strains, resulting in the strongest wound size reduction between 7- and 21dpci. However, no further reduction was observed between 21- and 90dpci, with scar remaining in all WIK hearts at 90dpci, likely caused by elevated ECM gene expression and increased collagen presence in the 7dpci WIK wound. Correlation analysis indeed identified collagen deposition at 7dpci as a predictor of the 90dpci regenerative outcome. At 90dpci, the NA line showed the highest percentage of scar-free hearts whereas 80% of the TU hearts failed to regenerate the compact wall. Comparing gene expression between the TU and NA showed a reduced activation of the metabolic genes required for regeneration in the TU after injury. In conclusion, comparing seven wild-type zebrafish strains has allowed us to identify correlations between different cellular processes occurring during regeneration.


Konstantinos LEKKOS (Oxford, United Kingdom), Zhilian HU, Jana KOTH, Madeleine LEMIEUX, Katherine BANECKI, Helen POTTS, Gennaro RUGGIERO, Mathilda MOMMERSTEEG
17:45 - 20:00 #29527 - 052. Examining the interaction between metabolism and heart regeneration in Mexican cavefish.
052. Examining the interaction between metabolism and heart regeneration in Mexican cavefish.

Unlike humans, certain species possess a robust ability to regenerate their hearts. We have identified the Astyanax mexicanus fish as a unique model for heart regeneration research. While Astyanax surface fish regenerate their heart after injury, their cave-dwelling counterparts cannot and, like humans, form a permanent scar. This model provides an opportunity for direct comparison of natural regeneration versus scarring within a single species. 

We have previously shown that there are similar levels of cardiomyocyte proliferation at the wound border zone in both surface fish and cavefish. However, BrdU pulse-chase revealed a higher number of BrdU-positive cardiomyocytes in surface fish compared to cavefish which may be a result of defective cytokinesis resulting in a failure to regenerate. Indeed, examining cavefish cardiomyocytes showed higher DNA content in comparison to the surface fish heart. 

Using RNA-seq to investigate mechanisms underlying cardiomyocyte proliferation in Astyanax hearts after injury, we identified metabolism-related genes among those most differentially expressed. Specifically, glycolytic genes were highly upregulated in surface fish border zone cardiomyocytes. Moreover, glucose levels were reduced in the cavefish heart after injury. Seahorse metabolic assays revealed a reduction in anaerobic glycolysis in the cavefish heart after injury but no difference in oxygen consumption rates or fatty acid utilisation between the fish. This was accompanied by reduced ATP in cavefish hearts compared to surface fish after injury. Inhibiting glucose metabolism in surface fish using 2-deoxy-d-glucose increased cardiomyocyte DNA content showing a similar failure in cell cycle progression to the cavefish heart.

Overall, this suggests an important role for glucose metabolism in cardiomyocyte cell cycle progression, and that inability to maintain sufficient ATP in the cavefish heart might be linked to their inability to support a regenerative response.


Rita ALONAIZAN (Oxford, United Kingdom), William STOCKDALE, Helen POTTS, Madeleine LEMIEUX, Konstantinos LEKKOS, John WALSBY-TICKLE, Mathilda MOMMERSTEEG
17:45 - 20:00 #30249 - 054. Novel Role of Proteoglycan Sulfation as a Barrier to Direct Cardiac Reprogramming.
054. Novel Role of Proteoglycan Sulfation as a Barrier to Direct Cardiac Reprogramming.

Direct cardiac reprogramming (CR) of fibroblasts into induced cardiomyocytes (iCMs) represents a promising therapeutic avenue to promote heart repair post injury. Our lab previously identified four transcription factors playing a conserved role as barriers to CR. Remarkably, knock-down of these barriers increases CR up to six-fold as compared to MGT (MEF2C; GATA4; TBX5) overexpression alone. Next, mechanistic and functional, exploration of downstream transcriptional targets led to the identification of SULF2 and CHST2 as novel regulators of CR, indicating a potential novel role for proteoglycan sulfation-modifying enzymes as regulators of cell fate reprogramming. In this context, functional screening of all genes involved in the regulation of PG modification and sulfation in CR assays, identified CHST7 as most potent and evolutionarily conserved barrier to CR. Collectively, these data confirm that PG sulfation plays a previously unrecognized role in the regulation of CR and cell fate stability in differentiated cells.


Michaela LYNOTT (San Diego, California, USA), Alexandre COLAS
17:45 - 20:00 #30320 - 056. Interleukin 4 and 13 Signaling in Macrophages Regulates Cardiac Regeneration and Capillary Morphology in Neonatal Mice.
056. Interleukin 4 and 13 Signaling in Macrophages Regulates Cardiac Regeneration and Capillary Morphology in Neonatal Mice.

Heart failure (HF) is a prevalent disease, projected to affect over 8 million Americans by 2030. Disease burden continues to be high, despite therapy. Thus, new strategies that target HF progression should be developed. Our lab employs the neonatal mouse model of heart regeneration to identify reparative pathways to apply them to HF. We previously showed that deletion of Interleukin 13 (IL13) or its receptor, IL4Rα, impair heart regeneration. However, the cell types mediating this response are not known. IL13 shares the receptor IL4Rα with IL4 and both cytokines are known to polarize macrophages into a pro-reparative phenotype. Here, we hypothesize that IL4/13 signaling to macrophages promotes neonatal heart regeneration after myocardial infarction (MI) and we explore the cell types producing IL13/IL4 during cardiac regeneration. We generated a genetic model whereby IL4Rα is depleted in macrophages by crossing IL4Rα floxed (IL4Rαfl/fl) mice with transgenic mice whereby Cre recombinase is driven by the CX3CR1 promoter (CX3CR1Cre). We performed MI on P1 mice and assessed cardiac function by ultrasonography 21 days post-injury. We found that Cre-positive mice had a lower ejection fraction compared to Cre-negative controls. Additionally, we found smaller capillaries in the myocardium of Cre-positive mice. To identify the cellular source of IL4 and IL13 in the heart we used fluorescent reporter mouse lines; IL4-green fluorescent protein and IL13-yellow fluorescent protein. In homeostasis, we found IL4 expression in innate lymphoid cells (ILCs) and T cells. Following P1 MI, IL13 was expressed primarily in ILCs and IL4 in ILCs and myeloid cells. This suggests a novel role for ILCs during cardiac regeneration. In addition, we found that lack of IL4/13 signaling in macrophages impairs cardiac function and capillary morphology after MI in neonatal mice. Future studies will be aimed to identify the downstream signaling in macrophages that mediates this reparative response.


Santiago ALVAREZ-ARGOTE (Milwaukee, USA), Caitlin C. O'MEARA
17:45 - 20:00 #30422 - 058. Parasympathetic Axon Development, Disease, and Neuroregeneration in the Cardiac Ventricles.
058. Parasympathetic Axon Development, Disease, and Neuroregeneration in the Cardiac Ventricles.

The heart relies on opposing signals from sympathetic and parasympathetic nerves to guide developmental, homeostatic, and repair functions. However, the cardiac innervation patterning that regulates these essential functions has been relatively unexplored. This is partially a result of technical limitations, as histological sectioning prevented nerve morphology to be accurately examined. In this work, we are analyzing the spatial patterning of cardiac innervation by employing organ clearing, whole-mount staining, and three-dimensional (3D) imaging techniques. 

 

We are particularly interested in mapping the parasympathetic nerves, which are thought to only innervate the atria and nodes; however, their contribution to ventricular innervation has not been analyzed in whole-mount and 3D. Excitingly, our results demonstrate for the first time that dense parasympathetic axon bundles innervate the cardiac ventricles. Moreover, preliminary data demonstrates that parasympathetic and sympathetic nerve fibers are intertwined in the ventricles through a developmental patterning mechanism that we are currently exploring. 

 

The intricate neuronal networks are susceptible to injury and fatal misfiring following an adult myocardial infarction (MI). Remarkably, an MI in the neonatal mouse results in robust regeneration and restoration of autonomic functions. Our lab discovered that neonatal heart regeneration is regulated by nerve signaling. Here, we demonstrate reinnervation of the regenerating myocardium following injury in contrast to non-regenerating hearts. Moreover, these newly formed nerves exhibit significant crossover with smooth muscle positive arteries, suggesting potential physiological restoration of neurovascular coupling. The results of our research will expand our knowledge of the development and plasticity of the cardiac nervous system during homeostasis, disease, and regeneration. 


Rebecca SALAMON (Madison, USA), Ahmed MAHMOUD
17:45 - 20:00 #30442 - 060. Cell-reprogramming and myocardial regeneration in a pig model of right-ventricular failure.
060. Cell-reprogramming and myocardial regeneration in a pig model of right-ventricular failure.

Heart failure is a major burden to our societies. Besides patients with left ventricular dysfunction following ischemic insults or hypertension, progress in pediatric surgery to repair cardiac malformations has led to a growing population of now-adult congenital heart diseases (CHD) patients. These patients with right ventricular (RV) failure are left without any efficient pharmacological therapeutic approach. Cell therapy has been an option to regenerate the myocardium although the mechanism of action of such an approach remains questionable.

Here, we used human embryonic stem cell-derived cardiac Nkx2.5+ progenitor cells seeded in a collagen-based patch covering the whole RV to regenerate failing RV of a pig model of repaired tetralogy of Fallot. We report that these cells migrate within the myocardium while reversing the interstitial fibrosis. They then engraft and fully differentiate into small clusters of fetal-like human myocytes within the myocardium. Degradation of the fibrotic extracellular matrix triggers an inflammatory reaction involving resident macrophages releasing cytokines in the neighborhood of myocytes. This leads to the activation of pig myocytes' inflammasome and their Nfkb-dependent reprogramming into Oct4+ cells. The reprogrammed myocytes redifferentiate and proliferate around human myocytes. Altogether, the graft of human CPC triggers a reprogramming of pig myocytes and in turn endogenous regeneration and improved contractility of the RV. 


Virginie LAMBERT (Paris), Ambre DELERIS, Fahd TIBOURTINE, Virginie FOUILLOUX, Pauline BRIDGE, Michel PUCEAT
17:45 - 20:00 #30445 - 062. Antigen presentation is critical for zebrafish cardiac regeneration.
062. Antigen presentation is critical for zebrafish cardiac regeneration.

Post-ischemic tissue remodeling involves the formation of a permanent scar that impairs cardiac function.  Unlike mammals, zebrafish can regenerate its heart by forming new functional tissues without a persistent scar.  The severity of tissue damage and the regenerative capacity depend on the quality, extent and temporal dynamics of the immune response.  However, how the immune system regulates regeneration, particularly how the antigen presentation-adaptive immunity axis plays a role in this process, remains to be elucidated.

In the present study, scRNA-Seq revealed a strong antigen presentation signature in the injured zebrafish heart, with distinct antigen-presenting cell clusters at different stages of the regenerative process.  Therefore, we hypothesized a role for the adaptive immune response during cardiac regeneration.  Indeed, we observed the infiltration of T-cells, especially Cd4+ T-cells, within the injured tissue at intermediate stages of regeneration.  Genetic targeting of Cd74, a key component of MHC class II antigen presentation, dampened regeneration as evidenced by impaired cardiomyocyte cell cycle re-entry and defective scar formation/resolution.  Pharmacological inhibition of T-cell activation further confirmed these data.  Importantly, these defects were accompanied by changes in T-cell populations and by an overall faulty immune response.  Further transcriptomic and immunohistochemical analyses revealed the involvement of the MAPK signaling pathway in the regulation of the regenerative response by antigen presentation.

Our work highlights the importance of antigen presentation in modulating the spectrum of regenerative/reparative outcomes post-cardiac injury.  Ultimately, these data will help identify mechanisms with the potential to ameliorate adverse remodeling and improve key aspects of mammalian heart regeneration.


João CARDEIRA-DA-SILVA (Bad Nauheim, Germany), Stephan LATTING, Stefan GÜNTHER, Michail YEKELCHYK, Bo HU, Janita MINTCHEVA, Philipp JUNKER, Didier STAINIER
17:45 - 20:00 #30469 - 064. 14-3-3 binding motif phosphorylation disrupts Hdac4 organized inhibitory condensates to stimulate cardiac reprogramming.
064. 14-3-3 binding motif phosphorylation disrupts Hdac4 organized inhibitory condensates to stimulate cardiac reprogramming.

Limited understanding of the molecular mechanisms of induced cardiomyocyte (iCM) reprogramming is a key obstacle preventing its effective clinical applications. We report here the identification of a phosphorylation code in 14-3-3 binding motifs (PC14-3-3) that greatly stimulates iCM formation. PC14-3-3 is identified in pivotal functional proteins for iCM reprogramming, including transcription factors and epigenetic factors. Akt1 kinase and PP2A phosphatase are a key writer and eraser of the PC14-3-3 code, respectively and PC14-3-3 activation induces iCM formation with only Tbx5 but without Mef2c and Gata4. In contrast, PC14-3-3 inhibition by mutagenesis or inhibitor-mediated code removal abolishes reprogramming. We further discover that key PC14-3-3 embedded factors, such as Mef2c, Nrip1, and Foxo1, form inhibitory nuclear condensates with Hdac4 under hypo-phosphorylation state and PC14-3-3 activation disrupts these condensates to promote cardiac gene expression. This study provides a framework in decoding post-translational modifications for cell reprogramming and organ regeneration.


Liu LIU (Ann Arbor, USA), Zhong WANG
17:45 - 20:00 #30488 - 066. Role of endothelial Tal1 during zebrafish heart regeneration: from single cell transcriptomic analysis to a functional study.
066. Role of endothelial Tal1 during zebrafish heart regeneration: from single cell transcriptomic analysis to a functional study.

Objectives: Unlike mammals, zebrafish are able to regenerate their heart after an injury at all stages of life. It has been identified that this process involves cardiomyocyte dedifferentiation/proliferation, but the implication of interstitial cell types during heart regeneration remains poorly understood. Our study aims to describe the non-myocyte cell types present in the heart during regeneration in adult zebrafish as well as focus on the functional role of endothelial Tal1 expression in this process.

Methods: We performed single cell RNA sequencing (scRNAseq) on unamputated, 3-, 7- and 14-days post amputation (dpa) zebrafish hearts to provide a transcriptomic analysis of the zebrafish regenerating heart. We identified genes of interest from the dataset obtained. We then used a zebrafish conditional line expressing a dominant negative isoform of Tal1 (DNTal1) specifically in endothelial cells. After validating our DNTal1 line with a developmental characterization, adult fish were amputated and their hearts were extracted at different timepoints to be fixed and stained in order to assess their regenerative abilities.  

Results: Using scRNAseq, we described the different interstitial cell types present in the zebrafish heart and the changes they undergo during heart regeneration. We observed an increase in the number of Tal1 expressing endothelial cells during heart regeneration compared to unamputated controls. After validating our zebrafish DNTal1 line showing it exerts cardiac developmental defects already described in the literature, we saw DNTal1 expression inhibited heart regeneration primarily by disrupting the formation of the regenerating vascular plexus.

Conclusion: We showed Tal1 expression is essential for the endothelial regenerative response and its inactivation leads to a failure of regeneration.


Laura ROLLAND (Montpellier), Alenca HARRIGNTON, Adèle FAUCHERRE, Jourdano MANCILLA ABAROA, Thomas MOORE-MORRIS, Chris JOPLING
17:45 - 20:00 #30512 - 070. A small molecule screen identifies novel activators of epithelial to mesenchymal transition in human epicardial cells.
070. A small molecule screen identifies novel activators of epithelial to mesenchymal transition in human epicardial cells.

Upon ischemic cardiac injury, the epicardium, the outer layer of the heart which is essential for cardiac development, becomes re-activated and displays reparative potential. In this process, epicardial epithelial-to-mesenchymal transition (epiMT) is an essential step. To understand and increase epicardial activation, we aim to identify novel epiMT-inducing pathways by performing a small molecule screen.

Primary human epicardial cells were derived from human heart auricles. These epicardial derived cells (EPDCs) were cultured as epithelial-like cells maintaining a cobblestone morphology, and could be induced to undergo EMT by adding e.g. TGFbeta. Using this cell culture model, a phenotypic screen was performed on epithelial-like EPDCs using the LOPAC1280 small molecule library to identify epiMT-inducing compounds. The screen was performed 3 times to exclude patient variability. EpiMT was confirmed using αSMA-positive immunostaining as a hallmark for a phenotypic switch to a mesenchymal cell.
After validation of the positive hits, five compounds were selected that reproducibly induced epiMT, as shown by: 1) a phenotypic switch from epithelial (cobble) to mesenchymal (spindle-shaped) cells, 2) a decrease in CHD1 expression, and 3) an increase in EMT-related transcription factors (Snail, Slug) and mesenchymal markers (a-SMA, PSTN). To further establish a potential mechanism, cells were treated for 3 hours with the two most promising compounds and were subjected to RNA sequencing. These results suggest novel EMT regulators in human epicardium which are currently being validated.

In conclusion, high-throughput experiments using human primary epicardial derived cells to identify novel epiMT-inducing compounds is feasible. Using this model, we have identified several novel inducers of epiMT.
This work is funded by the DHF (2017T059, to AMS)


Esther DRONKERS, Tessa VAN HERWAARDEN, Esmee GROENEVELD, Marie-Jose GOUMANS, Anke SMITS (Leiden, The Netherlands)
17:45 - 20:00 #30535 - 072. FGF10 gene transfer therapy promotes cardiac regeneration and repair.
072. FGF10 gene transfer therapy promotes cardiac regeneration and repair.

The stimulation of terminally differentiated cardiomyocyte proliferation represents one of the main therapeutic approaches for heart regeneration and repair. We uncovered a role for the Fibroblast Growth Factor 10 (FGF10) signalling in regulating both fetal and adult cardiomyocyte proliferation. Using Fgf10 gain and loss of function mouse models together with experimental mouse model of myocardial infarction (MI) we demonstrated FGF10 regenerative potential.

Our study aims to determine the clinical relevance of FGF10 therapy for heart regeneration. Thus, using AAV strategy, we aim to evaluate Fgf10 gene transfer therapeutic impact after myocardial infarction (MI).

We first aimed to determine the most efficient route of delivery for AAV9 administration by considering short- and long-term heart expression but also side organ targeting. We thus analyzed the spatiotemporal pattern of Fgf10 expression following AAV9-Fgf10 injection (retro-orbital or tail vein) with 1011 viral particles of either AAV9-GFP or AAV9-Fgf10Our results reveal that tail-vein administration route is more appropriated for a rapid, long term and efficient cardiac FGF10 treatment, with a pic of Fgf10 expression at 3 weeks post injection and a maintained expression 10 weeks post-injection. In order to investigate the impact of AAV9-Fgf10 administration following MI on cardiac function and remodeling, mice subjected to MI were tail vein-injected with 1011 particles of either AAV9-GFP or AAV9-Fgf10 at the time of surgery. Our preliminary experiments indicate that AAV9-Fgf10 treatment promotes cardiomyocyte proliferation and prevents cardiac fibrosis infiltration. Moreover, echocardiographic measurements show that AAV9-Fgf10 treatment preserves cardiac remodeling and function post-MI.

Altogether, our results are thus of particular interest regarding the use of AAV9-Fgf10 strategy as a clinical perspective to promote heart repair following myocardial injury.


Fabien HUBERT (Marseille), Sandy PAYAN, Francesca ROCHAIS
17:45 - 20:00 #30544 - 074. Interplay between calcium cycling and sarcomeres directs cardiomyocyte redifferentiation and maturation during regeneration.
074. Interplay between calcium cycling and sarcomeres directs cardiomyocyte redifferentiation and maturation during regeneration.

A promising strategy to repair the injured mammalian heart is by the induction of cardiomyocyte dedifferentiation followed by their proliferation. Thus, there is now a focus on identifying factors and pathways that can drive the production of new cardiomyocytes through proliferation. However, a largely underappreciated concept is the redifferentiation and maturation of cardiomyocytes following proliferation.  This becomes particularly prominent as current methods to induce cardiomyocyte proliferation often result in cardiomegaly due to uncontrolled cell numbers. Unlike mammals, the zebrafish adult heart can robustly regenerate its heart following injury.  Here, we have studied zebrafish heart regeneration to discover an essential mechanism that restricts proliferation and stimulates the redifferentiation and maturation of cardiomyocytes after injury. Using an ex vivo imaging technique that we developed, we observed differences in intracellular calcium dynamics in cardiomyocytes located at the wound border zone, where cardiomyocyte proliferation is induced. These changes in calcium dynamics provided evidence that after a period of strong proliferation, cardiomyocytes redifferentiate and mature. An adaptor protein lrrc10 was identified to be able to specifically halt proliferation and promote cardiomyocyte redifferentiation. Finally, these maturation mechanisms were similarly observed in mouse and human iPSC-derived cardiomyocytes. Hence demonstrating that rather than being a passive process, redifferentiation is actively regulated and highlights the importance on also determining its underlying mechanisms in order to generate fully functional cardiomyocytes.


Phong NGUYEN (Utrecht, The Netherlands), Iris GOOIJERS, Mara BOUWMAN, Jeroen BAKKERS
17:45 - 20:00 #30547 - 076. Live imaging of zebrafish cardiac slices to study heart regeneration.
076. Live imaging of zebrafish cardiac slices to study heart regeneration.

The adult zebrafish is able to fully regenerate its heart after injury, making it an attractive model to identify and study mechanisms that regulate heart regeneration. In addition, a wide range of transgenic tools are available for zebrafish which allows the live imaging of cellular events. Thus far, live imaging in adult zebrafish have been hampered by the opacity and movement of the heart. In addition, ex vivo cultures don’t allow for deep tissue imaging and show rapid cell death. To overcome these issues, we present the use of cardiac slices of the zebrafish heart to study heart regeneration ex vivo. Cardiac slices can be cultured for several days while retaining characteristics of the in vivo heart. We have used these slices to perform live imaging on proliferating adult cardiomyocytes within their native tissue context for the first time. Currently, we are expanding the use of cardiac slice cultures to study the dynamics of calcium cycling and energy metabolism to get a better understanding of the role of these processes during heart regeneration. In conclusion, culturing cardiac slices of the adult zebrafish heart allows us to gain temporal insights of cardiomyocyte biology during physiological and pathological conditions. 


Iris GOOIJERS, Hessel HONKOOP (Utrecht, The Netherlands), Phong NGUYEN, Jeroen BAKKERS
17:45 - 20:00 #30567 - 078. Distinct epicardial gene regulatory programmes drive development and regeneration of the zebrafish heart.
078. Distinct epicardial gene regulatory programmes drive development and regeneration of the zebrafish heart.

Unlike the adult mammalian heart, which has limited regenerative capacity, the zebrafish heart can fully regenerate following injury. Reactivation of cardiac developmental programmes is considered key to successfully regenerating the heart, yet the regulatory elements underlying the response triggered upon injury and during development remain elusive. Organ-wide activation of the epicardium is essential for zebrafish heart regeneration and is considered a potential regenerative source to target in the mammalian heart. Here we compared the transcriptome and epigenome of the developing and regenerating zebrafish epicardium by integrating gene expression profiles with open chromatin ATAC-seq data. By generating gene regulatory networks associated with epicardial development and regeneration, we inferred genetic programmes driving each of these processes, which were largely distinct. We identified wt1a, wt1b, and the AP-1 subunits junbb, fosab and fosb as central regulators of the developing network, whereas hif1ab, zbtb7a, tbx2b and nrf1 featured as putative central regulators of the regenerating epicardial network. By interrogating developmental gene regulatory networks that drive cell-specific transcriptional heterogeneity, we tested novel subpopulation-related epicardial enhancers in vivo. Taken together, our work revealed striking differences between the regulatory blueprint deployed during epicardial development and regeneration. These findings challenge the dogma that heart regeneration is essentially a reactivation of developmental programmes, and provide important insights into epicardial regulation that can assist in developing therapeutic approaches to enable tissue regeneration in the adult mammalian heart.


Michael WEINBERGER, Filipa SIMOES (Oxford, United Kingdom), Tatjana SAUKA-SPENGLER, Paul RILEY
17:45 - 20:00 #30569 - 080. The role of RUNX1 in CM cell cycle activity and its impact on cardiac regeneration.
080. The role of RUNX1 in CM cell cycle activity and its impact on cardiac regeneration.

Factors responsible for cardiomyocyte (CM) proliferation may serve as a potential therapeutic to stimulate endogenous myocardial regeneration. It is established that RUNX1 induction in CMs increases after injury. Here, we examine the effect of RUNX1 on CM cell cycle and establishment of the presumed proliferative-competent Mononuclear Diploid CM population (MNDCM) during postnatal development and cardiac regeneration using both CM-specific gain-and loss of function mouse models. We hypothesize that RUNX1 overexpression (OE) increases CM cell cycle activity with expansion of the MNDCMs, thereby extending the neonatal regenerative window and positively impacting adult cardiac remodeling post injury. 

During postnatal development, RUNX1 KO decreased postnatal CM cell cycle activity, while RUNX1 OE extended the period of cell cycle activation. This extension observed in RUNX1 OE mice is complete with cytokinesis resulting in an expansion of the MNDCMs and total CM endowment. To determine whether RUNX1 could similarly regulate CM cell cycle in a regenerative and non-regenerative model, we induce P6 and 8-week MIs to measure cell cycle activity and cardiac function post injury. RUNX1 OE neonatal mice with a P6 MI displayed no difference in CM cell cycle activity 7 days post injury compared to control littermates. However, RUNX1 OE in adult mice with an 8-week MI showed increased CM cell cycle activity with completion of cytokinesis 2 weeks post injury and limited improvement in cardiac function 28 days post injury. RUNX1 influences CM cell cycle activation in the context of normal postnatal development and adult MI. Conversely, this phenomenon does not appear to translate to the neonatal injury context. We are examining this possible discrepancy with further experiments to fully understand the role of RUNX1 in CM cell cycle activity and its impact on cardiac regeneration.


Kaelin AKINS (Milwaukee, USA), Samantha SWIFT, Mary KOLELL, Michael FLINN, PHD, Caitlin O'MEARA, PHD, Michaela PATTERSON, PHD
17:45 - 20:00 #30580 - 082. Macrophages regulate cardiomyocyte repopulation of fibrotic tissue during zebrafish heart regeneration.
082. Macrophages regulate cardiomyocyte repopulation of fibrotic tissue during zebrafish heart regeneration.

The last two decades of research have significantly increased our understanding of mechanisms regulating zebrafish heart regeneration.  However, an aspect that remains largely underexplored is how newly proliferated cardiomyocytes invade and repopulate fibrotic tissue following cryoinjury.  We have previously shown that blocking the function of AP-1 transcription factors leads to defects in regeneration, partly due to a defect in cardiomyocyte protrusion and invasion into injured tissue.

 

Here, we show that cardiomyocyte protrusion peaks at 10 days post cryoinjury, shortly following the peak in cardiomyocyte proliferation.  Furthermore, using live imaging of myocardial slices, we observe that border zone (BZ) cardiomyocytes are dynamic, extending multiple protrusions into the collagen-containing injured area.  This dynamic behavior is accompanied by collagen degradation by BZ cardiomyocytes, which may be regulated by Mmp14b, a membrane-tethered matrix metalloprotease with collagen-degrading ability.  In addition, we find that macrophages are very closely associated with protruding cardiomyocytes at the BZ.  In irf8 mutants, which lack macrophages, we observe a decrease in the length of cardiomyocyte protrusions while the number of extended protrusions remains unchanged.  Single-cell RNA-seq of BZ cells suggests that both macrophages and cardiomyocytes contribute to regulation of the BZ microenvironment to promote invasion of cardiomyocytes to replenish lost tissue following injury.

 

Altogether, our data reveal a crosstalk between BZ cardiomyocytes and macrophages to facilitate repopulation of injured tissue by proliferating cardiomyocytes.  Detailed understanding of these mechanisms may have therapeutic implications, e.g., to promote the engraftment of exogenous cardiomyocytes following myocardial infarction in the mammalian heart.


Arica BEISAW, Arica BEISAW (Heidelberg, Germany), Julia DALLMANN, Stefan GUENTHER, Till LAUTENSCHLAEGER, Didier Yr STAINIER
17:45 - 20:00 #30595 - 084. Modest upregulation of Tbx5 stimulates prenatal ventricular cardiomyocyte proliferation.
084. Modest upregulation of Tbx5 stimulates prenatal ventricular cardiomyocyte proliferation.

Heart development and rhythm control are highly Tbx5 dosage-sensitive. TBX5 haploinsufficiency causes congenital heart defects and conduction disorders, whereas slightly increased levels of TBX5 in human heart samples have been associated with atrial fibrillation. To gain insight into the impact of slight dosage changes of Tbx5 in vivo, we deleted the mouse orthologue of a conserved atrial fibrillation-associated regulatory region in the TBX5 locus (RE(int)-/- mice). Postnatal RE(int)-/- mouse atria showed slightly increased Tbx5 expression levels (30%) and increased susceptibility to atrial arrhythmia. Strikingly, we observed increased Tbx5 (30%) in the ventricles before birth, accompanied by increased heart size. We observed an increase in fetal cardiomyocyte proliferation rates specific to the left ventricle. Transcriptional profiling of ventricles of fetal control and RE(int)-/- mice revealed induction of several cell cycle related genes and Prrx1, a transcription factor associated with atrial fibrillation. When expression of Prrx1 was reduced by introducing a Prrx1 enhancer deletion in RE(int)-/- mice, we observed a normalization of heart size. These data indicate prenatal cardiomyocyte proliferation rates are Tbx5 dosage sensitive and act in part through moderating Prrx1 levels. To examine the potential of Tbx5 to induce postnatal cardiomyocyte renewal, AAV9-TNNT2-TBX5 was retro-orbitally administered to juvenile mice. Preliminary results show cardiomyocyte-specific expression of TBX5, and induction of target genes (Gja5, Nppa) and genes involved in proliferation. Future experiments will focus on the capacity of Tbx5 to induce cardiomyocyte proliferation and the underlying mechanisms.


Fernanda M. BOSADA, Alexandra GIOVOU (Amsterdam, The Netherlands), Bastiaan BOUKENS, Gerard J.j BOINK, Monika GLADKA, Vincent M. CHRISTOFFELS
17:45 - 20:00 #30635 - 086. Investigating the role of endocardial cell heterogeneity and EndoMT in the regenerative zebrafish heart.
086. Investigating the role of endocardial cell heterogeneity and EndoMT in the regenerative zebrafish heart.

Cardiovascular diseases account for nearly 18 million deaths per year, and majority of these deaths occur due to myocardial infarction (MI).  To augment cardiac repair and regeneration following MI, mechanistic insights derived from the regenerative zebrafish heart are considered to be of high translational value.  Unlike the mammalian heart, the zebrafish heart is able to restore lost cardiomyocytes (CMs) through mechanisms that allow spared cardiomyocytes to dedifferentiate, proliferate, and repopulate the injured tissue.  This regenerative ability is attributed to a stringent spatiotemporal regulation and crosstalk between the various cardiac cell types.  Therefore, for an effective bench-to-bedside application of cardio-regenerative strategies, it is imperative that the distinct roles and spatiotemporal dynamics of the injury-responsive cardiac cell populations in zebrafish are well characterized.  

The injury-responsive endocardial cells remain poorly characterized.  Since they provide direct structural support, and may activate several signaling pathways in the CMs, we aim to further investigate their role in facilitating cardiac regeneration in adult zebrafish.  Using single-cell and bulk transcriptomic approaches on the ET(krt4:EGFPsqet33-1A) line, in which endocardial cells express GFP, we have identified the injury-specific endocardial sub-populations enriched for EndoMT (serpine1, vim), and immune-cell (lgals2a) genes. Expression of these genes was mapped to the injury area and wound-border zone interface using the available Tomo-seq data for injured hearts.  In addition, several markers of mature endothelial/endocardial cells were identified, and observed to be repressed in these clusters.  These data corroborate recent findings in mouse that show EndoMT in endothelial cells of the infarcted hearts. Thus, we will perform functional studies on a few identified candidates, aiming to identify factors that promote CM dedifferentiation and/or proliferation.


Pooja SAGVEKAR (Bad Nauheim, Hesse, Germany), Stefan GÜNTHER, Khrievono KIKHI, Didier STAINIER
17:45 - 20:00 #30564 - 088. Duty cycle of cyclic stretch induces syncytial organization and functional maturation of biomimetic fetal ventricular tissue.
088. Duty cycle of cyclic stretch induces syncytial organization and functional maturation of biomimetic fetal ventricular tissue.

Afterload, also known as resistance to contraction, and preload, also known as stretch, are key mechanical stimulation parameters that influence ventricular maturation but are difficult to test in vivo. Current in vitro mechanical stimulation bioreactors can mimic preload via cyclically stretching cardiac tissue but fail to simultaneously mimic afterload due to the usage of high afterload anchorage points. Therefore, we developed a mechanical stimulation bioreactor system that better recapitulates the developmental cardiac cycle. This system can apply duty cycles that vary the afterload and preload during the stretch regimen, which can be used to assess how these mechanical parameters affect maturation of biomimetic cardiac tissues. To determine the optimal stage to construct biomimetic cardiac tissues, we conducted single cell RNA sequencing on embryonic chicken ventricular cells spanning four key ventricular development stages. This data revealed embryonic day 7 as an important ventricular maturation transitional stage where epicardial-derived cells are actively contributing to the compacting myocardium. Embryonic day 7 ventricular biomimetic tissues were created and cultured statically for seven days followed by three different stimulation regimens for seven additional days: static, cyclic stretch without a duty cycle, or cyclic stretch with a duty cycle. Cyclically stretched biomimetic tissues maintained their overall spontaneous contractile function and led to significant increases in contractile frequency and mechanical stiffness compared to static controls. Strikingly, biomimetic tissues conditioned with a duty cycle led to the greatest improvement in cardiomyocyte elongation, alignment, and sarcomere organization. This data demonstrates duty cycles are a driver of ventricular developmental maturation.


Gaetano SCUDERI, Jonathan BUTCHER (Ithaca, USA), Brianna HOU, Kathleen CLIFFORD
17:45 - 20:00 #30711 - 090. Massive Expansion of Functional Human iPSC-derived Cardiomyocytes by Concomitant Glycogen Synthase Kinase-3 Beta Inhibition and Removal of Cell-Cell Contact.
090. Massive Expansion of Functional Human iPSC-derived Cardiomyocytes by Concomitant Glycogen Synthase Kinase-3 Beta Inhibition and Removal of Cell-Cell Contact.

Therapeutic induction of cardiomyocyte proliferation in vivo by modulating Wnt, Hippo, neuregulin, and other signaling pathways has shown significant promise to treat congenital and acquired heart diseases. However, when these signaling molecules are applied in vitro to expand cardiomyocytes (CMs) from pluripotent stem cells, the extent of proliferation has generally been modest (<5 fold), precluding the use of these cells for industrial-scale applications. Here, we demonstrate the massive expansion of beating human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in vitro by glycogen synthase kinase-3 beta (GSK-3β) inhibition using CHIR99021 (CHIR), a small molecule GSK-3β antagonist, together with the removal of contact inhibition by low-density serial passaging, resulting in a total of 100-250-fold increase in CM number. Prevention of cell-cell contact stimulated hiPSC-CM cell cycle activity, providing 10 to 25-times greater expansion beyond GSK-3β inhibition alone. To disambiguate the role of Wnt/b-catenin signaling on hiPSC-CM proliferation upon CHIR-treatment and low-density passaging, we found that the maintenance of a more immature sarcomeric structure and gene expression was dependent on LEF/TCF transcriptional activity while cell cycle activation was regulated by cell contact-mediated AKT phosphorylation. To demonstrate the functionality of expanded hiPSC-CM, we engineered heart tissues and showed that the contractile performance is equivalent between expanded and unexpanded hiPSC-CMs. In summary, we uncovered a molecular interplay between GSK-3β inhibition and cell-cell contact that enables the massive expansion of functional hiPSC-CMs thereby facilitating large-scale drug screening efforts and engineering of therapeutically relevant-sized cardiac tissues.


Jan Willem BUIKEMA (Amsterdam, The Netherlands), Soah LEE, Sean M. WU
17:45 - 20:00 #29415 - 092. Building the muscular wall in the cardiac atrium involves cell elongation and reorganization of tissue polarity.
092. Building the muscular wall in the cardiac atrium involves cell elongation and reorganization of tissue polarity.

During vertebrate development, cardiomyocytes undergo cellular rearrangements to form complex myocardial structures.  Malformations of these structures lead to diseases that include cardiomyopathies and cardiac arrythmias.  Previous studies have mostly focused on the formation of the trabecular network in the ventricle; however, morphogenetic processes that drive atrial myocardial complexity, which is crucial to propagate the action potential for cardiac contraction, have largely been overlooked.  Our study uses zebrafish larvae to elucidate cardiomyocyte behaviors during atrial development, as they allow for high-resolution live imaging and are easily amenable to genetic manipulations.

Using live 3D confocal imaging of zebrafish hearts, combined with mosaic labelling and temporal tracking of individual atrial cardiomyocytes, we found that atrial myocardial morphogenesis is driven by complex cell behaviors.  Specifically, we observed that atrial cardiomyocytes in zebrafish larvae form membrane protrusions and adopt an elongated shape in a non-stochastic orientation that establishes atrial tissue-level polarity.  These shape changes lead to restricted multilayering between neighboring cardiomyocytes, and the formation of new cell contacts, resulting in populations of elongated cardiomyocytes that span the atrium in an orientation parallel to the direction of blood flow.  These cell behaviors lead to the appearance of muscle ridges on the inner surface of the atrium.  Notably, these atrial cardiomyocyte behaviors appear to be independent from factors important in ventricular morphogenesis such as Erbb2 and Notch signaling.  Altogether, these data suggest that atrial morphogenesis is driven by oriented cell elongation as well as by distinct molecular and environmental/physical factors, all of which need further investigation.

 


Marga ALBU (Bad Nauheim, Germany), Felix GUNAWAN, Rashmi PRIYA, Alessandra GENTILE, Didier STAINIER
17:45 - 20:00 #29456 - 094. Egr3 regulates atrioventricular valve formation in zebrafish.
094. Egr3 regulates atrioventricular valve formation in zebrafish.

Congenital heart disease (CHD) is the leading cause of death from congenital anomalies, with 7.4% of the cases caused by abnormal development of the atrioventricular (AV) canal. The AV canal gives rise to the atrioventricular valves and node as well as the septae in mammalian hearts, all of which are essential for heart function.  A previous transcriptomic analysis performed in our laboratory identified the transcription factor gene egr3 as a novel endocardial AV valve marker.  Here, we investigate egr3 function during AV valve development and valve leaflet formation using zebrafish as a model system.  We generated a promoter-trap line and several mutant alleles, including a full locus deletion, by Crispr/Cas9-mediated genome engineering.  We find that egr3 expression is specific to the presumptive endocardial AV canal, and that loss of Egr3 function leads to the absence of AV valve leaflets, resulting in severe retrograde blood flow.  In addition, we observed that egr3 expression in the AV endocardial cells was lost in non-beating hearts, and that it was expanded in perturbed cardiac flow conditions, suggesting that it is responsive to mechanical forces.  Altogether, this work identifies Egr3 as a critical regulator of AV valve formation, downstream of blood flow, thereby uncovering a new candidate gene for valve-related CHDs.


Agatha RIBEIRO DA SILVA (Bad Nauheim, Germany), Thomas JUAN, Felix GUNAWAN, Didier STAINIER
17:45 - 20:00 #29486 - 096. Cell-autonomous role of mesodermal Fn1 in outflow tract elongation.
096. Cell-autonomous role of mesodermal Fn1 in outflow tract elongation.

Introduction: Defective outflow tract (OFT) elongation causes congenital heart disease (CHD) resulting in newborn lethality. OFT formation involves the addition of second heart field (SHF) cells to the nascent heart tube, and regulation of SHF cell proliferation, migration, and differentiation in the OFT. Fibronectin (Fn1) is an extracellular matrix glycoprotein essential for the development of cardiac structures, including the OFT. Although, the requirement for Fn1 in the OFT elongation is clear, the cellular and molecular mechanisms underlying the role of Fn1 in OFT development remain unknown. Materials and Methods: We ablated Fn1 from the anterior mesoderm, including the SHF, using the Mesp1Cre/+ knock-in strain of mice. To study the role of mesodermal Fn1, we performed immunostainings and western blots using Fn1f/+;Mesp1Cre/+ (control) and Fn1f/-;Mesp1Cre/+ (mutant) embryos isolated at different stages of development. Results: The ablation of mesodermal Fn1 results in a shortened OFT. This defect was due to an altered SHF cell shape and polarity, defects in cell-cell adhesion, proliferation, and premature cell differentiation in the SHF. Interestingly, even when mesodermal Fn1 was depleted, Fn1 expression between the SHF and endoderm was similar in control and mutants. However, the presence of endodermal Fn1 was not sufficient to rescue SHF cell defects in mutant embryos, suggesting that mesodermal Fn1 has a cell-autonomous role in OFT elongation. Discussion: In these studies, we have gained new insights into mechanisms regulating the addition of SHF-derived cells to the primitive heart tube during the formation of the OFT. Our work will lead to a deeper understanding of mechanisms regulating normal cardiac development and alterations that cause CHD.  Future work will be concentrated to understand if SHF cells migration is affected in vivo and why mesodermal Fn1 is required in a cell-autonomous manner.


Cecilia ARRIAGADA (Newark, USA), Sophie ASTROF
17:45 - 20:00 #29515 - 098. Genomic insertion location and orientation affect Bacterial Artificial Chromosome reporter expression patterns.
098. Genomic insertion location and orientation affect Bacterial Artificial Chromosome reporter expression patterns.

Transgenic mouse models are an essential tool to understand mammalian genetics and cell biology. We used a piggyBac-on-BAC strategy to generate transgenic mice carrying a Bacterial Artificial Chromosome (BAC) containing the human ADAMTS19 enhancer and gene, with a tamoxifen-inducible Cre (CreERT2) replacing the first exon. hADAMTS19-CreERT2 mice (F0-F3) were crossed with Rosa26-tdTomato reporter mice and timed-pregnant females dosed with tamoxifen at E13.5. Surprisingly, E16.5 whole embryonic hearts showed three separate, but complementary and founder lineage-specific tdTomato expression patterns that together represent all mouse Adamts19-expressing cell types: atrial and ventricular (AV) cardiomyocytes, outflow tract (OFT) cells, and valve interstitial cells (VICs). Nanopore gDNA sequencing of a selection of founder lineages revealed that the hADAMTS19-CreERT2 BAC had uniquely integrated in a 5’ to 3’ orientation in either N-Cadherin (Cdh2) intron 14 (AV line) or Plexin A2 (Plxna2) intron 9 (OFT line). Immunofluorescence and mouse heart single cell data analyses confirmed that tdTomato expression patterns were consistent with Cdh2 or Plxna2 expression patterns, suggesting that the genomic region surrounding the BAC dominated CreERT2 expression. In contrast, analysis of the VIC subline revealed BAC integration in a reverse (3’ to 5’) orientation in intron 9 of RNA Polymerase II Associated Protein 2 (Rpap2), which is ubiquitously expressed. This reverse orientation allowed CreERT2 expression to be regulated by the hADAMTS19 BAC enhancer. We conclude that the genomic location and insertion orientqtion of BAC transgenes affects their expression pattern.


Johanna COMES (paris), Piet VAN VLIET, Marieke ROZENDAAL, Florian WUNNEMANN, Smith MARTIN, Gregor ANDELFINGER
17:45 - 20:00 #29548 - 100. Mechano-Molecular Control of Heart Formation.
100. Mechano-Molecular Control of Heart Formation.

The heart is the first functional organ to form during vertebrate development. First, a linear heart tube is formed, followed by cardiac looping, chamber formation, and maturation. As for many organs, the heart arises from a simple epithelium with planar polarity properties. It has been established that cardiac chamber remodeling is coordinated through tissue-scale polarization of actomyosin. However, the mechanism governing the looping process is not fully understood. Here, using Zebrafish as a model, we describe the role of actomyosin to generate and distribute the tension forces necessary across the cardiac epithelium during cardiac looping and chamber formation. We found a supracellular actomyosin network required for this process to occur, which is regulated mainly by two kinases (Mylk3 and Rock2a) to achieve the proper distribution of phosphorylated myosin across the ventricular myocardium. Moreover, we also identified that the planar cell polarity pathway (non-canonical Wnt signaling) modulates the appropriate phosphorylation pattern in the cardiac epithelium by controlling the kinase activity in a spatio-temporal manner. Our findings describe a mechano-molecular mechanism necessary for proper looping and chamber formation during cardiogenesis.


Kevin Manuel MÉNDEZ-ACEVEDO (Berlin, Germany), Motahareh MOGHTADAEI, Luca HUMMEL, Anca MARGINEANU, Anne MERKS, Daniela PANÁKOVÁ
17:45 - 20:00 #29726 - 102. High throughput screen of mouse mutant lines identifies new genetic mutations implicated in congenital heart disease.
102. High throughput screen of mouse mutant lines identifies new genetic mutations implicated in congenital heart disease.

Congenital heart disease (CHD) affects 0.8-1.1% of live births each year, making it the most common birth defect globally. Although the significant role of genes in the pathogenesis of CHD has been previously established, the etiology of 60% of CHD remains unexplained. To uncover additional genetic causes of CHD, we analyzed 3D microCT images of lethal and subviable mouse mutant strains generated by the International Mouse Phenotyping Consortium  and looked for evidence of CHD at embryonic days 15.5 and 18.5. We evaluated 430 mutant lines and 2,479 idividual mutants, and scored them for the following CHDs: atrial septal defects (ASD), ventricular septal defects (VSD), patent ductus arteriosis (PDA), truncus arteriosis (TA), double outlet right ventricle (DORV), atrioventricular septal defect (AVSD), transposition of the great arteries (TGA), pulmonary atresia, dextrocardia, situs inversus totalis (SIT), and cleft palate. From our data we identified 28.5% of null mutant mouse strains that exhibited at least a 25% penetrance of CHD. 65.1% of the mutant lines meeting our penetrance threshold were novel, meaning that they have not been associated with CHD previously. Thus, we have identified 79 new genes that give rise to CHD when mutated. We therefore hold that this type of analysis is valuable to identify novel genes implicated in CHD that have not been revealed by other conventional methods. We will extend our findings by performing pathway analyses to uncover novel pathways associated with CHD. We will also use sequence data from CHD patients to uncover novel mutations associated with CHD in patients. Results will eventually be used to help to guide clinical prognoses and treatments.  

 


Monika CHUNG (Newark, USA)
17:45 - 20:00 #30419 - 106. Mechanisms of cardiac de novo sarcomerogenesis.
106. Mechanisms of cardiac de novo sarcomerogenesis.

Defective sarcomeric assembly is implicated in the pathophysiology of several inherited cardiomyopathies. While the mature sarcomere structure has been extensively studied, the molecular mechanisms that drive de novo sarcomere assembly are largely unknown. One of the earliest components in de novo sarcomere assembly is alpha-actinin 2 (ACTN2). ACTN2 localizes to the z body which fuses to form the z disc that serves as a linker for adjacent sarcomeric units.  Here we used human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs), which closely follow this temporal progression of sarcomere formation, to interrogate the early ACTN2 interactome.  We performed RNAseq and mass spectrometry on hPSC-CMs and ACTN2 co-IP components at different times during cardiac differentiation. We find that ACTN2 sequentially associates with structural components such as intermediate filament and junctional proteins, and functional regulators such as calcium channel and metabolic proteins reflecting a coordinated program over time. The early cardiomyocyte coordinates translation and processing of sarcomeric proteins with their spatial organization and incorporation into developing myofibrils. ACTN2 in the early z body associates with chaperones and actin binding proteins, suggesting that the z body acts as a nidus for de novo actin filament assembly. In support of this role WNT and ROCK inhibition result in an increase of ordered sarcomeric filaments. We further identified ribosomal subunits and RNA binding proteins in the ACTN2 interactome, suggesting a role of the z body as a dynamic condensate. We confirmed this dynamic state of the z body using ACTN2 FRAP. We further show that epiblast-specific loss of Ddx3x, an early ACTN2 interactor and RNA binding protein, leads to embryonic lethality and cardiac defects in the mouse. Together these findings highlight the importance of the z body in the assembly process and provide a window to study the mechanisms of sarcomere organization.


Bhavana SHEWALE (New York, USA)
17:45 - 20:00 #30447 - 110. Ploidy reversal as a postnatal developmental program in murine hearts.
110. Ploidy reversal as a postnatal developmental program in murine hearts.

Polyploidization is a normal cellular adaptation for a variety of cell types in mammals, including cardiomyocytes (CMs); however, the development of polyploidization understudied. Recent work suggests that genetics contribute to diverse displays of ploidy in the adult murine heart. Thus, we hypothesized that the developmental progression to differing end-states may similarly be affected. Here, we assessed CM endowment, cell cycle, and ploidy temporally across two diverse inbred mouse strains, A/J and C57Bl/6J. Consistent with previous work, C57Bl/6J hearts displayed rapid cell cycle activation in the first postnatal week. Total CM numbers are relatively unchanged after P7, while final ploidy states are generally constant from P14 on. In contrast, A/J mice displayed depressed cell cycle activation in the first week of life.  Polyploidy in A/Js reaches its peak at P21, whereby only ~3.5% of CMs remained mononuclear and diploid. Interestingly, from 3-6 weeks of age, we observed an expansion of total CM numbers and of the 1x2N subpopulation to ~8%, which could not be explained by proliferation of the residual diploid population. Instead, the expanded diploid population appears to come from a polyploid CM. This finding was confirmed by analysis identifying a population of CMs which had completed cytokinesis by 6 weeks which was not present at 3 weeks. We believe this is the first report of ploidy reversal by a mammalian CM, a phenomenon first observed by the hepatocyte field. Ongoing single nuclear RNA seq is examining the transcriptomic differences between A/J and C57Bl/6J CMs at P21 to identify this unique population competent to undergo ploidy reversal. Preliminary results suggest A/J CM nuclei, regardless of their ploidy state, are more likely to be in the cell cycle in comparison to C57Bl/6J. Understanding the developmental paths to end state CM endowment and ploidy states helps us understand the biology of organ size and can be applied to stimulating regeneration.


Samantha SWIFT (Milwaukee, USA), Mary KOLELL, Alexandra PURDY, Michael FLINN, Caitlin O'MEARA, Christoph RAU, Michaela PATTERSON
17:45 - 20:00 #30467 - 112. Embryonic macrophage loss results in cardiac dysfunction and disrupts adult heart health.
112. Embryonic macrophage loss results in cardiac dysfunction and disrupts adult heart health.

Macrophages are well-characterized as sentinel immune cells that coordinate cellular responses to injury and infection. However, recent developments in cardiac macrophage biology have broadened our understanding of this critical cell type in heart development and function. Still, it is not known how loss of embryonic-derived macrophages affects heart health into adulthood. Here, we are the first group to show in larval and adult zebrafish as well as in adolescent mice that macrophages reside at the sinoatrial node, where they couple to nodal cardiomyocytes via connexin-45. Using a combination of histology, echocardiograms, electrocardiograms (ECGs), and physiological stress tests, we demonstrate that loss of embryonic macrophages significantly disrupts adult heart health and function, leading to arrhythmia and fibrosis. Adult irf8st96/st96 embryonic-macrophage knockout zebrafish have significant disruption in epicardial integrity, detachment, and expansion, known indicators of heart stress or damage. Mutant zebrafish also exhibit increased collagen deposition within the myocardium and along the atrial wall. Longitudinal analysis of echocardiographic measurements reveals significant age- and sex-specific changes in irf8 mutant heart function, including arrhythmia with notable variability in diastasis time as well as consistently prolonged isovolumic relaxation time. ECG traces of macrophage mutants following a 1-hr swim tunnel-based endurance test reveal significantly prolonged P-wave durations and PR intervals compared to wildtype. A subset of mutants also exhibited intermittent P-wave absence and potential flutter, indicating sinus or atrial dysfunction. Overall, these data reveal significant cardiac abnormalities following the specific loss of embryonic-derived macrophages and expand our knowledge of critical macrophage functions governing long-term heart health. 


Shannon PAQUETTE (Providence, USA), Cadence LEE, Alan MORRISON, Jessica PLAVICKI
17:45 - 20:00 #30473 - 114. Investigating the role of DOCK6 and EOGT in cardiac development and congenital heart disease.
114. Investigating the role of DOCK6 and EOGT in cardiac development and congenital heart disease.

Congenital Heart Defects (CHDs) are structural malformations that occur during development and affect 1% of live births worldwide. Correct heart development requires tight spatiotemporal control of both the Rac1 and Notch signalling pathways, and mutations in components or regulators of both pathways can lead to CHDs. Adams-Oliver Syndrome (AOS) is a multisystemic disorder in which 20% of patients also suffer from CHDs. All known causative mutations in AOS have been identified in Rac1 or Notch signalling pathways, including homozygous recessive mutations in DOCK6 and EOGT which regulate Rac1 and Notch signalling respectively. However, it is unknown how mutations in these genes lead to onset of this disease. We aim to understand how mutations in DOCK6 and EOGT lead to dysregulation of Rac1 and Notch signalling during heart development and cause CHDs in patients with AOS, using zebrafish as a model. 

We find that mRNA of both dock6 and eogt is expressed transiently during early zebrafish cardiovascular development. eogt mRNA expression is restricted to a subset of developing vascular structures, while dock6 exhibits slightly broader expression. Interestingly, both genes also have non-coding transcripts which are expressed in the myocardium during early heart morphogenesis. Using CRISPR mediated genome editing we have created zebrafish disease models of AOS with mutations in both dock6 and eogt, in which we are assessing cardiac morphology throughout development. In addition, we are investigating whether non-coding transcripts of dock6 and eogt play a role in heart development, or the onset of CHDs in AOS patients.


Emma ARMITAGE (Sheffield, United Kingdom), Emily NOEL
17:45 - 20:00 #30487 - 116. Sequential action of jnk genes establishes the embryonic left-right axis.
116. Sequential action of jnk genes establishes the embryonic left-right axis.

Establishment of the left-right body axis is critical for the placement, morphogenesis and function of organs, with disruption to laterality often associated with congenital heart malformations – known as heterotaxy. Left-right specification has long been proposed to be dependent on asymmetric fluid flow in the embryonic node, driven by motile (nodal) cilia. Nodal flow ultimately results in asymmetric expression of Nodal in the left lateral plate mesoderm, which directs organ morphogenesis.

Positioning of nodal cilia is governed by the highly conserved Wnt planar cell polarity (PCP) pathway, yet the exact effectors of PCP signalling remain elusive. We have examined the role of the highly conserved JNK (c-Jun N-terminal Kinase) gene family – a proposed downstream component of PCP signalling – in the development and function of the zebrafish node.

We show that together jnk1 and jnk2 function to specify length of nodal cilia, generate asymmetric flow in the node and restrict Nodal to the left lateral plate mesoderm. Crucially, loss of asymmetric Nodal expression does not result in disturbances to heart looping morphogenesis, supporting an emerging model that nodal flow may be dispensable for organ laterality.

This work uncovers multiple roles for the JNK gene family acting at different points during embryonic development. Furthermore, it reinforces observations that other node-independent pathways function in parallel to establish cardiac laterality and that genetic causes underlying heterotaxy are likely to be multi-faceted.


Christopher J DERRICK (Newcastle, UK, United Kingdom), Adrian SANTOS-LEDO, Lorraine ELEY, Isabela Andhika PARAMITA, Deborah J HENDERSON, Bill CHAUDHRY
17:45 - 20:00 #30497 - 118. Establishing the Role of the Conserved TN Domain in Tinman.
118. Establishing the Role of the Conserved TN Domain in Tinman.

Congenital heart disease (CHD) is a major factor in mortality and morbidity in children and adults. To advance our abilities to identify and manage CHD, we need to further understand the key genetic factors involved in causing these maladies. Drosophila melanogaster shares similar cardiac developmental mechanisms with humans and has been an essential model for human heart development. One similarity occurs between Tinman (Tin), a transcription factor in Drosophila vital for cardiac cell differentiation, and its mammalian ortholog NK2 Homeobox 5 (Nkx2.5). These proteins share two conserved regions: the homeobox and the tin (TN) domain. Although the TN domain is completely conserved between these two proteins, there is little known about its significance. By utilizing CRISPR/Cas9 gene editing, I established a line of Drosophila containing an in-frame deletion of the TN domain. Staining Drosophila embryos for a marker of cardial cells revealed mutant embryos generate a significantly more cardial cells than wild type (p<0.01). I also stained embryos for Tin and Svp, a Drosophila protein important for heart development, to determine if the TN domain deletion affected cardial cell specification. Mutant embryos contained more cells expressing tin and more cells expressing svp than wild type embryos. These results support the importance of the TN domain in heart development and indicate the increase in cardial cells occurs before cardial cells differentiate into cells exclusively expressing tin or svp. Characterizing the role of the conserved TN domain in Tin and Nkx2.5 can expand our understanding of cardiac formation and maturation


Cayleen BILECKYJ (San Diego, USA), Richard CRIPPS
17:45 - 20:00 #30520 - 120. Electrical organization of the first heartbeats.
120. Electrical organization of the first heartbeats.

The heart starts beating early in development, and maintains its function throughout dramatic developmental changes in electrophysiology, tissue morphology, and cell fate specification. However, little is known about the heart’s initial transition from silent to active - what are the intermediate dynamics between a quiescent tissue and a periodic and spatially coordinated heartbeat? Capturing the first heartbeats is a severe technical challenge because these sub-second events only occur once in each animal’s life, and could emerge at any point in a window of hours.  To study these initial dynamics, we developed an imaging system to observe calcium dynamics in up to 72 developing zebrafish embryos simultaneously. The first beats, occurring 20-21 hours post fertilization, were initially infrequent and irregular but from onset were coherent propagating waves that swept the heart field. We then developed genetic tools for simultaneous targeted optogenetic perturbation and voltage or calcium imaging, which revealed that the heart cone supports electrical wave propagation and becomes increasingly excitable before spontaneous activity emerges. Using spatially resolved calcium imaging, we mapped the spatial origin of the first heartbeats. This origin is highly variable between individuals, can move suddenly within a short time interval, and is not correlated with genetic markers of future pacemaker cells. Together our data suggest that broad excitability and electrical connectivity impose biophysical constraints that enforce initial spatial organization and timing of the first heartbeats, despite little specialization of cardiac cell function.


Bill JIA (Boston, USA), Sean MEGASON, Adam Ezra COHEN
17:45 - 20:00 #30527 - 122. ECM asymmetry in ‎the early heart tube: dissecting the links between morphology and ECM dynamics in ‎the developing zebrafish ‎heart.
122. ECM asymmetry in ‎the early heart tube: dissecting the links between morphology and ECM dynamics in ‎the developing zebrafish ‎heart.

During development, the heart tube comprises two cellular layers, an outer myocardium and an inner endocardium, between which lies a layer of extracellular matrix (ECM). This cardiac ECM creates specialised extracellular environments important in mediating the chemical and biomechanical signals that shape the heart tube into a three-dimensional (3D) organ. Visualising and linking ECM and tissue morphology is challenging since sample processing may impact matrix hydration and tissue structure. Using morphoHeart, a novel and in-house-developed quantitative image analysis tool that allows the 3D segmentation of the heart layers  -including the ECM- from live embryos, we have identified an early left-right axis of ECM-regionalisation linked to heart looping and orientation of chamber ballooning.

To understand if defects in heart tube lateralisation result in abnormal ECM-regionalisation and hence disrupted morphogenesis, we investigated zebrafish hearts in which spaw (zebrafish homolog of Nodal responsible for left-right laterality) is disrupted. Analysis of cardiac ECM distribution in spaw mutants identified that ECM asymmetry in the early tube is maintained but mispositioned. Irrespective of subsequent looping direction, this early ECM asymmetry translates into a highly regionalised ECM in the inner and outer curvatures of the atrium at looping and ballooning stages. Further linking laterality, ECM and heart morphogenesis, quantification of heart size in spaw mutants demonstrated a role for Nodal in timely chamber growth and chamber-specific ECM remodelling, even in dextrally-looped mutant hearts. Together we propose a model whereby laterality cues orient an early asymmetric cardiac ECM that intrinsically sets an axis for chamber growth and looping morphogenesis to occur.


Juliana SÁNCHEZ-POSADA (Sheffield, United Kingdom), Emily S. NOËL
17:45 - 20:00 #30536 - 124. Developmental origins of Lmna-related dilated cardiomyopathies and impact of FGF10 treatment.
124. Developmental origins of Lmna-related dilated cardiomyopathies and impact of FGF10 treatment.

Dilated cardiomyopathies (DCM) including ischemic- and genetic-related DCM are characterized by cardiomyocyte necrosis and fibrosis associated with impaired cardiac function leading to severe heart failure. LMNA is mutated in 10% of DCM cases in humans and is responsible for rapidly progressing DCM with severe cardiac defects. While the role of LMNA has been extensively studied in adult heart, its role during heart development remained to be determined. We recently identified the fibroblast growth factor 10 (FGF10) as a potential target to promote cardiac regeneration in ischemic DCM.

This study thus aims to uncover the role and the cell-type requirement of Lmna during heart development, maturation and homeostasis and to evaluate FGF10 as a therapeutic target in Lmna-related DCM.

Transgenic mouse lines will be used to perform temporal (development, maturation and adult) and cell-type specific (cardiomyocytes and cardiac fibroblasts) deletion of the Lmna and conditional Fgf10 overexpression.

Expression profiling revealed increased LMNA myocardial expression from embryonic to postnatal stages. Interestingly, our results revealed that LMNA preferentially accumulates in cardiac fibroblasts in fetal and postnatal heart. Cardiomyocyte specific Lmna deletion in fetal, postnatal and adult heart displays rapid cardiac function alteration with dramatic left ventricular dilatation and massive fibrosis, unveiling a critical requirement in developing cardiomyocytes. Interestingly, specific Lmna-deletion in embryonic cardiac fibroblasts leads to embryonic lethality, revealing an unsuspected role for LMNA in cardiac fibroblasts. Finally, we evaluated the impact of FGF10 treatment on Lmna-related DCM and our results suggest that FGF10 preserves cardiac fibrosis infiltration and cardiac remodelling in Lmna-related DCM.

Altogether, this study unveils the key developmental role of LMNA and identifies FGF10 as a therapeutic target for cardiac regeneration in Lmna-related DCM.


Laetitia BOUCHARD (Marseille), Antoine MUCHIR, Francesca ROCHAIS
17:45 - 20:00 #30541 - 126. Functional analysis of the novel BAV candidate gene muc4 in zebrafish cardiac valve leaflet development.
126. Functional analysis of the novel BAV candidate gene muc4 in zebrafish cardiac valve leaflet development.

Bicuspid aortic valve (BAV) is the most common congenital heart defect that affects around 1.5% of population. The majority of patients develop aortic valve stenosis, aortic root dilatation or infective endocarditis. As a consequence, BAV accounts for nearly 50% of all aortic valve replacements. The genetical heterogeneity of BAV makes identification of its genetic causes complicated, thus, to date they remain largely elusive.

Based on unpublished GWAS study of 2236 BAV patients and 11604 controls, we identified a new risk locus for BAV – MUC4. The goal of this study was to functionally characterize muc4 in zebrafish and analyze its involvement in cardiac valve morphogenesis.

We used two alternative approaches for functional analyses of muc4 in zebrafish atrioventricular (AV) valvulogenesis: transient F0 CRISPR/Cas9-mediated gene knockout and antisense oligonucleotide Morpholino- (MO)-mediated gene knockdown. The 1-cell stage embryos were injected either with a mix of Cas9 protein and muc4-gRNAs or with muc4-MOs. At 56-58 hours post fertilization (hpf) the crispant and morphant embryos were fixed, genotyped and imaged using confocal microscopy against Alcam and kdrl:GFP.

Using both approaches, we observed a delay in AV valvulogenesis. In the wild-type at 56 hpf, a few AV endocardial cells had ingressed into the extracellular matrix (ECM) to form a multilayered valve leaflet. However, in muc4 crispants or morphants, the valve remained monolayered or only one cell had ingressed into the ECM. This phenotype had a penetrance of approx. 50%.

Here, we have functionally characterized a new BAV candidate gene, MUC4. Knockout and knockdown of this gene in zebrafish revealed that muc4 may impact cardiac AV valvulogenesis by causing a developmental delay during the multilayering of valve leaflets. Our research constitutes the basis for future studies on the relevance of MUC4 in BAV as one of the candidate genes, contributing to this polygenic pathology.


Dinara SHARIPOVA (Potsdam, Germany), Federica FONTANA, Jan GEHLEN, Anja STUNDL, Radoslaw DEBIEC, Markus KRANE, Salim ABDELILAH-SEYFRIED, Nilesh SAMANI, Jaenette ERDMANN, Teresa TRENKWALDER, Johannes SCHUMACHER
17:45 - 20:00 #30545 - 128. Conditional deletion of Wt1 in Wt1Cre cells reveals a partial lethality of Wt1LoxP/GFP;Wt1Cre mice.
128. Conditional deletion of Wt1 in Wt1Cre cells reveals a partial lethality of Wt1LoxP/GFP;Wt1Cre mice.

The Wilms tumor 1 homolog (Wt1) encodes a zinc finger protein essential for heart development. Over the last ten years, several Wt1 transgenic mouse models have been generated and used by several laboratories to study the behaviour of WT1 expressing cells during heart development and repair. In this study, we generated a new mouse model of the Wt1 gene, the Wt1LoxP/GFP;Wt1Cre mice. In these mice, one copy of the Wt1 allele is flanked by loxP sites, whereas the other copy has a GFP knock-in in exon 1, which disrupts WT1 expression. In addition, the Wt1Cre line used to generate this model is a BAC transgenic line that has been extensively used in the cardiovascular field. The genotyping of postnatal mice of our model revealed a sub-Mendelian distribution of the Wt1LoxP/GFP;Wt1Cre mice. The observed lethality of Wt1LoxP/GFP;Wt1Cre mice probably corresponds to the late embryonic stages since the proportion of this genotype was not reduced when embryos were examined on days E14.5. Interestingly, all mutant mice lacked mature gonads and displayed genital tracts containing both male and female genital structures and ambiguous genitalia. In addition, the histological examination of embryonic Wt1LoxP/GFP;Wt1Cre mice demonstrated severe heart defects, which could be the main cause of lethality of some mutant mice. Our results call for cautious interpretation of data obtained by conditional gene deletion experiments using this Wt1Cre line, as mouse models in which Wt1 has been deleted in the embryonic heart are embryonic lethal.


Rosa PORTELLA-FORTUNY (BARCELONA, Spain), Otilia GLIGA, Marina RAMIRO-PARETA, Claudia MÜLLER-SÁNCHEZ, Alejo TORRES-CANO, Manuel REINA, Francesc X. SORIANO, Ofelia M. MARTÍNEZ-ESTRADA
17:45 - 20:00 #30553 - 130. Whole genome sequencing identifies evolutionarily constrained genes with chromatin regulators and CHD genes as modifiers of conotruncal heart defects in 22q11.2DS.
130. Whole genome sequencing identifies evolutionarily constrained genes with chromatin regulators and CHD genes as modifiers of conotruncal heart defects in 22q11.2DS.

The prevalence of CHD in 22q11.2 deletion syndrome (22q11.2DS) patients is 65% as compared to the general population at ~0.7%, of which most have conotruncal heart defects (CTD).  To understand the basis of variable phenotypic expression, we analyzed whole-genome sequence data of 456 CTD cases and 537 controls with 22q11.2DS retaining the most damaging coding/splicing rare variants (MDRV), as identified by two independent variant calling pipelines with multiple annotation algorithms and examined 19 gene-sets for the association.  We found a significantly higher burden of MDRVs in three interdependent gene-sets comprising evolutionarily constrained genes (OR=3.31; P-value, 4.00E-07), chromatin regulator genes (OR=4.16; P-value, 7.00E-05) and known congenital heart disease genes (OR=6.63; P-value, 5.9E-04) based upon a weighted gene-set based test.  When removing chromatin regulators and considering constrained genes, most are neuronal synapse genes, while the CHD genes are associated with cardiac development.  Further, by focusing on recurrently affected genes, we found the chromatin regulatory genes were overrepresented in cases, but no enrichment was found in controls.  Overall, using both methods, there are a total of 46 affected chromatin genes in 42 CTD cases accounting for 9.21% of the total, with six recurrently affected chromatin genes including EP400, KAT6A, KMT2C, KMT2D, NDS1, and PHF21A, involved in histone peptidyl-lysine modification.  Although ubiquitously expressed, the chromatin genes have enriched expression in cardiac progenitor cells in the pharyngeal apparatus, where TBX1, the transcription factor gene for 22q11.2DS is expressed.  Modifier genes identified might act in the genetic and epigenetic pathways of TBX1 function.


Yingjie ZHAO (Bronx, USA), Lijie SHI, International 22Q11.2Ds Brain And Behavior CONSORTIUM, Bernice MORROW
17:45 - 20:00 #30557 - 132. Mendelian randomization reveals sex-differences in atherogenic cardiovascular disease pain perception.
132. Mendelian randomization reveals sex-differences in atherogenic cardiovascular disease pain perception.

Clinical manifestation of atherogenic cardiovascular disease differs between men and women. Men often present with classical symptoms i.e., chest pain radiating to left arm and jaw. Women, on the other hand, seem to show a wider range of symptoms that are considered less typical, including but not limited to back pain and nausea. The anatomical pathways feeding cardiac pain perception have not been properly characterized with respect to sex-related differences.

 

In our study, we explored the UK Biobank cohort to perform sex-stratified genome-wide association studies for myocardial infarction, coronary artery disease and a variety of pain types to identify instrumental variables for Mendelian randomization. This approach enables determination of sex-differences of the causality between heart disease and its clinical manifestation, thus allowing for large-sample size evidence of the differences in atherogenic cardiovascular disease manifestation between sexes.

 

We observed that a genetically-influenced chest pain was associated with a higher risk of atherogenic cardiovascular disease in men (OR: 1.49 95% CI: 1.01 – 2.19)Surprisingly, for women, this chest pain was not linked with atherogenic cardiovascular disease (OR: 1.06, 95% CI: 0.96 – 1.17), suggesting chest pain by itself is a less reliable predictor of heart disease. Furthermore, we characterized sex-stratified causal relationships of other pain types with myocardial infarction and coronary artery diseases.

 

Our study identified differences in clinical manifestation of myocardial infarction and coronary artery disease between men and women, that form the basis for our research on developmental and anatomical differences in pain perception between the sexes. 


Ruben METHORST (Leiden, The Netherlands), R NOORDAM, M.r.m. JONGBLOED, M.c DERUITER
17:45 - 20:00 #30565 - 134. Fibulin Genes Regulate Outflow Tract Caliber and Extracellular Matrix Integrity.
134. Fibulin Genes Regulate Outflow Tract Caliber and Extracellular Matrix Integrity.

Critical developmental roles for matrix molecules in proliferation and differentiation and in generating tissue structural integrity are evident by the range of clinical disorders induced by mutations in extracellular matrix (ECM) genes. In the cardiovascular system, recent data underscore the dynamic nature of ECM composition during embryogenesis. This emerging focus on the function of ECM in cardiac development and disease has advanced our understanding of the morphogenetic basis of congenital heart defects (CHD). Yet, we have limited appreciation of the cellular and biophysical mechanisms regulated by individual ECM proteins during formation of the cardiac outflow tract (OFT). Our data reveal that Fibulin molecules, a family of glycoproteins that regulate cell behaviors and structural contributions to the ECM, have previously unrecognized requirements in establishing the proper dimensions of the vertebrate OFT. Zebrafish fibulin mutants also exhibit decreased TGF-β signaling in second heart field (SHF) progenitors of the linear heart tube. Consequently, fibulin-deficient SHF progenitors supply fewer endothelial and smooth muscle cells, resulting in a narrower arterial pole. In addition to effects on SHF progenitor differentiation, we find that deposition of Elastin, a crucial ECM component of the OFT, is impaired as evidenced by decreased expression, fiber number and alignment in fibulin mutant embryos. Importantly, tissue stiffness of the OFT auxiliary chamber is impaired by disrupted ECM integrity and these factors govern biomechanical function. Our insights will yield valuable strategies to improve engineered biomaterials for surgical intervention in congenital OFT anomalies and will fuel further identification of novel disease genes in CHD.


Di YAO, Angelika ALEMAN (New York, USA), Caitlin FORD, Micah WOODARD, Carmen DE SENA TOMAS, Kimara TARGOFF
17:45 - 20:00 #30571 - 136. Estimation of tissue-scale dynamics during mouse cardiac morphogenesis based on microscopic observation.
136. Estimation of tissue-scale dynamics during mouse cardiac morphogenesis based on microscopic observation.

Diabetic pregnancy has increased risk of asymmetric septal hypertrophy in the fetus, and we aim to elucidate how the nutritional and metabolic environment of the embryo regulates cardiac morphogenesis. In a previous study, we revealed that cardiomyocyte maturation is impaired in a high glucose environment in vitro, however it remains unclear how ventricular chamber undergoes asymmetric hypertrophy at organ scale.

To understand morphogenetic mechanisms, it is essential to clarify the hierarchical relationship among organ, tissue and cellular activities. Histological analysis revealed disarrayed muscle fibers in the hypertrophic tissue at cellular scale, however little is known about the tissue scale deformation process. Based on histological observation, we hypothesized that the tissue scale morphogenetic process of hypertrophy is isotropic growth caused by disruption of proper orientation in tissue elongation. To test this hypothesis, we developed a new method to estimate the size and direction of local tissue growth during heart morphogenesis. Specifically, 3D whole organ imaging is performed to detect the cells of the same origin in a fixed heart using MADM-based tracking. Then, based on the detected spatial distribution of these cells, thickening of myocardial wall and the direction of tissue stretch at each local tissue are calculated. In this presentation, we provide a proof of concept for this method through the analysis of artificial data. We discuss how to apply this method to reveal the anisotropy of tissue growth and how to dissect the cellular mechanisms that cause tissue scale dynamics.

This new methodology for estimating the tissue scale deformation process during organogenesis will help us understand the multiscale morphogenetic mechanisms of organ development.


Naofumi KAWAHIRA (Los Angeles, USA), Haruko NAKANO, Atsushi NAKANO
17:45 - 20:00 #30576 - 138. Adaptation of the sinus venosus reveals insights into emergent CHDs and evolutionary transitions of the vertebrate heart.
138. Adaptation of the sinus venosus reveals insights into emergent CHDs and evolutionary transitions of the vertebrate heart.

Nr2f transcription factors play an integral role in vertebrate atrial development and have been linked to congenital heart defects (CHDs) in humans. Adult patients with structural CHDs affecting the atrium often present with secondary complications, such as pulmonary hypertension and vascular remodeling. Here, we report a zebrafish nr2f1a mutant allele with embryonic atrial defects that can survive to adulthood. Remarkably, despite essentially lacking an atrium, the adjacent sinus venosus (SV) in these mutants undergoes adaptive remodeling, which entails substantial thickening due to increased cell proliferation. Transcriptomic analysis of the bulbus arteriosus (BA) and SV, transgenic reporters, and genetic lineage tracing showed previously unknown molecular and cellular similarity between the wild-type BA and SV, including the presence of vascular smooth muscle (VSM) and neural crest cells. Furthermore, single cell RNA-seq revealed novel heterogeneity of VSM cell populations in the BA and SV and that VSM and endothelial cells within the adapting SV of the nr2f1a mutants take on transcriptional signatures of the BA. Echocardiography showed retrograde blood flow in adult nr2f1a mutant hearts, suggesting that aberrant blood flow contributes to the adaptive remodeling of the SV. Consistent with this hypothesis, prolonged treatment with a vasodilator mitigated thickening, while increased exercise exacerbated the adaptive thickening. Altogether, our data demonstrate an unexpected latent similarity between the large chamber-like vessels at the arterial and venous poles of the zebrafish heart and adaptive remodeling of the SV in nr2f1a mutants due to hemodynamic forces, which may provide insights into complications associated with CHDs in humans.


Jacob GAFRANEK (Cincinnati, OH, USA), Enrico D'ANIELLO, Nathan SALOMONIS, Joshua WAXMAN
17:45 - 20:00 #30585 - 140. Vascular Outflow Tract Development: A New Source of Smooth Muscle Cells in the Ascending Aorta.
140. Vascular Outflow Tract Development: A New Source of Smooth Muscle Cells in the Ascending Aorta.

The ascending aorta harbors a heterogenic smooth muscle cell (SMC) population. Embryonic lineage tracing studies have shown that extracardiac cells contributing to the vascular outflow tract (OFT), originate in the cardiac neural crest and second heart field (SHF). The cardiac neural crest is important for proper outflow tract septation and formation of the inner SMCs layer. The SHF mainly contributes to the SMC population of the pulmonary trunk and a small part of the ascending aorta. Currently, the origin of the outer medial SMC and adventitial fibroblasts is unknown. Recently we described a distinct Wilms Tumor 1 (WT1) positive cell population in the area of the OFT. We hypothesize that these cells contribute to the aortic SMC and fibroblast population. Using the Cre-LoxP recombination system driven by an epicardial specific promotor (WT1-CreER2) and the ROSAmT/mG reporter line we studied the fate of this cell populationfrom stage E9.5 till E17.5 in mouse embryos.

Immunohistochemistry revealed that this  population of cells shares other SHF markers.. GFP-labeled epicardial cells undergo -mesenchymal transformation and migrate into the arterial vessel wall of the ascending aorta. During this process,these cells lose their epicardial phenotype and contribute to the mural cells of the outer layer of the aorta and pulmonary trunk. Our results demonstrate a new embryonic origin of SMC which specifically contributes to media and adventitia of the ascending aorta but not to the aortic arch or descending aorta. Future studies will focus on their specific role in normal development and during pathological circumstances.


Tamara BORSBOOM (Leiden, The Netherlands), Esther DRONKERS, Lambertus WISSE, Conny MUNSTEREN, Bart LOEYS, Marie-Jose GOUMANS, Monique JONGBLOED, Marco DERUITER
17:45 - 20:00 #30591 - 142. A dedicated approach for transmural visualization and quantification of nerve density: application in myocardial infarction biopsies.
142. A dedicated approach for transmural visualization and quantification of nerve density: application in myocardial infarction biopsies.

Abnormal cardiac innervation plays a role in arrhythmogenicity after myocardial infarction(MI). Nerve quantification is challenging as visualization of  nerves requires high magnifications. We developed a method to analyze nerve density(ND) in large transmural biopsies.

Myocardial biopsies from 4 swine, 3 months after MI,  were stained with Sirius Red(fibrosis) and β3Tubulin(autonomic nerves). Fibrosis cut-offs classified biopsies in: MI core(>59.8%), borderzone(BZ)(14.3-59.8%) or remote zone(RZ)(<14.3%). Using a custom software pipeline, biopsies were graphically divided into 1x1mm grids. Nerve and myocardial tissues were thresholded and after  quality check and artefact removal, divided into 0.1x0.1mm squares. ND/square was calculated and classified into denervation, hypoinnervation, normal innervation and hyperinnervation according to control derived cut-offs(5th, 95th %, n biopsies=38). Squares were located back to their original biopsy position for visualization and quantification of innervation types.

All 83 MI biopsies showed variable innervation patterns. Denervation, hypo- and hyperinnervation were largest in MI core and BZ. Innervation was normal in the RZ: 94.9%(93.8-96.0%), decreased in the BZ: 87.9%(85.1-92.4%, p<0.001), and even lower in the MI core: 72.8%(66.3-75.8%, p<0.001). Denervation and hypoinnervation were highest in core biopsies, 16.7%(13.5-23.5%) and 3.8%(2.9-5.0%) resp., lower in the BZ, 2.6%(1.6-6.5%, p<0.008) and 1.2%(0.8-1.7%, p=0.015) and even lower in the RZ, 1.2%(0.7-1.8%, p<0.001), 0.6%(0.3-1.3, p<0.001). Hyperinnervation was observed mainly in the BZ, 5.3%(3.7-9.4%) and in the core, 5.3%(3.7-9.4%, p>0.999), whilst being low in the RZ 2.6%(1.7-4.3%, p=0.002 and p=0.038 respectively).

This method allows detailed visualization and quantification of ND after cardiac damage and can be broadly applied to high resolution nerve imaging. Variable innervation types within the same MI-biopsies indicate a heterogeneous arrhythmia substrate.


H.s. CHEN (Leiden, The Netherlands), J.c. VAN MUNSTEREN, L.m. VOORTMAN, L.j. WISSE, C.a. CLASHAN, M.c. DERUITER, K. ZEPPENFELD, M.r.m. JONGBLOED
17:45 - 20:00 #30594 - 144. The sex of epicardium-derived cells influences the outgrowth of cardiac sympathetic nerves in vitro.
144. The sex of epicardium-derived cells influences the outgrowth of cardiac sympathetic nerves in vitro.

In the past decades, attention on sex differences in the prevalence and outcomes of a wide range of cardiac diseases has increased. Next to overt sex differences in disease presentation and outcome, also differences in autonomic function between males and females have been exposed. After myocardial infarction, male patients have an increased risk for ventricular arrhythmias and sudden cardiac death. Part of these arrhythmias have been attributed to an increase in cardiac sympathetic nerve fibers occurring after cardiac damage. Although mechanical studies of post-myocardial infarction cardiac sympathetic hyperinnervation have raised growing awareness on the role of the autonomic nervous system in arrhythmogenesis, data on the role of sex herein are still scarce and not conclusive. We found that co-cultures of male superior cervical ganglia with male myocardium and male mesenchymal epicardium-derived cells (EPDCs) results in significant higher neurite directional outgrowth towards myocardium compared to entirely female co-cultures. Moreover, male EPDCs in a female setting enhanced the female neurite outgrowth comparable to an entire male environment. We are currently exploring by RNA sequencing of male and female EPDCs, whether  male and female EPDCs have a differential expression of axon repellent guidance cues. Our results confirm the stimulating effect of EPDCs on neurite outgrowth and also demonstrate that sympathetic nerve outgrowth and density differs between male and female in vitro. Our data underlines the potential relevance of sex differences in post-myocardial infarction cardiac hyperinnervation.


Yang GE, Janine VAN GILS (Leiden, The Netherlands), Ruben METHORST, Conny VAN MUNSTEREN, Lieke VAN ROON, Anke SMITS, Marie-José GOUMANS, Marco DERUITER
17:45 - 20:00 #30603 - 146. Role of reactive oxygen species in maternal hyperglycemia-induced congenital heart defects: a potential therapeutic target.
146. Role of reactive oxygen species in maternal hyperglycemia-induced congenital heart defects: a potential therapeutic target.

Congenital heart defects (CHD) affect 1% of all live births in the US annually. The etiology of CHD is multifactorial, that is, both pathogenic genomic variation and environmental risk factors act as CHD contributors. Among the environmental teratogens, maternal pre-gestational diabetes mellitus (matDM) is associated with up to ~5-fold increase in the risk of having an infant with CHD.  Generation of excess reactive oxygen species (ROS) due to maternal hyperglycemia is routinely observed in embryonic hearts exposed to matDM but the teratogenic effect of ROS on cardiac developmental pathways is not fully understood. The Notch and Nitric oxide (NO) signaling pathways, which are highly expressed in the endocardium, are critical for normal heart development.  Previously, we identified a reduction in NO bioavailability and Notch signaling in mouse embryonic hearts exposed to matDM and reported a gene-environment interaction between Notch1 haploinsufficiency and matDM in the development of CHD. Here, we investigated the effect of modulating intracellular levels of oxidative stress in endocardial and endocardial-derived cell lineages to determine the role of ROS in matDM-associated CHD. Endocardial-derived cells exposed to oxidative stress elicit similar responses as hyperglycemia in reduction of NO bioavailability and Notch signaling. To study the role of oxidative stress in vivo, we generated WT and Notch1+/- embryos overexpressing the antioxidant gene, SOD1, to determine whether mitigating oxidative stress can rescue matDM associated CHD. We have confirmed a reduction of oxidative stress in SOD1-tg embryonic hearts and preliminary data shows lower incidence of CHD in SOD1-tg compared to WT embryos exposed to matDM. The role of oxidative stress in matDM-associated CHD requires further investigation, as mitigation of endocardial oxidative stress may be a promising approach to lower the incidence of CHD in high-risk populations.


Talita CHOUDHURY (Columbus, USA), Sara ADAMCZAK, Emily CAMERON, Madhumita BASU, Vidu GARG
17:45 - 20:00 #30612 - 148. Probing the functional necessity of cardiac enhancers associated to key regulators of heart development.
148. Probing the functional necessity of cardiac enhancers associated to key regulators of heart development.

Mammalian heart development depends on the contributions of two major sources of cardiac progenitor cells, the first and the second heart field (FHF/SHF). While FHF cells establish the early heart tube and are major contributors to the left ventricle (LV), SHF progenitors enter the heart tube at the poles and are essential for outflow tract (OFT) and right ventricle (RV) formation. The cardiac transcription factors (TFs) Gata4 and Hand2 are linked in a key gene regulatory module orchestrating SHF progenitor specification, differentiation, proliferation and migration. Mouse embryonic hearts deficient for these TFs exhibit OFT and RV defects, pointing to essential roles of the cis-regulatory landscapes of these genes in controlling SHF progenitors and mirroring congenital heart defects. Using transgenic reporter assays, previous studies have identified sets of enhancers with overlapping activities in SHF progenitors in each locus. However, the related subregional specificities and functions of these enhancers have not yet been characterized in vivo, leaving their functional necessity and involvement in heart disease unexplored. Here, we have generated mouse lines harboring individual deletions of Hand2 and Gata4 cardiac enhancers. Our results based on qualitative and quantitative transcriptional profiling and cardiac marker analysis have revealed considerable cis-regulatory robustness in both, Gata4 and Hand2 topologically associated domains. Nevertheless, we have identified a genomic cardiac enhancer module essentially contributing to Gata4 gene dosage in a heart compartment-specific manner. Our results highlight the complexity of cardiac TF enhancer landscapes and provide a framework for assessment of heart disease-related loss-of-function effects of genomic heart enhancers in vivo.


Virginia ROLAND (Bern, Switzerland), Matteo ZOIA, Julie GAMART, Marco OSTERWALDER
17:45 - 20:00 #30622 - 152. Transcriptional and epigenetic regulation of the cardiac neural crest from induction through migration.
152. Transcriptional and epigenetic regulation of the cardiac neural crest from induction through migration.

The cardiac neural crest is a transient embryonic cell population that contributes to portions of the heart, including the septum of the cardiac outflow tract. Here, we elucidate regulatory changes that may confer ectomesenchymal ability onto the cardiac neural crest, enabling them to contribute to elements of the heart. To this end, we analyzed transcriptional and epigenetic changes in the cardiac neural crest as a function of time from induction to migratory stages. Analysis of single-cell RNA-seq data reveals groups of genes with differential expression profiles, with those associated with ectomesenchymal potential expressed at premigratory stages and then up-regulated during migration. In contrast, genes associated with specification show a reciprocal profile, peaking at the premigratory stages. We next looked for transcription factors co-expressed in the premigratory cardiac but absent from the cranial neural crest at a similar developmental timepoint, identifying FoxC2, FoxP1, and Twist1 as putative regulators of ectomesenchymal fate. Targeted knockouts in the cardiac neural crest using CRISPR-Cas9 revealed that individual loss of each transcription factor results in severe persistent truncus arteriosus. Finally, to build regulatory linkages, we analyze the chromatin landscape of cardiac neural crest cells using Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq). The results reveal numerous enhancers activated in the cardiac crest population at premigratory stages that are further upregulated during migration, characterized by GO terms associated with heart morphogenesis. Taken together, our results suggest that cardiac crest-specific transcription factors expressed during the specification stage may be critical for mediating their ectomesenchymal differentiation at later stages of heart development.


Shashank GANDHI (Berkeley, USA), D. Ayyappa RAJA, Max EZIN, Marianne BRONNER
17:45 - 20:00 #30626 - 154. Congenital heart defects in the fetus, classifications and embryology: cladistics or phenetics?
154. Congenital heart defects in the fetus, classifications and embryology: cladistics or phenetics?

Congenital heart defects (CHD) are a common cause of fetal death and termination of pregnancy. The aim of this study was to phenotype all fetal cardiac specimens of our  collection, and to create a user-friendly database.

Each specimen was thoroughly examined according to segmental analysis. The main CHD was determined according to the 11 categories and 23 subcategories of the clinical and anatomical classification of CHD. Associated lesions were coded using IPCCC ICD-11 Nomenclature. Codes and photographs for each of the 1236 specimens were linked into the database. Among them, 120 were normal hearts (10%). The three main groups of CHD were ventricular outflow tracts anomalies (32%), functionally univentricular hearts (23%), and anomalies of atrioventricular junctions and valves (14%). The most frequent lesions were valvar anomalies (55%), ventricular septal defects (VSD, 40%), ventricular hypoplasia (35%), interatrial communications (35%). A cladistic grouping can superimpose on the anatomical classification to identify similar characteristics and raise embryological hypotheses. For example, the term “conotruncal” anomaly comprises defects supposed to have the same embryological origin/”common ancestor”. A phenetic approach explores how a segmental characteristics associates with other defects. For example, common arterial trunk is almost always associated with outlet VSD, but also with intact interventricular septum, hypoplastic ventricle, or right aortic arch. Clustering segmental cardiac characteristics without a priori developmental hypotheses may help identifying new mechanisms for CHD. Detailed anatomical phenotyping of fetal hearts should allow us to identify rare associations of malformations, and can help elaborating different approaches of describing and grouping CHD.


Manon HILY (Paris), Damien BONNET, Bettina BESSIERES, Nicolas GARCELON, Hassan FAOUR, Lucile HOUYEL
17:45 - 20:00 #30638 - 156. Induced overexpression of rnx2a in cardiovascular cells triggers ectopic calcification.
156. Induced overexpression of rnx2a in cardiovascular cells triggers ectopic calcification.

Cardiovascular calcification (CVC) is one of the most frequent forms of cardiovascular disease, with increasing prevalence showing high morbidity and mortality rates worldwide.  CVC is characterized by the progressive dysfunction of the cardiac muscle, arteries, and cardiac valves due to ectopic calcification of their extracellular matrix (ECM).  Previously considered a passive accumulation of calcified tissue, it is now recognised that these diseases result from a cell-mediated active process.  In fact, it has been suggested that before calcification cardiovascular cells differentiate to an osteoblastic (bone-cell) fate.  Previous work has shown that overexpression of Runt-related Transcription Factor 2 (Runx2) in mouse is sufficient to induce calcification in the heart and aorta.

Here we use the zebrafish to study the initiation of CVC in vivo using genetic tools and live-imaging at single-cell resolution.  We overexpressed runx2a in cardiovascular cells using cell-specific promoters and confirmed the differentiation of these cells to a pro-osteoblastic lineage. Importantly, these cells were in direct contact with ectopic calcification sites and triggered the recruitment of immune cells.  To determine the role of the immune system in the progression of the CVC phenotype we are now using cell type-specific genetic ablation tools.

Overall, we introduce the zebrafish as a unique model to study the events leading to the initiation and progression of CVC, aiming to identify new therapeutic targets.


Inês CRISTO (Lisbon, Portugal), Didier STAINIER, Anabela BENSIMON-BRITO
17:45 - 20:00 #30641 - 158. Dysmorphic sarcoplasmic lumen, altered calcium transients, and depressed contractile ability seen in a patient-specific iPSC-CM model of Ebstein’s anomaly and left ventricular noncompaction.
158. Dysmorphic sarcoplasmic lumen, altered calcium transients, and depressed contractile ability seen in a patient-specific iPSC-CM model of Ebstein’s anomaly and left ventricular noncompaction.

Patients presenting with congenital heart diseases (CHDs), Ebstein’s anomaly (EA) and left ventricular noncompaction (LVNC), suffer higher morbidity than either CHD alone. The genetic etiology and pathogenesis of combined EA/LVNC remain largely unknown. In a familial case of EA/LVNC associated with the KLHL26 (p.R237C) variant, we discovered an altered electrostatic surface profile of the variant protein, which likely decouples the CUL3-interactome and alters protein turnover. 

To investigate the underlying mechanisms, we differentiated a family trio (EA/LVNC-unaffected daughter and EA/LVNC-affected mother and daughter) of induced pluripotent stem cells into cardiomyocytes (iPSC-CMs), assessed for significant differences in CM structure and function (P<0.05), and synthesized cellular and pathway insights. We characterized morphology via immunofluorescence and electron microscopy and contractile function via biomechanical and automated video methods. We analyzed differential mRNA expression via RNA Sequencing of iPSC-CMs and cardiac tissues, and iPSC-CM protein abundances via label-free quantification.

In comparison to the unaffected, EA/LVNC-affected iPSC-CMs exhibited aberrant morphology and contractile ability, mainly a distended endo/sarcoplasmic reticulum (ER/SR), decreased beat rate, and altered calcium transients. The affected daughter iPSC-CMs further showed decreased apoptosis and increased proliferation. From pathway enrichment analyses, we saw a suppression of myocardial contraction and an activation of ER lumen and lipid metabolism. Via RT-PCR, relative to GAPDH and unaffected iPSC-CMs, we found significantly decreased expression of Sarcoplipin (SLN) and increased Junctophilin-2 (JPH2).

Together, these results from cell and developmental studies suggest that this familial EA/LVNC associated with the KLHL26 variant develops dysregulated ER/SR signaling and altered calcium transients with subsequent lesions in CM contractility and cell cycle progression.


Suma THAREJA (Milwaukee, USA), Melissa ANFINSON, Matthew CAVANAUGH, Min-Su KIM, Peter LAMBERTON, Jackson RADANDT, Ryan BROWN, Huan Ling LIANG, Karl STAMM, Muhammad Zeeshan AFZAL, Jennifer STRANDE, Michele FROMMELT, John W. LOUGH, Robert FITTS, Michael E. MITCHELL, Aoy TOMITA-MITCHELL
17:45 - 20:00 #30644 - 160. Characterization of Hippo:YAP1 and Retinoic Acid signaling pathways during atrial cardiomyocyte acquisition.
160. Characterization of Hippo:YAP1 and Retinoic Acid signaling pathways during atrial cardiomyocyte acquisition.

Abnormalities in early development of Cardiac Progenitor Pool (CPC) located in the Second Heart Field can progress to a myriad of congenital heart defeats that can contribute to neonatal mortality. Thus, understanding the regulatory mechanisms that contribute to cell lineage differentiation and fine tuning in early human development will help to prevent or cure heart defects. Specifically, we focus on Retinoic Acid since it is a key regulator in cardiogenesis where it promotes the posterior specifications of CPCs and their progression toward atrial Cardiomyocyte (CM) lineages. Despite its importance, the specific mechanisms of RA are not well understood. In Embryonic stem cells YAP functions differentially and is controlled by multiple different signaling pathways. We have identified YAP1;TEAD 4 factors as important cell fate determinants of atrial CM in cooperation with RA signaling. It has been shown that RA induces NR2F2 (Nuclear Receptor Subfamily 2 group F member 2) expression to confer atrial identity during CPC differentiation. NR2F2 is an orphan nuclear receptor that binds DNA and it forms heterodimers with the retinoid X receptor to regulate transcription of RA target genes. Our data suggest that RA recruits YAP to the chromatin through NR2F2 and that there is a collaborative effect in CM differentiation with YAP and NR2F2. By combining a range of molecular approaches we aim to establish a Link between NR2F2 and YAP;TEAD in vitro and in vivo. Overall, our ongoing studies suggest that RA signaling induces the activation of non-canonical YAP:TEAD enhancers which are integrated with the cardiac transcription factor network of CPCs essential for the specification of atrial lineages.


Conchi ESTARAS, Corrine LITTLE (Philadelphia, USA), Elizabeth ABRAHAM, Mikel ZUBILLAGA, Clara DE PABLO REVOLTOS
17:45 - 20:00 #30650 - 162. “The role of SHROOM3 in congenital heart disease”.
162. “The role of SHROOM3 in congenital heart disease”.

We implicated a novel CHD candidate, SHROOM3, in cardiac defects within mice and humans. Mechanistic studies have focused on SHROOM3 interaction with ROCK1/2 to impact the cytoskeleton. However, a recent study indicates SHROOM3 interaction with cell adhesion protein N-CADHERIN is important during neural tube development. Given the range and severity of defects attributed to SHROOM3, we hypothesize mechanism beyond ROCK1/2 are important during cardiac development. To test this hypothesis we performed complementary analysis of the transcriptome, global proteome, and phospho-proteome, in Shroom3+/+ vs Shroom3gt/gt embryo heart lysates using RNA-Seq and tandem mass tag labeling, mass spectroscopy (MS). Pathway analysis of both the transcriptome and proteome revealed altered G-Protein coupled receptor signaling. However, GO analyses of the phospho-proteome, indicate the top pathway disrupted in Shroom3gt/gt embryos is cardiac cell-cell adhesions. An in vitro cell dissociation assay confirmed SHROOM3-loss-of-function downregulates cell-cell adhesions.  Next, we generated a SHROOM3 protein interactome using co-immunoprecipitation/MS (co-IP/MS), with both full length GFP-SHROOM3 and deletion constructs. The SHROOM3 protein interactome confirms N-CADHERIN is a top binding component and the deletion constructs co-IP/MS reveal a likely interaction site, with the deletion of a.a. 286-776, disrupting SHROOM3-N-CADHERIN interaction. Finally, we interrogated exome sequencing of 283 patients with left-right patterning and cardiac defects, and identified 12 rare, potentially damaging variants in SHROOM3. Whereas no variants fell within the ROCK1/2 binding region, one third cluster to the proposed N-CADHERIN binding region, indicating these variants may interrupt N-CADHERIN interaction and cell adhesions during cardiac development, resulting in CHD. Taken together these data indicate novel role for SHROOM3 interacting with N-CADHERIN driving cell adhesions during cardiac development.


Matthew DURBIN (INDIANAPOLIS, USA), James ZWIERZYNSKI, Stephanie WARE
17:45 - 20:00 #30657 - 164. In Vivo and In Vitro Cartilage Differentiation from Embryonic Epicardial Progenitor Cells.
164. In Vivo and In Vitro Cartilage Differentiation from Embryonic Epicardial Progenitor Cells.

The presence of cartilage tissue in the embryonic and adult hearts of different vertebrate species is a well-recorded fact. However, while the embryonic neural crest has been historically considered as the main source of cardiac cartilage, recently reported results on the wide connective potential of epicardial lineage cells suggest they could also differentiate into chondrocytes. In this work, we describe the formation of cardiac cartilage clusters from proepicardial cells, both in vivo and in vitro. Our findings report, for the first time, cartilage formation from epicardial progenitor cells, and strongly support the concept of proepicardial cells as multipotent connective progenitors. These results are relevant to our understanding of cardiac cell complexity and the responses of cardiac connective tissues to pathologic stimuli.


Paul PALMQUIST-GOMES, Ernesto MARÍN-SEDEÑO, Adrián RUIZ-VILLALBA, Gustavo Adolfo RICO-LLANOS, José María PÉREZ-POMARES, Juan Antonio GUADIX (Málaga, Spain, Spain)
17:45 - 20:00 #30686 - 166. Genome-wide transcriptomics analysis of genes regulated by GATA4, 5 and 6 during cardiomyogenesis.
166. Genome-wide transcriptomics analysis of genes regulated by GATA4, 5 and 6 during cardiomyogenesis.

The transcription factors GATA4, GATA5 and GATA6 are key regulators of vertebrate heart muscle differentiation (cardiomyogenesis), but specific target genes regulated by these individual cardiogenic GATA factors remain unknown. We have identified genes that are specifically regulated by each of them, as well as those regulated by either of them using genome-wide transcriptomics analysis in Xenopus laevis. The genes regulated by gata4 are particularly interesting because GATA4 is able to induce differentiation of beating cardiomyocytes in Xenopus and in mammalian systems. Among the specifically gata4-regulated transcripts we identified two SoxF family members, sox7 and sox18. Experimental reinstatement of gata4 restores sox7 and sox18 expression, and loss of cardiomyocyte differentiation due to gata4 knockdown is partially restored by reinstating sox7 or sox18 expression, while (as previously reported) knockdown of sox7 or sox18 interferers with heart muscle formation. In order to test for conservation in mammalian cardiomyogenesis, we confirmed in mouse embryonic stem cells (ESCs) undergoing cardiomyogenesis that knockdown of Gata4 leads to reduced Sox7 (and Sox 18) expression and that Gata4 is also uniquely capable of promptly inducing Sox7 expression.  Our genome-wide transcriptomics analysis therefore identifies an important and conserved gene regulatory axis from gata4 to the SoxF paralogs sox7 and sox18 and further to heart muscle cell differentiation. Our identification of genes that are differentially regulated by each of cardiogenic gata factors also provides a platform for future investigations on the gene regulatory network underpinning embryonic cardiomyogenesis.


Boni A AFOUDA (Aberdeen, United Kingdom), Adam T LYNCH, Stefan HOPPLER
17:45 - 20:00 #30690 - 168. Neural crest cell-derived Wnt5a regulates second heart field planar cell polarity during cardiac outflow tract development.
168. Neural crest cell-derived Wnt5a regulates second heart field planar cell polarity during cardiac outflow tract development.

Wnt5a is a known regulator of planar cell polarity (PCP) signals in second heart field (SHF) progenitors. However, the exact identity of the cell source of Wnt5a has not been fully elucidated. We studied murine heart development following neural crest cell (NCC)- and SHF-specific conditional knock out of Wnt5a. Our results demonstrate that SHF-derived Wnt5a regulates PCP within the early SHF cells required for initial OFT elongation. Mef2c-cre mediated loss of SHF-derived Wnt5a leads to reduced SHF incorporation into the early OFT and a spectrum of OFT phenotypes, ranging from common arterial trunk arising from RV (25%) to double-outlet right ventricle (DORV, 25%) to normal OFT development (50%). As NCC migrate into the developing OFT, they also secrete Wnt5a. Wnt1-cre mediated loss of NCC-derived Wnt5a causes downregulation of PCP signaling and migratory arrest of more cranial, late wave SHF progenitors. The resulting foreshortened OFT is unable to align over the ventricles such that the appropriate rotation and development of the pulmonary trunk is perturbed. All NCC-mutants demonstrate DORV (100%), with additional pulmonary outflow defects observed in 62%. Neither conditional mutant demonstrates disrupted NCC migration into OFT or venous pole defects. Overall, these results demonstrate that different subsets of SHF progenitors respond to PCP signals from SHF and NCC depending on their location and timing during embryonic development. Apart from providing new insights into the source of Wnt5a during heart development, our data are also the first to demonstrate a novel role for NCC in regulating PCP in SHF cells.


Omar TOUBAT, Jongkyu CHOI, Prashan DE ZOYSA, Drayton HARVEY (Los Angeles, USA), Henry SUCOV, Jianbo WANG, Ram Kumar SUBRAMANYAN
17:45 - 20:00 #29372 - 170. Ventricular septation in wild-type and Down Syndrome mice.
170. Ventricular septation in wild-type and Down Syndrome mice.

Down syndrome (DS) is caused by trisomy of human chromosome 21 (Hsa21) and is the most common cause of congenital heart defects (CHDs), with approximately half of all DS births being born with a form of CHD. Most of these arise from aberrant heart septation during development, leading to a combination of shunting between the left and right heart chambers at either the ventricular or atrial levels and incorrect valve formation. However, the developmental origins of the CHDs in DS remain poorly understood. To address this, we are using the Dp1Tyb mouse model of DS, which contains an extra copy of mouse chromosomal regions orthologous to Hsa21, and recapitulates human DS CHD phenotypes, with ~60% of Dp1Tyb embryos at embryonic day 14.5 (E14.5) showing defects in cardiac septation. Analysis of morphogenetic changes during ventricular septation in Dp1Tyb E10.5-E13.5 embryos found no difference in growth of the muscular ventricular septum but showed abnormalities in the outflow tract cushions (OFTCs), the transient developmental structures that complete the final stages of ventricular septation and separation of the pulmonary artery from the aorta. Results indicate that Dp1Tyb OFTCs have altered morphology and reduced cell density. Since most of the OFTCs are derived from cardiac neural crest cells, these results imply that the CHDs found in DS may be caused by changes in neural crest differentiation, potentially linking these defects to other DS-associated abnormalities such as craniofacial dysmorphology.


Mint HTUN (London, United Kingdom), Rifdat AOIDI, Eva LANA-ELOLA, Dorota GIBBINS, Jeremy GREEN, Victor TYBULEWICZ
17:45 - 20:00 #30515 - 172. Identifying key genes in the formation of mammalian heart valves.
172. Identifying key genes in the formation of mammalian heart valves.

Heart valve defects are the most common birth defects in new-borns worldwide. Endocardial (EC) cells are a subpopulation of endothelial (ET) cells that have a unique ability to undergo endothelial-to- mesenchymal (EndoMT) transition, a process critical for heart valve formation. To date, only one molecular maker, NFATc1, is known that uniquely labels EC cells during valve development. However, it is not the master regulator of EC cell specification since mice deleted for Nfatc1 forms underdeveloped heart valves. The ability of EC cells to undergo EndoMT makes them distinct from ET cells, despite both sharing a common precursor origin during cardio-genesis. We hypothesize that EC cells become distinct from ET cells via the expression of a sub-set of genes that are regulated differently at genetic and epigenetic levels.

The integrated analysis of bulk RNA-sequencing and whole-genome bisulphite sequencing data alongside single-cell RNA-sequencing datasets from public domain has identified candidate genes involved in EC-fate determination. These candidates have been validated at the gene expression level. Additionally, inducible shRNA knockdowns of candidate genes have been studied in an Nfatc1-mCherry mouse ES cell line using an in vitro hanging drop culture differentiation system. This has provided a functional assay for the temporal knockdown effects of these genes on the EC differentiation and specification. One of the top candidates is Zfpm1 and the effect of its knockdown on the functional ability of EC cells to undergo EndoMT is assessed using trans-well invasion and migration assays. Altogether, this will not only facilitate our understanding of the role of Zfpm1 in heart development but will also provide a basis for stem cell-based therapies for valve-related defects. 


Punkita LOHIYA (London, United Kingdom)
17:45 - 20:00 #30719 - 174. Epigenetic control of cardiac metabolism by the histone demethylase Kdm8 prevents heart failure.
174. Epigenetic control of cardiac metabolism by the histone demethylase Kdm8 prevents heart failure.

Kdm8 demethylates the di-methylated form of lysine 36 of histone H3 (H3K36me2) and prevents spurious gene expression. Kdm8 is enriched in the developing mouse heart, and its constitutive inactivation is embryonically lethal. Moreover, H3K36me distributes abnormally genome wide in the failing heart, in which metabolism shifts towards glycolysis. However, the function of Kdm8 in the heart or the contribution of metabolic imbalance to initiate the events leading to heart failure are unknown. We show that Kdm8 maintains the expression of genes controlling mitochondrial metabolism by repressing Tbx15 to prevent dilated cardiomyopathy leading to lethal heart failure. Kdm8 inactivation in cardiomyocytes by cre-mediated homologous recombination increased global levels of H3K36me2, caused adverse myocardial remodeling beginning at 4 months, and dead due to heart failure by 9 months of age. Genome wide mRNA profiling revealed that Tbx15 target genes controlling NAD+ metabolism were downregulated in Kdm8 mutant hearts before the initiation of adverse myocardial remodelling. Moreover, metabolomics showed decreased NAD+ pathway intermediates, and NAD+ treatment blunted the initiation of cardiac deterioration towards heart failure. Furthermore, Tbx15 overexpression in cardiomyocytes blunted the respiratory increase induced by NAD+. This suggests that dysregulation of a KDM8 – TBX15 axis sits high in the hierarchy of events initiating cardiac deterioration towards heart failure. Indeed, KDM8 was downregulated in human hearts affected by heart failure. Furthermore, higher expression of TBX15 tracked with stronger downregulation of genes encoding mitochondrial proteins. Our findings suggest that epigenetically controlled metabolic gene networks are dysregulated to initiate adverse myocardial remodelling towards heart failure.


Abdalla AHMED, Carmina PEREZ ROMERO, Lijun CHI, Jibran SYED, Yaxu WANG, Quetzalcoatl ESCALANTE COVARRUBIAS, Dorothy LEE, Etri KOCAQI, Lorena AGUILAR ARNAL, Gerard Bryan GONZALES, Kyoung-Han KIM, Paul DELGADO OLGUIN (Toronto, Canada)
17:45 - 20:00 #30322 - 176. A digital framework to analyse, understand, and stereotypically compare early myocardium morphogenesis in the mouse model.
176. A digital framework to analyse, understand, and stereotypically compare early myocardium morphogenesis in the mouse model.

A dynamic Atlas with cellular resolution is an essential resource for understanding at a multiscale level the key mechanics of cardiac morphogenesis and assessing variability between embryos.
Reconstructing a 4D atlas of the mouse heart is a challenge; indeed, although new live-cell imaging techniques allow a crucial understanding of cellular and tissue kinetics, they do not guarantee a whole-organ acquisition at a spatiotemporal resolution necessary for a detailed multidimensional reconstruction.
To overcome this technological limitation, we designed a dedicated computational framework that, starting from a high-resolution stereotyped atlas, incorporates cellular kinetics and dynamic data of the myocardium provided by time-lapse images from a collection of specimens.
The incorporation of live-embryo data into the Atlas is performed in two fundamental steps: a temporal staging and a spatial mapping of cardiac shapes. Our framework implements a staging system that, through a morphometric feature, assigns to each frame of the time-lapse a developmental stage of the Atlas. Given the temporal matching, each cardiac shape is then mapped, by combined applications of image processing and registration techniques, in the respective Atlas shape.
This last step allows to map the individual cells tracked in the time-lapse images and to project dynamic features such as growth rate, anisotropy and strain to which the myocardium is subjected during its morphogenesis.
This computational method is intended as a novel tool to compare different live-embryos in a single time-space convention, which is necessary to investigate key processes in heart development.


Morena RAIOLA (Spain, Spain), Isaac ESTEBAN, Miguel TORRES
17:45 - 20:00 #30521 - 180. QUANTITATIVE CHARACTERISATION OF A CARDIAC PROGENITOR CELL EPITHELIUM.
180. QUANTITATIVE CHARACTERISATION OF A CARDIAC PROGENITOR CELL EPITHELIUM.

During development, cells adopt diverse strategies to sculpt epithelial tissues into characteristic 3D shapes with molecularly and mechanically distinct regions. The vertebrate heart tube is a good example, as it extends by progressive addition of second heart field (SHF) progenitor cells within an epithelial sheet in the dorsal pericardial wall. Initially contributing to the entire cardiac primordium across the dorsal mesocardium, SHF cell addition is restricted to the poles of the heart tube after dorsal meoscardial breakdown. Perturbation of SHF deployment results in a spectrum of congenital heart defects (CHD). T-box transcription factors implicated in CHD regulate the emergence of a boundary segregating progenitor cells to alternate cardiac poles. In addition, the epithelial properties of SHF cells, including cell elongation, clonal anisotropy and epithelial tension, have been identified as regulatory targets during heart tube elongation. However, the relationship between epithelial properties, mechanical tissue stress and progenitor cell patterning in the SHF remains unknown. Indeed, understanding how cellular and tissue forces contribute to growth and form during organogenesis is a major challenge in developmental biology. Here we present quantitative phenomenological analysis of epithelial cellular properties of cells throughout the dorsal pericardial wall, including cell elongation, polarity, anisotropy and cell contacts.  Patterns of epithelial stress and tension in the SHF are integrated in our dataset using force inference image analysis. We document the dynamic relationship between epithelial features and progenitor cell patterning at embryonic days 8.5 and 9.5, before and after dorsal mesocardium breaks down, and in T-box mutant embryos. By integrating findings from cell scale feature patterning, mechanical tissue stress and cell biology our project will provide new mechanistic insights into cardiac morphogenesis and the origins of CHD.


Clara GUIJARRO CALVO (Marseille), Benoît AIGOUY, Paul VILLOUTREIX, Robert G. KELLY
17:45 - 20:00 #30537 - 182. Role of the cAMP-dependent protein kinase A regulatory subunit RIα in cardiac morphogenesis.
182. Role of the cAMP-dependent protein kinase A regulatory subunit RIα in cardiac morphogenesis.

Second heart field (SHF) progenitors allow the rapid elongation of the embryonic heart tube. The precise control of SHF deployment is a prerequisite for correct heart tube elongation and any alteration results in congenital heart defects (CHD). We previously identified the transcriptional repressor Hes1 as a critical regulator of SHF deployment and preliminary results suggest that regionalized SHF expression of the cAMP-dependent protein kinase (PKA) regulatory subunit RIα (Prkar1a) may control Hes1 expression in the SHF. In the adult heart, PKA is known to regulate heart contraction nevertheless its role during heart development still remains unknown. In human, mutations in PRKAR1A gene cause haploinsufficiency which results in a rare Carney Complex syndrome, associated with benign tumors including cardiac myxomas described to be associated with congenital heart defects.

To evaluate the role of R1α during early heart morphogenesis, we developed transgenic mouse lines allowing conditional deletion of Prkar1a in diverse cardiac progenitor cell lineages.

Our results demonstrate a key role for RIα in controlling SHF deployment. Indeed, RIα deletion in SHF progenitors results in impaired SHF proliferation, associated with early embryonic lethality. Upregulated Hes1 expression was detected in RIα mutants and increased phenotype severity observed in RIα/Hes1 compound mutants strongly support RIα and Hes1 genetic interaction. The extracardiac cell population, cardiac neural crest (CNC), plays a critical role in heart morphogenesis and our results suggest key role for RIα in CNC cell deployment. Additionally, heterozygous RIα deletion in CNC leads to a 5 week-old lethality associated with cardiac hyperplasia and CNC lineage tracing reveals the existence of subendocardial CNC progenitors in the adult heart, suggesting a potential role for CNC derivatives in the development of cardiac myxomas.

Altogether our study reveals a critical role for RIα in early cardiac development.


Corentin PORADA (Marseille), Fabien HUBERT, Grégoire VANDECASTEELE, Francesca ROCHAIS
17:45 - 20:00 #30563 - 184. The lipid sensor Pparg and non-canonical Notch pathway contribute to the regional identity of the outflow tract.
184. The lipid sensor Pparg and non-canonical Notch pathway contribute to the regional identity of the outflow tract.

The arterial pole of the heart is a hotspot for life-threatening forms of congenital heart defects (CHDs). This cardiac region results from the elongation of the embryonic outflow tract (OFT) that requires the addition of Second Heart Field (SHF) progenitor cells to provide the myocardium at the base of the ascending aorta and pulmonary trunk. Investigating the transcriptional program of future sub-aortic and sub-pulmonary myocardium we found that the gene encoding the lipid sensor Pparγ is preferentially expressed in the inferior OFT wall. Here we show that TBX1 is required for the regional expression of PPARγ and its downstream target genes in the inferior OFT. Using mouse genetics and ex vivo embryo culture in the presence of PPARγ agonists or antagonists, we demonstrate that Pparg controls cardiac progenitor cell proliferation and addition of the future subpulmonary myocardium to the OFT, both critical determinants for normal arterial pole development. Moreover, we show that the non-canonical DLK1/NOTCH/HES1 pathway negatively regulates Pparγ in future subaortic myocardium. In conclusion, we present evidence that Pparg is a regulatory component of the cross-circuitry controlling regional identity at the arterial pole of the heart, thus providing new insights into gene, and potential gene-environment, interactions involved in CHD.


Magali THÉVENIAU-RUISSY (Marseille), Mayyasa RAMMAH, Rachel STURNY, Francesca ROCHAIS, Robert G KELLY
17:45 - 20:00 #30590 - 186. Uncovering the signalling cues that influence the development and differentiation of the cardiopharyngeal mesoderm.
186. Uncovering the signalling cues that influence the development and differentiation of the cardiopharyngeal mesoderm.

The heart is built by distinct progenitor populations that contribute to specific cardiac chambers, valves and vessels. These progenitors arise from different germ layers in the gastrulating embryo. The mesoderm-derived cardiac progenitors include a population called the cardiopharyngeal mesoderm (CPM), that contributes to second heart field-derived cardiac regions, but also to head skeletal muscles by populating the pharyngeal arches. While the development and differentiation of cardiac cell types has been examined closely, the developmental mechanisms regulating fate commitment of the head muscle progenitors in the CPM are unclear. I have addressed the early development of the CPM, and its differentiation into skeletal muscle. Using mouse embryos and an embryonic stem cell model, our work shows that inhibition of Wnt/beta-catenin and Nodal pathways underlies the fate specification of these progenitors. Using our findings, we directed embryonic stem cells to differentiate into the CPM and subsequently, into cardiomyocytes and skeletal muscles. Our in vitro model also suggests that there may be a distinct myogenic cue for skeletal muscles derived from the CPM, as compared to skeletal muscles derived from somitic mesoderm. By generating derivatives in vitro, specifically via the CPM, this approach can provide a tractable model of syndromes including both congenital heart diseases and muscular dystrophies.


Nitya NANDKISHORE (Marseille), Fabienne LESCROART, Ramkumar SAMBASIVAN
17:45 - 20:00 #30616 - 188. The role of the apelin receptor in cardiac lineage specification in zebrafish.
188. The role of the apelin receptor in cardiac lineage specification in zebrafish.

The existence of an early cardiac progenitor (CP) population has long been supported by fate mapping experiments which identify presumptive CPs at specific embryonic positions as gastrulation begins. These presumptive CPs then migrate to the anterior lateral plate mesoderm and engage a conserved transcriptional network to direct specification, migration, and differentiation of the cardiac lineage. While later stages of cardiac development are well characterized, initial specification remains poorly understood, due to a lack of specific molecular markers for early CPs. To address the outstanding question of CP specification our group is studying the Apelin receptor (Aplnr), a highly conserved GPCR, first shown to regulate cardiac development in zebrafish where aplnra/b mutants do not express nkx2.5, the earliest specific CP marker, following gastrulation and do not develop cardiomyocytes. Fate mapping analysis has shown that migration of presumptive CPs during gastrulation requires Aplnr. Following loss of Aplnr function Nodal signalling fails to activate correctly and this failure disrupts mesendoderm specification. scRNA-seq has identified that with loss of Aplnr function a cardiac-like mesoderm population fails to develop during gastrulation. Our results indicate that Aplnr is required for the initial commitment of mesodermal cells to the cardiac lineage. Current work is focused on characterizing this specification failure to understand both the function of Aplnr and the mechanisms regulating early CP development.


Nathan STUTT (Toronto, Canada), Ian SCOTT
17:45 - 20:00 #30685 - 190. Exploring the interface between the first and second heart fields: implications for cardiac septation.
190. Exploring the interface between the first and second heart fields: implications for cardiac septation.

The embryonic heart elongates by progressive addition of second heart field (SHF) cardiac progenitor cells to the arterial and venous poles of the early heart tube, derived from the first heart field (FHF). The SHF gives rise to the right ventricle, outflow tract and part of the atrial myocardium. Defects in SHF deployment contribute to a spectrum of congenital heart defects (CHD) affecting the cardiac poles. Septation defects account for over 50% of CHD. The primary atrial septum and dorsal mesenchymal protrusion originate in Tbx1 expressing cells in the posterior dorsal pericardial wall where they activate the FHF regulator Tbx5 in response to retinoic acid (RA) signaling. Transient coexpression of Tbx1 and Tbx5 is followed by downregulation of the SHF program, resulting in emergence of a sharp boundary between Tbx1-positive arterial pole progenitors and Tbx5-positive venous pole progenitors. A similar history of expression of both FHF and SHF regulators, followed by downregulation of the SHF program, demarcates boundary formation at the site of ventricular septation. Here we show that transient overlap between the FHF and SHF programs is observed along the dorsal mesocardium during early heart development, followed by downregulation of the FHF program in the midline of the dorsal pericardial wall. Single cell RNA-seq analysis of the SHF lineage identifies venous pole progenitor cells that switch to a FHF program and mouse genetics reveals that SHF genes are downregulated through T-box gene dependent and independent mechanisms. Finally, using a conditional dominant negative RA receptor we show that RA signaling is required at the heart field interface for morphogenesis of the muscular ventricular septum. Our results point to the importance of a transient regulatory module at the heart field interface in boundary formation during early heart morphogenesis and patterning of future septal structures, providing insights into the etiology of CHD and septum evolution.


Charlotte THELLIER (Marseille), Marcel GRUNERT, Marie COUDERC, Clara GUIJARRO-CALVO, Claudio CORTÉS, Nicolas BERTRAND, Rachel STURNY, Christopher DE BONO, Magali THÉVENIAU-RUISSY, Silke RICKERT-SPERLING, Stéphane ZAFFRAN, Robert G. KELLY
17:45 - 20:00 #30329 - 192. Development of an in vitro model of cardiomyocyte cell competition.
192. Development of an in vitro model of cardiomyocyte cell competition.

Heart regeneration after infarction has been an important goal in the last decades. The low regenerative potential of the mammalian heart has set an essential barrier to this achievement, and innovative strategies need to be developed to recover heart function. Cell competition (CC) has been described in multiple tissues, including the mammalian developmental and adult heart. In this context, fitter cardiomyocytes outcompete their neighbors, thus improving the proportion of more functional cells with no deleterious consequences. However, this process has not been thoroughly characterized yet in the mammalian heart, even though understanding it could help unlock cardiomyocytes' regenerative capacity and heal the injured heart.

We have developed a model to closely study cardiomyocyte CC based on Myc mosaic overexpressing hearts (iMOS Myc). By dissociating P1 iMOS Myc;MHC-Cre mouse hearts, we obtain highly reproducible cultures in which the winner population can be tracked over time. Using this approach we have been able to detect the expansion of Myc-overexpressing cardiomyocytes at the expense of the wildtype ones. This indicates that CC is potentially happening in our experimental setting. We are currently characterizing key features of CC, such as winner cell proliferation and loser cell death. Preliminary data suggests increased proliferation in iMOS-Myc cultures compared to controls, although further characterization is warranted.

Additionally, we have set an in vitro system to overexpress factors of our interest in our cardiomyocyte cultures, taking advantage of the specific tropism of Serotype 9 AAVs. So far, we have expressed YAP5SA, a constitutively active form of YAP that elicits LATS1/2 repressive phosphorylation. We found 2,24% proliferating cardiomyocytes at 3 dpi (compared to a 0,16% rate in controls) with clear signs of sarcomere disassembly. We plan to use this model to test for new factors capable of triggering CC.


Jorge PEÑA PEÑA (Madrid, Spain), Cristina VILLA DEL CAMPO, Miguel TORRES SÁNCHEZ
17:45 - 20:00 #30476 - 194. Generation of iPSC-derived human cardiac valve cells.
194. Generation of iPSC-derived human cardiac valve cells.

Congenital heart disease affects nearly 1% of births and includes valvular abnormalities, such as pulmonary valve stenosis, which commonly occurs in the RASopathies. During valvulogenesis, a subpopulation of endocardial cells overlaying the cardiac jelly undergoes endothelial-to-mesenchymal transition (EndMT), producing valvular interstitial cells (VICs). These resident mesenchymal cells of the cardiac valve are responsible for extracellular matrix production and homeostasis. Furthermore, a subpopulation of endocardial cells escapes EndMT and becomes valvular endothelial cells (VECs), which line the outside of cardiac valve leaflets. Critically, a differentiation strategy to generate both cell populations is needed so that human valvular disease can be modeled in vitro. Recently, a feeder-dependent embryoid body differentiation strategy utilizing BMP10 has been shown to generate an endocardial population that is distinct from vascular endothelial cells. Here, we show that iPSCs maintained without feeder cells can be differentiated as a monolayer towards mesoderm with the GSK3 inhibitor CHIR-99021 and subsequently to an endocardial lineage with BMP10 and bFGF. Importantly, these endocardial cells express high levels of endocardial markers, such as NKX2.5, GATA4, GATA5, and NRG1. EndMT can then be induced on this endocardial population. Following six days of EndMT, some of these endocardial cells become VICs and upregulate key mesenchymal genes, such as SOX9, CDH11, and COL1A1. A subpopulation of endocardial cells escapes EndMT to become VECs and maintain high expression of endothelial genes, such as PECAM1, CDH5, and EMCN. These cell populations can be utilized for in vitro modeling of human cardiac valves.


Clifford LIU (New York, USA), Bruce GELB
17:45 - 20:00 #30501 - 196. Human embryonic stem cell derived cardioids as a model of human development.
196. Human embryonic stem cell derived cardioids as a model of human development.

Chamber-like cardiac organoids or « cardioids » constitute a promising in vitro model to study human physio-pathological cardiac development. A WNT-BMP modulation-based protocol for the generation of self-organizing cardioids recently published (Hofbauer & al., Cell 2021) was optimized using several pluripotent stem cell lines. Cardioids featured expected Ca2+ spiking and contractions from differentiation day 14 when chambers were formed. The physiological significance of cardioids was further challenged.

Both the rate and the amplitude of beating cardioids were challenged using several pharmacological agents including the pacemaker channel inhibitor ivabradine, or the positive inotropic agents phenylephrine and epinephrine, a1-adrenergic (Phenylephrine) or b-adrenergic agonist, respectively. Contractility rate of cardioids was not impaired by a pacemaker current inhibitor (Ivabradine) pointing to a myocyte cell-autonomous generation of beating rate. This observation was confirmed by the presence of functional inositol 1,4,5-trisphosphate receptors (IP3R) within cardiomyocytes, revealed by the inhibition of spontaneous beating rate by Xestospongin.

We identified the early cell aggregation step as a key process which determines both the fate and position of cells in the self-generating cardioids. To improve cell aggregation  and reproducibility  of cardioids over time, micro-patterning used within a Sound Induced Morphogenesis (SIM) device (CymatiX MimiX Biotherapeutics), should turn out to be an extremely powerful technology.

Regardless of their still immature states, we report the functional and physiological properties of chamber-like cardioids and thus their relevance as a potent platform to study cardiac development and physiopathology.


Valentin AZEMARD (Marseille), Michel PUCEAT
17:45 - 20:00 #30518 - 198. Deciphering transcription factor regulatory networks during human cardiac development.
198. Deciphering transcription factor regulatory networks during human cardiac development.

Introduction – Transcription factors (TFs) are key regulators that control the temporal extensive variations of gene expression, the hallmarks of sequential stages of human cardiac development. Genetic alterations of these TFs cause human cardiac defects. TFs act within regulatory networks which have been only partially described. Using cardiac differentiation of human induced pluripotent stem cells (hiPSCs), this study aims at unravelling the global TF regulatory network that govern human cardiac development.

Methods & Results – Transcriptomic data were generated daily, from day 0 to day 30, in triplicates, during directed cardiac differentiations of three hiPSC lines reprogrammed from healthy donor somatic cells. The top 3,000 differentially expressed genes (DEGs) along cardiac differentiation were pooled into 12 gene clusters, illustrating successive temporal alterations of gene expression. Functional annotation of these gene clusters and comparison of their expression profiles to publicly available transcriptomic data of murine cardiac development, demonstrated that hiPSC cardiac differentiation accurately recapitulates transcriptomic processes governing cardiac development. Next, a regulatory network that includes all 216 TFs from the 3,000 DEGs was investigated using an expression-based correlation score involving time delay. Overall, 6,817 activation-type and 6,497 inhibition-type relationships were inferred between these 216 TFs. Compared to the literature-based STRING database, our regulatory network was 3.3 fold denser and could also inform about directions, types and time delays of each interaction. Identified sub-networks included well-known interactions, such as between FOXC1, ISL1, MEF2C and HAND2, but also new candidate sub-networks.

Conclusion – This work shows that hiPSC cardiac differentiation is relevant for developmental studies, and provides a valuable database to explore new TF regulation networks during human cardiac development.


Bastien CIMAROSTI (Nantes), Robin CANAC, Aurore GIRARDEAU, Virginie FOREST, Lemarchand PATRICIA, Redon RICHARD, Nathalie GABORIT, Guillaume LAMIRAULT
17:45 - 20:00 #30558 - 200. The Role of YAP1 in the most important event of everyone’s life: Gastrulation.
200. The Role of YAP1 in the most important event of everyone’s life: Gastrulation.

Long-standing studies in embryos have shown that polarized extraembryonic signals generate a NODAL:SMAD2/3 gradient in the epiblast that regulates gastrulation patterning and the establishment of the anterior-posterior (A-P) axis. However, epiblast-like cells (hESCs and mESC) organize in 2D and 3D gastruloid structures in vitro in the absence of extraembryonic tissues. These observations highlight the importance of intrinsic self-organizing mechanisms in hESCs, independent of polarized extraembryonic signals. Indeed, human 3D-gastruloids display features of Carnegie-stage-9 embryos, with a high degree of organization in gene expression along the A-P axis, including a posterior-to-anterior signature of somitogenesis. The A-P pattern correlates with the organization of signaling components along the length of the gastruloid. In elongated gastruloids, BMP signals are predominantly anterior, while the expression of WNT3 and NODAL genes are restricted to the posterior end, consistent with a role of the latter in the mammalian tailbud at the onset of gastrulation. However, how the regionalization of the signaling components is achieved in the 3D-gastruloids is unknown. Our ongoing analysis suggest that the Hippo-effector YAP1 regulates the antero-posterior organization of the 3D-gastruloids by restricting the posterior expression of Nodal signaling components. Our data show that YAP1 represses Nodal activity through the recruitment of the Polycomb-repressor complex to the chromatin of Nodal genes in hESCs. Hence, in the 3D-gastruloids, YAP1 deletion lead to the anterior expansion of Nodal:Smad2/3 activity. As a consequence, the YAP1 KO-derived 3D-structures are longer than controls, suggestive of an increase in Nodal-induced mesoderm derivatives. Our findings highlight a crucial interaction between YAP1 and NODAL signaling essential for the germ-layer formation and morphogenetic events occurring during the establishment of the body plan.  


Abraham ELIZABETH, Stronati ELEONORA, Zubillaga MIKEL, Conchi ESTARAS (Philadelphia, USA)
17:45 - 20:00 #30608 - 202. Functional characterization of human pluripotent stem cell-derived left ventricle-like cardiomyocytes.
202. Functional characterization of human pluripotent stem cell-derived left ventricle-like cardiomyocytes.

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) offer an attractive experimental platform to model cardiovascular diseases and advance potential regenerative therapies. However, hPSC-CM cultures exhibit heterogeneity and immature characteristics.

 

In our laboratory, we have developed a “Left Ventricle” (LV) differentiation protocol which efficiently generates LV-like cardiomyocytes (hPSC-LV-CMs), characterised by higher maturity and improved functionality than those generated using the “Standard” WNT-On/WNT-Off differentiation protocol. The CytoCypher MultiCell High Throughput System was used to prove improved contractility, calcium dynamics and increased dependence of β2-adrenergic receptor signalling as well as a positive force-frequency relationship as indicators of superior maturity at 30 days of differentiation. To address if hPSC-LV-like CMs could be further functionally matured, we grew cells from day 20-30 in the presence of fatty acid containing maturation media. Results show this media was unable to improve the cells’ contractility and overall cells loss the ability to display a positive force-frequency response. However, a trend for a higher contraction amplitude was noted. This media also led to improved calcium dynamics, but the calcium amplitude of these cells was lower, suggesting that fatty acid supplementation per se is insufficient to improve functional maturity of LV-like CMs.

 

Maturity can be measured in multiple ways and not all media supplements improve cellular maturity in the same way. Some might lead to metabolic maturity (fatty acids) and others may help cytoarchitecture maturity (mechanical loading and pacing) or functional maturity, thus raising the need to develop a media to more holistically improve hPSC-CM maturity.


Elisa FERRARO (London, United Kingdom), Wenjun LI, Lorenza Iolanda TSANSIZI, Marie-Victoire COSSON, Jose SANCHEZ ALONSO-MARDONES, Julia GORELIK, Andreia Sofia BERNARDO
17:45 - 20:00 #30614 - 204. Fast generation of left ventricle-like cardiomyocytes with mature properties from human pluripotent stem cells.
204. Fast generation of left ventricle-like cardiomyocytes with mature properties from human pluripotent stem cells.

Decreased left ventricle (LV) function caused by genetic mutations or injury often leads to debilitating and fatal cardiovascular disease. LV-cardiomyocytes (LV-CMs) are, therefore, a desirable therapeutical target. Here we made use of developmental-cues to devise a protocol for generating LV-cardiomyocytes from human pluripotent stem cells (hPSC). Voltage clamping confirmed hPSC-LV-CMs display a ventricular action potential shape with hallmarks of maturity, including increased expression of IK1. Moreover, hPSC-LV-CMs have an elongated shape, exhibit well-defined sarcomere structures with a length typical of adult cardiomyocytes, express mature cytoarchitecture markers such as TCAP and have a better respiratory capacity. Functionally, LV-cardiomyocytes display more mature calcium transients, supported by higher RYR expression. Engineered heart tissues (EHTs) generated with hPSC-LV-CMs consist of longitudinally organised cardiomyocyte bundles whereas those generated using the standard WNT-On/WNT-Off protocol (hPSC-Std-CMs) are mostly populated by clumps of cardiomyocytes. LV-EHTs produce more force, have faster contraction dynamics and a slow beating rate but can be paced, confirming LV-CMs are losing the pacemaker potentials typical of immature cardiomyocytes. Collectively, we show that: 1) it is possible to rapidly obtain LV-CMs with mature properties, even before they are exposed to reported maturation regimes; and 2) the generation of EHTs per se is unable to rescue the lag in maturation observed between hPSC-Std-CMs and hPSC-LV-CMs. This work demonstrates that hPSC-LV-CMs are a suitable model to study LV development and disease and could enable more faithful LV-specific cardiotoxicity screens. Moreover, it opens the possibility of like-for-like cell replacement therapy becoming an accessible therapy to treat heart failure patients.


Bernardo ANDREIA SOFIA (London, United Kingdom), Nicola DARK, Marie-Victoire COSSON, Thomas OWEN, Lorenza I TSANSIZI, Elisa FERRARO, Alice J FRANCIS, Selina TSAI, Anne WESTON, Lucy COLLINSON, Ken T MACLEOD, Elisabeth EHLER, Sian HARDING, Jim C SMITH
17:45 - 20:00 #30632 - 206. Defining the downstream genetic networks regulated by GATA6 during human cardiogenesis using hESC and iPSC models.
206. Defining the downstream genetic networks regulated by GATA6 during human cardiogenesis using hESC and iPSC models.

Haploinsufficiency for GATA6 is associated with various forms of congenital heart disease (CHD) including septal and conotruncal defects. Genetic loss-of-function studies in model organisms confirmed that GATA6 regulates critical aspects of heart morphogenesis but have not been useful to model human haploinsufficiency. The phenotypic diversity of CHD in patients containing GATA6 mutations is likely due to an unknown combination of interacting gene variants that converge to influence the cardiac phenotype. A greater understanding of the GATA6 genetic regulatory network that controls human heart development is thus essential for better understanding the pathophysiology and ultimately developing therapies. In the present study, we used cardiac directed differentiation with human embryonic stem cells (hESCs) and induced pluripotent stem cells (iPSCs) as a platform to study GATA6 function. GATA6-/- hESCs failed to generate cardiomyocytes (CMs) or cardiac progenitor cells (CPCs) during cardiac-directed differentiation. The expression of markers for cardiac mesoderm were markedly reduced in GATA6-/- cells compared to controls, indicating a defect in mesoderm patterning. Profiling by RNA-seq and CUT&RUN at the mesoderm patterning stage identified genes of the BMP and WNT pathways as being regulated by GATA6, including LGR5, a GPCR associated with WNT signaling and a putative key target. In contrast to the homozygous mutants, GATA6+/- hESCs generated CPCs and CMs but did so less efficiently than controls. Analysis of an iPSC line containing a heterozygous mutation in GATA6 (c.1071delG) derived from a patient with CHD had similar defects in CM differentiation efficiency, suggesting that it can be used to study in vitro the human disease phenotype. Together, this study provides evidence for a regulatory function for GATA6 during human pre-cardiac mesoderm patterning and describes a system for examining GATA6 haploinsufficiency in vitro


Joseph BISSON (New York, USA), Ritu KUMAR, Kelly BANKS, Ellen YANG, Zhong-Dong SHI, Kihyun LEE, Danwei HUANGFU, Todd EVANS
17:45 - 20:00 #30688 - 208. Cardioids unravel mechanisms of compartment-specific cardiac defects.
208. Cardioids unravel mechanisms of compartment-specific cardiac defects.

The number one cause of fetal death are defects in heart development. Determining the underlying causes faces many challenges, including the complexity and inaccessibility of the embryonic heart, the unclear impact of drugs and environmental factors during pregnancy, and the lack of in vitro models representing all the compartments of the human heart. Here, we established a cardioid organoid platform recapitulating the development of the major compartments of the human embryonic heart, including the right and left ventricles, the atria, the outflow tract, and the atrioventricular canal. These cardioids have the compartment-specific in vivo-like gene expression profile, morphology, and functionality. We use this platform to unravel the developmental electrochemical signal propagation between interacting heart chambers and dissect how genetic and environmental factors cause specific defects in different regions of the developing human heart.


Clara SCHMIDT (Vienna, Austria), Alison DEYETT, Tobias ILMER, Aranxa TORRES, Simon HAENDELER, Lavinia CECI GINISTRELLI, Lokesh PIMPALE, Sasha MENDJAN
17:45 - 20:00 #30468 - 210. Cell lineage tracing and local gene ablation based on Cre recombinase microinjection.
210. Cell lineage tracing and local gene ablation based on Cre recombinase microinjection.

Microinjection of lipophilic dyes, iontophoresis and infection by modified viruses allow tracing the embryonic cell-lineages prospectively.  One the one hand, dyes are easy to use but fail to label single cells. On the other hand, iontophoresis and viral vectors require specialized equipment, unavailable in most laboratories. Here, we developed a new method for cell lineage tracing based on the microinjection of the TAT-Cre recombinase protein in embryos carrying floxed reporters. This approach allows the prospective monitoring of cell progenitors and their descendants from primitive streak stages in mouse (E6.5) to organogenesis (E8.5). Using a low titer of TAT-Cre allows single-cell labelling, which makes this method suitable for prospective clonal analysis. Moreover, TAT-Cre microinjection in a floxed targeted gene embryo yields local genetic ablation at a defined region and developmental stage. In summary, this new method is accessible and efficient for cell lineage tracing system, enabling fate mapping, clonal analysis and targeted genetic perturbation in vivo.

 


Miquel SENDRA, Juan De Dios HOURCADE, Antonio José SARABIA, Oscar Horacio OCAÑA, Miguel TORRES, Jorge N DOMINGUEZ (Jaén, Spain)
17:45 - 20:00 #30586 - 212. Retinoic-acid dependent cis-regulatory elements and mechanisms of non-coding genetic disease in heart and human cardioids.
212. Retinoic-acid dependent cis-regulatory elements and mechanisms of non-coding genetic disease in heart and human cardioids.

Congenital heart defects (CHDs) are the most common form of birth defects, occurring in nearly 1% of newborns. Retinoic acid (RA) is a signaling molecule synthesized from dietary vitamin A. An excess of RA has dramatic effects on human embryonic development. This can occur in either the offspring of women undergoing therapeutic treatment with the synthetic retinoid isotretinoin (13-cis-retinoic acid), or in the offspring of women with excess dietary Vitamin A supplementation. 13-cis-RA, known as isotretinoin (INN) and used for the treatment of acne, is correlated with a high risk of birth defects. Administration of INN during pregnancy causes multiple types of malformations in infants, including cardiovascular defects. There is also evidence that reduced maternal levels of Vitamin A can also increase the risk of CHD in her offspring. Despite the tremendous progress in deciphering the implication of the retinoic acid signaling in heart development, many gaps regarding the underlying mechanisms of RA-mediated gene regulation remains to be unraveled. Here, we are aiming at generating atlases of retinoic acid cis-regulatory elements in the developing heart of a Vitamin A deficient mouse model as in human cardioids through the use of multiomic approaches. Our pilot results indicate that retinoic acid signaling regulates a highly conserved, disease-associated candidate enhancer regulating sino-atrial development.


Sonia STEFANOVIC, Stephanie IBRAHIM (Marseille)
17:45 - 20:00 #30651 - 214. The role of Folic Acid in congenital heart disease.
214. The role of Folic Acid in congenital heart disease.

Supplementation of Folic Acid (FA) during pregnancy prevents neural tube defects (NTD). Recent studies indicate FA supplementation may similarly prevent heart defects, though the relationship remains unclear. Proposed etiologies for FA prevention of birth defects include a role in methylation, cell proliferation/differentiation and G-Protein signaling, with disruption affecting susceptible cell populations, including neural crest cells. However, the mechanism remains undetermined. To determine the mechanistic role of FA supplementation during cardiac development, we utilized an established model of high and low FA supplementation (2ppm versus 10ppm) in pregnant wild type mice, and in the resulting embryo hearts at day E12.5, we performed complementary analysis of the cardiac transcriptome, global proteome, and phospho-proteome utilizing RNA-Seq and tandem mass tag mass spectroscopy (TMT/MS). Ingenuity pathway analysis of differentially expressed genes and proteins indicates altered protein translation, with eiF2 signaling topping the list in the transcriptome and global proteome. Within the phospho-proteome, pathway analysis revealed alteration in DNA methylation, cell cycle control, tight junction signaling and RHOA signaling. The protein SHROOM3 is similarly associated with DNA methylation, cell proliferation, tight junctions and RHOA signaling and is expressed int the neural crest. SHROOM3 loss-of-function leads to NTDs that can be reversed with high FA supplementation. We recently demonstrate SHROOM3-loss-of-function leads to cardiac defects. We utilized short term FA supplementation in SHROOM3-loss-of-function mice, and preliminary analysis of 43 embryos reveals reduced penetrance of membranous VSDs from 36% to 11% with high folic acid supplementation. Overall, these findings suggest a mechanistic role for FA during cardiac development that includes DNA methylation, cell proliferation, tight junction and RHOA signaling within susceptible cell populations.


Matthew DURBIN (INDIANAPOLIS, USA), Korre FAIRMAN, James ZWIERZYNSKI, Laura HANELINE, Stephanie WARE
17:45 - 20:00 #29431 - 216. Single cell transcriptomic analysis of murine binucleated and mononucleated cardiomyocytes reveals heterogeneity within ventricular heart muscle.
216. Single cell transcriptomic analysis of murine binucleated and mononucleated cardiomyocytes reveals heterogeneity within ventricular heart muscle.

Mammalian heart regeneration has been the focus of study in the past decades, aiming to promote recovery after ischemic injury. However, mammalian cardiomyocytes have a notoriously slow turnover, incapable of compensating for the loss of contractile muscle.

 

In regenerative models (zebrafish, neonatal mammalian heart), new cardiomyocytes arise form preexisting ones; suggesting the presence of a cardiomyocyte pool able to reenter cell cycle. One hallmark of naïve, proliferative cardiomyocytes is their ploidy levels. Therefore, it has been proposed that mononucleated cardiomyocytes in mice and diploid cardiomyocytes in zebrafish and human can pose a potential population with the ability to replace lost myocardium.

 

However, defining a trancriptomic signature of these myocytes in order to understand and promote a proliferative phenotype has remained elusive due, in part, to the nature and sensitivity of the cardiac muscle cell.

 

Here we describe a method that has enabled us to perform single-cell RNA Sequencing in binucleated and mononucleated cardiomyocytes from murine ventricles. This method allows for a very high-quality RNA purification and depth of sequencing, enabling us to define several cardiomyocyte clusters with distinct transcriptomic profiles, both among binucleated and mononucleated populations, suggesting a higher heterogeneity among cardiomyocytes than what was previously described.

 


Cristina VILLA DEL CAMPO, Cristina VILLA DEL CAMPO (Madrid, Spain), Rocío SIERRA MUÑOZ, Sergio CALLEJAS ALONSO, Ana DOPAZO GONZÁLEZ, Miguel TORRES
17:45 - 20:00 #29462 - 218. Single cell characterization of valve development.
218. Single cell characterization of valve development.

Valvular heart disease affects 1-2% of the population, often requiring surgery. However, current replacement valves do not respond normally to biological signals and cannot undergo growth or proper remodeling. An alternative is the use of valves generated via differentiation of human pluripotent stem cells into valve cells, recapitulating in-vivo development. Valve leaflets are composed of 3 layers of extracellular matrix containing valve interstitial cells (VICs), ensheathed by valve endothelial cells (VECs). Valve development involves the formation of endocardial cushions populated by cells undergoing endothelial-to-mesenchymal transition (EndoMT), followed by elongation and layer stratification. The transcriptional programs driving these changes remain insufficiently defined, and little is known regarding VIC subtypes. Moreover, current differentiation protocols generate heterogeneous rather than specific valve cell populations. Our study aims to identify novel factors driving EndoMT and mechanisms driving specification of VIC subpopulations.

We performed scRNA-seq on E8.0-P1 whole mouse hearts and computationally generated cell type-specific clusters. Endocardial, VEC and VIC markers were identified. Transcriptomic changes corresponding to VICs were noted by E9.0, and two distinct VIC clusters were found at E10.25. Increased EndoMT marker expression was noted at E11.5. Early endothelial and VIC cells were analyzed using URD (Farrell et al 2018), which reconstructs differentiation trajectories in the form of a branching tree. Cells at branch points were subject to gene regulatory network analysis using SCENIC (Aibar et al 2017), and top hits included key transcription factors for valve development. CellPhoneDB (Efremova et al 2020) was then used to identify important cell-cell signalling interactions. The knowledge gained is expected to assist in optimization of differentiation protocols for VICs, and ultimately generation of “biological” stem cell-derived valves.


Lara FEULNER (Montréal, Québec, Canada), Piet VAN VLIET, Hicham AFFIA, Severine LECLERC, Florian WÜNNEMANN, Guy WOLF, Gregor ANDELFINGER
17:45 - 20:00 #30513 - 222. Single-cell RNA sequencing of the human fetal epicardium reveals novel markers and regulators of epithelial-to-mesenchymal transition.
222. Single-cell RNA sequencing of the human fetal epicardium reveals novel markers and regulators of epithelial-to-mesenchymal transition.

The heart is covered by the epicardium, consisting of epithelial cells and a mesenchymal layer. The epicardium has been shown to be essential during cardiac development by contributing cells through epithelial-to-mesenchymal transition (EMT) and the secretion of paracrine factors. In the adult, the epicardium conveys a cardioprotective response after myocardial infarction, albeit suboptimal compared to the epicardial contribution to cardiac development. Direct analysis of the human fetal epicardium is vital as it provides new insights into the cellular and biochemical interactions within the developing heart, which can potentially contribute to enhancing the post-injury response. Epicardial layers were isolated from human fetal hearts (14-15 weeks gestation) and digested into single cells. Single-cell RNA sequencing was used to determine the cellular composition of human fetal epicardium, and data were further explored to identify regulators of epicardial EMT. Analysis of 2073 cells reveals an enrichment of epicardial derived populations and displays a clear clustering of the epicardial epithelial and mesenchymal populations. Importantly, we found that in humans ‘classical’ markers, such as Wilms’ Tumor 1, Transcription factor 21 and T-box transcription factor 18, are not specific enough to reliably classify the epicardium. Our analysis has provided markers that do allow for robust identification of the epicardium validated by immunohistochemistry. To establish the regulation of epicardial activation we focus on the process of EMT within our dataset. Here we present a novel pathway involved in epicardial EMT and several promising candidates that influence key developmental processes.

This work was funded by a Senior Researcher Dekker grant by the Dutch Heart Foundation (2017T059)


Tom STREEF (Leiden, The Netherlands), Tessa VAN HERWAARDEN, Esmee GROENEVELD, Marie-Jose GOUMANS, Anke SMITS
17:45 - 20:00 #30533 - 224. Single-cell analysis implicates early dysregulation of Nanog in the production of syndromic heart defects.
224. Single-cell analysis implicates early dysregulation of Nanog in the production of syndromic heart defects.

Approximately 30% of individuals with Cornelia de Lange Syndrome (CdLS) exhibit congenital heart defects (CHDs). The most common form of CdLS is caused by haploinsufficiency for NIPBL, which encodes a cohesin-associated protein. In Nipbl+/- mice, NIPBL-haploinsufficiency causes hundreds of small gene expression changes in every cell and CHDs similar to those in CdLS. Using the Nipbl+/- mouse as a model system for discovering new causal factors for CHDs, we documented abnormal heart development by cardiac crescent (CC) stage, suggesting that CHDs originate as early as gastrulation. To investigate this, we performed single-cell RNA sequencing on both CC- and gastrulation-stage wildtype and Nipbl+/- mouse embryos. Strikingly, we found that Nipbl+/- embryos dramatically overexpressed NanogNanog encodes a transcriptional repressor required for pluripotency in pre-implantation embryos and is normally transiently re-expressed during gastrulation. In Nipbl+/- mice, however, Nanog fails to shut off appropriately post-gastrulation, so that by CC-stage its levels are many-fold above normal. Interestingly, numerous gene expression changes detected in Nipbl+/-mice involve post-implantation-stage targets of Nanog. These include genes associated with pluripotency (Pou5f1/Oct4), left-right patterning (Tdgf1Lefty2Nodal), anterior-posterior patterning (Hox genes), and primitive erythropoiesis (Tal1Lmo2Hbb-bh1). Accompanying these changes are changes in the allocation of cells to clusters representing Mesp1-expressing cardiac progenitors, the first and second heart fields, and neural crest. These results suggest that a failure to downregulate Nanog expression after gastrulation, and the transcriptional dysregulation that ensues, lead to misallocation and/or dysfunction of cardiogenic cell populations. Funded by NIH-NHBLI.


Stephenson CHEA (Irvine, USA), Arianna G FAVELA, Arthur D LANDER, Anne L CALOF
17:45 - 20:00 #30611 - 228. Refinement of the four-dimensional human heart atlas during the first trimester of gestation with rare and diverse cell states.
228. Refinement of the four-dimensional human heart atlas during the first trimester of gestation with rare and diverse cell states.

Congenital heart defects are frequent and collectively represent a significant burden for human health. Understanding the developmental origins of such anomalies is key to improving diagnoses, prognoses and therapies. The Human Developmental Cell Atlas (HuDeCA) consortium in France gathers as much data and metadata as possible about normal human embryonic and early fetal tissues donated to research, complete with strict quality control procedures and use of standardized annotations, as a complement to the international Human Cell Atlas and a baseline for the scientific community worldwide. Here, we contribute new data about the identities and trajectories of cells in human male and female hearts from 8 to 12 post-conceptional weeks through judicious comparisons of individual hearts assessed through single-nucleus and spatial transcriptomics and advanced imaging approaches. We integrate and validate this information both across our datasets and with existing atlases, using multiple technical platforms to provide four-dimensional analyses at varying levels of resolution. Delineating the gene expression networks deployed within individual cells at specific positions has revealed an unsuspected diversity of cellular states. This data enables new hypotheses about the paracrine influences exerted by and on the many lineages necessary for the complex functions of the first and longest-lived vital organ.

* These authors contributed equally to this work and are joint first authors

** These authors share joint last authorship; correspondence may be addressed to both (stephane.zaffran@inserm.fr, heather.etchevers@inserm.fr).


Samina KAUSAR *, Marine HERBANE *, Elise MARECHAL, Fabienne LESCROART, Nicolas LENFANT, Céline CHEVALIER, Mathias MORENO, Yorick GITTON, Séverine MAZAUD-GUITTOT, Paolo GIACOBINI, Alain CHÉDOTAL, Anaïs BAUDOT, Stéphane ZAFFRAN ** (Marseille), Heather C. ETCHEVERS **
17:45 - 20:00 #30660 - 232. Single-cell transcriptomic profiling reveals maturation signatures in cardiomyocytes derived from LMNB2-inactivated human induced pluripotent stem cells.
232. Single-cell transcriptomic profiling reveals maturation signatures in cardiomyocytes derived from LMNB2-inactivated human induced pluripotent stem cells.

Mammalian cardiomyocyte maturation entails phenotypic and functional optimization during the

late fetal and postnatal phases of heart development, processes that are driven and coordinated by

complex gene regulatory networks. Cardiomyocytes derived from human induced pluripotent stem

cells (iPSCs) are heterogeneous and immature, barely resembling their adult in vivo counterparts.

To characterize relevant developmental programs and maturation states during human iPSC-cardiomyocyte

differentiation, we performed single-cell transcriptomic sequencing. This revealed

six cardiomyocyte subpopulations, with heterogeneity defined by cell cycle and maturation states.

Among those subpopulations, two of them demonstrated a non-proliferative and mature

transcriptional profile. To further investigate the proliferation-maturation transition in

cardiomyocytes, we induced LMNB2 loss-of-function, which represses cell cycle progression in in

vivo cardiomyocytes. This resulted in increased maturation, characterized by transcriptional

profiles related to myofibril structure and energy metabolism, in LMNB2-inactivated

cardiomyocytes. Unique maturation signatures and two maturational trajectories were identified

for control and LMNB2-inactivated cardiomyocytes. By comparing these datasets with single-cell

transcriptomes of human fetal hearts, we were able to define spatiotemporal maturation states in

human iPSC-cardiomyocytes. Our results provide an integrated approach for comparing in vitro differentiated

cardiomyocytes with their in vivo counterparts and suggest a strategy to promote

cardiomyocyte maturation.


Lu HAN (Milwaukee, USA)
17:45 - 20:00 #30649 - 234. SLU7 regulates cardiac developmental programs and contributes to the pathogenesis of hypoplastic left heart syndrome.
234. SLU7 regulates cardiac developmental programs and contributes to the pathogenesis of hypoplastic left heart syndrome.

Hypoplastic left heart syndrome (HLHS) is a severe form of congenital heart disease characterized by underdevelopment of the left ventricle. There are conflicting theories regarding the pathogenesis of this disease. We sought to further define the genetic landscape of HLHS through exome sequencing and mechanistic interrogation using a novel, functional genomics approach (fly, mouse, human). We previously reported whole exome sequencing of a cohort of patients with HLHS identifying the mutational landscape. Now, we extend our analysis, using a functional genomic Drosophila screen to analyze our identified variants more completely for developmentally important genes. We identified new loci that were previously filtered out by in silico techniques, and disregarded. In Drosophila, slu7 knockdown with human SLU7 rescue experiments demonstrate a critical role for SLU7 in heart tube formation. In mice, SLU7 IHC demonstrated cardiac-enriched expression in the developing heart. We created Slu7 gene-targeted mice, that were embryonic lethal, demonstrating an essential role for Slu7 in mammalian development. SLU7 knockdown resulted in defects in transcriptional and alternative splicing of developmental programs, many of which are essential for cardiac embryogenesis. One of the aberrantly spliced products is the chromatin-modifying gene KMT2D, one of the most common genes mutated in HLHS. Furthermore, our studies demonstrate SLU7 as a critical factor in RNA processing and cardiac development in multiple species. These pathways include novel, as well as known (KMT2D), genes, building a knowledge base to enable the development of rational therapeutic and counseling strategies for this complex set of patients.


Yujia YANG (New Haven, USA), Markus KRANE, Neil BOWLES, Hannah LESTER, Thomas MEITINGER, Gabriel BACCAM, Karl-Ludwig LAUGWITZ, Alessandra MORETTI, Zion HAN, Peter GRUBER
17:45 - 20:00 #29497 - 236. Epicardial Meis function is required for cardiac lymphangiogenesis.
236. Epicardial Meis function is required for cardiac lymphangiogenesis.

A single layer of cells could be perceived as insignificant in comparison to a whole organ. However, extensive evidence has shown that the epicardium, the outermost layer of the heart, is essential for cardiac development and regeneration. Epicardium-derived cells (EPDCs) and epicardial signalling are crucial for coronary vasculature development and myocardial growth.

MEIS transcription factors (TFs) are important regulators of cardiac development. Single Meis1 or Meis2-null mice die at midgestation with severe cardiac alterations (Stankunas et al. 2008; Machon et al., 2015). Here, we show that MEIS TFs are expressed and play a function in the epicardium during cardiac development. Meis1 and Meis2 epicardial-specific conditional deletion show prenatal agenesis of the lymphatic vasculature. This reveals a previously unknown non-autonomous function of the epicardium in promoting cardiac lymphangiogenesis. We characterize a population of subepicardial, fibroblast-like lymphatic-associated EPDCs (LEPDCs) that completely enseaths lymphatic vessels as they grow towards the apex of the ventricles. LEPDC and Lymphatic Endothelial Cell (LECs) association is disrupted in Meis mutants, which suggests that LEPDC-LEC crosstalk is important for cardiac lymphangiogenesis. Transcriptomic analysis of Meis-mutant epicardium/subepicardium showed a downregulation of Vegfc and Vegfd lymphoangiocrine signals. Epicardial-specific Vegfc deletion provokes less developed and immature coronary lymphatic vessels, whereas analysis of the lymphatic vasculature of Vegfd knockout hearts shows that VEGFD is important for the development of ventral coronary lymphatics. These results show Meis-regulated direct cellular interactions and paracrine signalling from the epicardium/EPDCs is essential for cardiac lymphatic development.


María Ester DE LA CRUZ CRESPILLO (Madrid, Spain), Vanessa CADENAS, Xiaolei LIU, Miguel TORRES
17:45 - 20:00 #30529 - 238. Epicardial-derived cells play critical role in atrioventricular valve developmental dynamics.
238. Epicardial-derived cells play critical role in atrioventricular valve developmental dynamics.

Myxomatous valve degeneration (MVD) is a common valve disease affecting approximately 2.5% of individuals in the western world. While a lot is known about the clinical manifestations of MVD, the cellular mechanisms that cause MVD largely remain to be elucidated. Using the Wilms Tumor 1 epicardial cre mouse, it has been demonstrated that during development, cells from the epicardium undergo an epicardial-to-mesenchymal transformation and invade the atrioventricular (AV) junction. These epicardial-derived cells (EPDCs) preferentially populate the parietal valve leaflets which are derived from the lateral AV cushions during embryonic valvulogenesis. The mechanisms governing the directional migration of AV-EPDCs into these parietal leaflets are slowly emerging, yet their functional role in regulation of valve development and maturation remains unknown. In order to gain insight into the role of EPDCs in valve development, we conditionally knocked out SOX9, a transcription factor implicated in EMT, valve development, and cell migration, utilizing the Wt1 epicardial cre recombinase. This leads to a significant reduction in the number of EPDCs that populate the parietal leaflets.  Postnatal Wt1cre;SOX9fl/fl specimens have a valvular phenotype reminiscent of myxomatous valve degeneration (MVD) with abnormalities in extracellular matrix (ECM) organization. Transcriptomic analysis of these leaflets display aberrant regulation of genes associated with cell adhesion, regulation of ECM disassembly, and extracellular structure organization. Together, these data indicate an important role for EPDCs in valve development and homeostatic remodeling of the maturing valve and points to the possible involvement of the epicardium in MVD pathogenesis.


Andrew HARVEY (Charleston, SC, USA), Renélyn WOLTERS, Ray DEEPE, Jenna DRUMMOND, Andy WESSELS
17:45 - 20:00 #30550 - 240. The RIG-I Signaling Pathway Contributes to the Pathogenesis of Aortic Valve Stenosis in the Mouse Model of Wire-Injury.
240. The RIG-I Signaling Pathway Contributes to the Pathogenesis of Aortic Valve Stenosis in the Mouse Model of Wire-Injury.

Aortic valve stenosis is characterized by an active calcification process driven by high mechanical stress-induced inflammation in the aortic valve leaflets. Interestingly, Singleton-Merten-Syndrome (SMS), a rare monogenetic disease, which can be caused by gain-of-function mutations in the retinoic acid-inducible gene I (RIG-I), is also characterized by diverse calcification abnormalities including calcification of aortic valve and proximal aorta. Therefore, we aim to investigate the role of the RNA receptor RIG-I in the development and progression of aortic valve stenosis. By using the mouse model of wire-injury, aortic valve stenosis was induced in wildtype (WT), RIG-I and MAVS (mitochondrial antiviral signaling protein) knockout (KO) mice. The development of the disease was monitored by ultrasound, while tissue samples were collected 6 weeks after the induction of aortic valve stenosis and analyzed using immunohistochemistry and fluorescence microscopy. The results showed a significant reduction of aortic peak blood velocity, an indicator of aortic stenosis, in mice deficient for RIG-I as well as for MAVS, the central adaptor molecule of RIG-I. Immunofluorescence staining revealed less CD45 positive immune cells infiltrating the aortic valve in MAVS KO mice compared to WT mice. In addition, MAVS deficiency led to a reduced activation of valvular cells in response to injury, indicated by the decreased expression of the mesenchymal cell marker aSMA in MAVS KO mice. The reduced severity of the aortic valve stenosis in RIG-I and MAVS KO mice suggests a possible role of RIG-I like receptor pathway in the pathophysiology of this disease.


Paraskevi VASILEIADOU (Bonn, Germany), Madeleine GRÄF, Marta STEI, Sandra ADLER, Sebastian ZIMMER, Eva BARTOK, Marcel RENN, Gunther HARTMANN
17:45 - 20:00 #30589 - 242. Wall shear stress activation of EGR1 and KLF2 transcription in valvular cells is mediated by the ERK1/2 mitogen-activated protein kinase pathway.
242. Wall shear stress activation of EGR1 and KLF2 transcription in valvular cells is mediated by the ERK1/2 mitogen-activated protein kinase pathway.

Valvular endothelial cells (VECs) covering the aortic valve leaflets are exposed to different stresses, in particular wall shear stress (WSS). Perturbation in the mechanical environment of the valve can lead to valve leaflet remodeling which ultimately can result in calcification or degeneration of the valve. EGR1 and KLF2 are known as flow responsive transcription factors. However, the pathway activating these factors in response to mechanical signals is not well understood. Our study aims to decipher how EGR1 and KLF2 are activated in response to the WSS. We used a unique fluid activation device that applies physiologically relevant pulsatile WSS to identify the signal required to activate EGR1 and KLF2. We performed gene expression and/or western blot analyses to assess the downstream effect of EGR1 and KLF2. VECs exposed to the WSS were treated with U0126 (ERK1/2 inhibitor). Interestingly, VECs treated with U0126 show a downregulation of EGR1 and KLF2 transcription factors suggesting that the MAPK pathway and more precisely ERK1/2 is required for their activation.  We also used adenovirus or siRNA to overexpress EGR1 and KLF2 in valvular cells. We found that addition of EGR1 adenovirus results in the transcriptional activation of TGFB1 and eNOS while KLF2 is downregulated under the same condition. However, valvular cells treated with EGR1 siRNA over-expressed KLF2, suggesting that both genes are reversibly regulated. Interestingly, we found that EGR1 is a direct activator of the eNOS transcription as confirmed by the increase of luciferase activity when EGR1 is added to the experiment. Our ex vivo model using explanted valve confirm these data. In conclusion our results support that EGR1 and KLF2 are upregulated in VECs exposed to WSS and that this activation is dependent on the MAP kinase pathway such as ERK1/2. We showed that EGR1 is enabled to upregulate the transcription of eNOS an essential regulator of valve homeostasis.


Amélie GASTE (Marseille), Eric BERTRAND, Jean-François AVIERINOS, Valérie DEPLANO, Stéphane ZAFFRAN
17:45 - 20:00 #30598 - 244. Leukocytes play a role in the progression of Mitral Valve Dystrophy rather than its onset.
244. Leukocytes play a role in the progression of Mitral Valve Dystrophy rather than its onset.

INTRODUCTION: Understanding Mitral Valve Dystrophy (MVD) pathophysiological mechanisms is a current challenge. We have generated a unique knock-in (KI) animal model bearing the FlnA-P637Q mutation associated with MVD in humans and confirmed the presence of MVD in 3-week-old KI rats. The main signaling pathway identified by RNA-seq referred to immune cell recruitment, along with extracellular matrix remodeling and response to molecular stress. Those findings were corroborated with cytometry showing the increased proportion of myeloid cells in 3-week-old KI MV.

OBJECTIVE: The aim of the present study was to analyze earlier developmental time points and the specific role of endothelial, interstitial and immune cells in the onset and the progression of MVD.

METHODS: 7-day post-natal (D7) KI and WT rats were studied. Classical histology, FACS and qPCR experiments were performed to determine the presence and the nature of the valve remodeling, the proportion and the molecular signature for each cellular subpopulation.

RESULTS: At D7, histological analyses showed a myxomatous remodelling in KI MV with thickened and proteoglycan-rich MV as previously found at 3-week-old. On the other hand, contrary to 3-week-old, no difference was observed in myeloid cell proportion at this age (6% vs 6% for 12 WT and 10 KI animals, respectively; p=0.63). CD45+ leukocytes and CD206+ developmental macrophages were located more diffusely in KI MV. The ongoing molecular signature characterization for the different cellular subpopulation uses primers targeting Esm1, Cspg4, Wnt2 and Grem1.

CONCLUSIONS: Our results revealed a specific role of myeloid cells in the progression of MVD rather than the onset of the disease. The typical inflammatory, ECM, and cellular activation markers currently under investigation at D7 need to be tested at 3-week-old. Cellular cross-talk of endothelial, interstitial and immune cells needs to be further studied to decipher the molecular mechanisms leading to MVD.


Constance DELWARDE (Nantes), Benjamin LE VELY, Amir H. KAYVANJOO, Pascal AUMOND, Séverine REMY, Laurent MONASSIER, Claire TOQUET, Jean-Jacques SCHOTT, Thierry LE TOURNEAU, Elvira MASS, Jean MEROT, Romain CAPOULADE
17:45 - 20:00 #30601 - 246. Disruption of Notch1 and Gata5 in mice results in clinically relevant congenital aortic valve stenosis.
246. Disruption of Notch1 and Gata5 in mice results in clinically relevant congenital aortic valve stenosis.

Congenital aortic valve disease (AVD) is a most common type of congenital heart defect, but the underlying mechanisms remain largely unknown. NOTCH1 mutations are associated with AVD, and we previously reported that Notch1;Nos3 mutant mice display AVD. However, this model has limitations since ~65% of mutant mice suffer neonatal lethality. GATA5 is also implicated in human AVD, and Gata5-/- mice exhibit bicuspid aortic valve but with only 25% incidence, showing an associated reduction in Notch signaling. We hypothesized that the interaction of Notch1 and Gata5 was critical for aortic valve morphogenesis, and generated novel murine models by intercrossing Notch1 and Gata5 heterozygote mice. Mice heterozygous for Notch1 and Gata5 (Notch1+/-;Gata5+/-), and heterozygous for Notch1 and null for Gata5 (Notch1+/-;Gata5-/-) demonstrated no neonatal lethality. Echocardiographic analyses demonstrated aortic valve stenosis (AVS) in ~50% of Notch1+/-;Gata5+/- mice and ~90% of Notch1+/-;Gata5-/- mice by 16 weeks of age. Notably, progression of AVS, as measured by an increase in aortic velocity from 6 to 16 weeks, was found in Notch1;Gata5 compound mutant mice. Furthermore, thickened and malformed aortic valves were found by histological examination in Notch1;Gata5 compound mutant mice at 16 weeks and 1 year as well as embryonic day 18.5 and postnatal day 10, consistent with a congenital phenotype. Interestingly, the expression of NICD and Nos3 was decreased in aortic valves of Notch1;Gata5 compound mutant mice as compared to wildtype littermates. In conclusion, our findings demonstrate a novel genetic interaction between Notch1 and Gata5. These new compound mutant mouse models display highly penetrant congenital and progressive AVS without lethality, and allow for unbiased genomic approaches to identify novel therapeutic targets for disease initiation and progression.


Jun YASUHARA (Columbus, USA), Uddalak MAJUMDAR, Yukie UEYAMA, Sara ADAMCZAK, Emily CAMERON, Vidu GARG
17:45 - 20:00 #30630 - 248. Mechanisms of neutrophil extravasation and migration.
248. Mechanisms of neutrophil extravasation and migration.

Neutrophils typically roll along the endothelium, ready to extravasate at a moment’s notice to the sites of tissue damage and/or pathogen invasion. The ability of extravasated neutrophils to re-enter the vasculature was first described in the zebrafish and later confirmed in the mouse model, establishing its relevance to mammalian systems. This endothelial re-entry has implications for  a rapid termination of inflammatory cytokine release by neutrophils. We examined neutrophil migration using zebrafish embryos at 2 days of development. After tail fin amputation, embryos were treated with selective chemical compounds targeting a number of receptor tyrosine kinases and intracellular signaling molecules (VEGFR, PDGFR, Eph, Abl, PI3K, mTOR and gamma-secretase) at concentrations that did not induce visually observable harm to tissues and the functioning vasculature. At least 50 embryos were scored for neutrophil migration at the end of 2h. Robust immobilization of neutrophils was observed upon treatment with selective inhibitors for the PI3K-delta and PI3K-gamma isoform (AS605240 and AS-252424), but not with pan-PI3K or PI3K-alpha-selective inhibitors. Unexpectedly, treatment with DAPT, an inhibitor of gamma-secretase, accelerated neutrophil migration. This observation and was confirmed using another, structurally distinct compound, PF03084014. So far, neutrophil-active compounds identified through library screening have reduced neutrophil migration rates. One compound derived from a medicinal extract, tanshinone IIA (protein target unknown), accelerated reverse migration. From our work, gamma secretase activity appears to negatively regulate directed, injury-induced neutrophil migration rates. Additional assays are underway to examine how these chemicals may alter netosis and/or vascular re-entry. Results from this study could provide novel targets for the development of neutrophil modifying drugs for research and for therapy.

 


Joanne CHAN (Tampa, USA), John MABLY
17:45 - 20:00 #30637 - 250. A functional investigation of cardiovascular defects in adult zebrafish.
250. A functional investigation of cardiovascular defects in adult zebrafish.

Although most cardiovascular defects result from genetic mutations, many of the symptoms may be detectable only in adulthood.  Zebrafish is a well-established genetic model to investigate cardiovascular development, disease and regeneration.  However, the tools to analyze cardiovascular adult phenotypes in vivo remain limited.

Here, we combine multiple imaging techniques including echocardiography, magnetic resonance imaging, micro-computed tomography, electrocardiography, and light-sheet microscopy to study cardiovascular phenotypes in adult zebrafish.

To determine the sensitivity of our approach we analyzed genetic models with variable and mild cardiovascular phenotypes only manifested in adulthood.  Using these mutant and cell-ablation models, we identified functional impairment resulting from morphological alterations.  Specifically, we performed a correlation analysis of the different parameters and found that cardiac valve injury and reduced outflow tract wall thickness cause blood regurgitation, confirming the association between haemodynamic defects and structural alterations of the heart.

Overall, we propose a new approach to identify phenotypes in adult zebrafish that are too variable or subtle to identify with conventional histological analyses.  Importantly, this study helps set the zebrafish as an important model to analyze adult cardiovascular phenotypes potentially translatable to clinical settings.


Anabela BENSIMON-BRITO (Marseille), Giulia L. M. BOEZIO, Joao CARDEIRA-DA-SILVA, Astrid WIETELMANN, Inês CRISTO, Ana Beatriz BARBOSA, Srinath RAMKUMAR, Pia R. LUNDEGAARD, Christian S. M. HELKER, Radhan RAMADASS, Janett PIESKER, Arno NAUERTH, Clemens MUELLER, Didier Y. R. STAINIER
17:45 - 20:00 #30689 - 252. Differential Etv2 threshold requirement for endothelial and erythropoietic development.
252. Differential Etv2 threshold requirement for endothelial and erythropoietic development.

Endothelial and erythropoietic lineages arise from a common developmental progenitor.  Etv2 is a master transcriptional regulator required for the development of both lineages.  However, the mechanisms through which Etv2 initiates the gene regulatory networks (GRNs) for endothelial and erythropoietic specification and how the two GRNs diverge downstream of Etv2 remain incompletely understood.  Here, by analyzing a hypomorphic Etv2 mutant, we demonstrate different threshold requirements for initiation of the downstream GRNs for endothelial and erythropoietic development.  We show that Etv2 functions directly in a coherent feedforward transcriptional network for vascular endothelial development, and a low level of Etv2 expression is sufficient to induce and sustain the endothelial GRN.  In contrast, Etv2 induces the erythropoietic GRN indirectly via activation of Tal1, which requires a significantly higher threshold of Etv2 to initiate and sustain erythropoietic development.  These results provide important mechanistic insight into the divergence of the endothelial and erythropoietic lineages.


Tanvi SINHA, Kelly LAMMERTS VAN BUEREN, Diane E. DICKEL, Ivana ZLATANOVA, Reuben THOMAS, Carlos O. LIZAMA, Shan-Mei XU, Ann C. ZOVEIN, Kohta IKEGAMI, Ivan P. MOSKOWITZ, Katherine S. POLLARD, Len A. PENNACCHIO, Brian L. BLACK (San Francisco, USA, USA)
20:00 Reception at the Musée Regards de Provence
Friday 13 May
08:45

"Friday 13 May"

Added to your list of favorites
Deleted from your list of favorites
PS3
08:45 - 10:45

Session III
CHD Mechanisms II

Moderators: Brian BLACK (Attendee, Poster Presenter) (San Francisco, USA, USA), Kelly SMITH (Group Leader) (Melbourne, Australia)
08:45 - 10:45 #30588 - 011. A phenotypic rescue approach identifies lineage regionalization defects in a mouse model of DiGeorge syndrome.
011. A phenotypic rescue approach identifies lineage regionalization defects in a mouse model of DiGeorge syndrome.

TBX1 is a key regulator of pharyngeal apparatus (PhAp) development. Vitamin B12 treatment partially rescues aortic arch patterning defects of Tbx1+/- embryos. Here we show that it also improves cardiac outflow tract septation and branchiomeric muscle anomalies of Tbx1 hypomorphic mutants. At molecular level, the in vivo vB12 treatment let us to identify genes that were dysregulated by Tbx1 haploinsufficiency and rescued by treatment. We found that SLUG, encoded by the rescued gene Snai2, identified a population of mesodermal cells that was partially overlapping with but distinct from ISL1+ and TBX1+ populations. In addition, SLUG+ cells were mislocalized and had a greater tendency to aggregate in Tbx1+/- and Tbx1-/- embryos and vB12 treatment restore cellular distribution. Adjacent neural crest-derived mesenchymal cells, which do not express TBX1, were also affected, showing enhanced segregation from cardiopharyngeal mesodermal cells. We propose that TBX1 regulates cell distribution in core mesoderm and the arrangement of multiple lineages within the PhAp.


Gabriella LANIA, Gabriella LANIA (naples, Italy), Monica FRANZESE, Noritaka ADACHI, Annalaura RUSSO, Erika D'AGOSTINO, Claudia ANGELINI, Robert G. KELLY, Antonio BALDINI
08:45 - 10:45 #29531 - 012. A deeply conserved GATA4/5/6-dependent cis-regulatory architecture in cardiac development and disease.
012. A deeply conserved GATA4/5/6-dependent cis-regulatory architecture in cardiac development and disease.

Mutations in the deeply conserved transcription factors and their bound cis-regulatory elements (CREs), which orchestrate early cardiac development, may lead to heart disease. GATA4/5/6 paralogs function near the top of the cardiac regulatory network, with essential roles in heart development. Leveraging a zebrafish heartless modelGata5/6 knockdown, we identified 1,470 differentially accessible regions (DARs) with reduced accessibility upon Gata5/6 loss (GATA-dependent DARs) at mid-gastrulation, prior to nkx2.5 expression. GATA motifs were strongly enriched in these CREs, suggesting a direct role for Gata5/6 in their activity.

To further enrich for functional cardiac CREs, we applied a comparative genomics approach. We hypothesize that GATA-dependent DARs with deep evolutionary conservation likely contribute to early cardiac development and disease. To this end, we identified 47 mesendodermal-specific GATA-dependent DARs as accessible regions conserved between zebrafish and human, which we termed GATA-dependent accessible conserved non-coding elements (GaCNEs). 17 out of 18 GaCNEs tested displayed cardiac activity in transgenic zebrafish embryos. Supporting functional conservation, three GaCNEs were identified as being GATA4 targets in human cardiomyocytes and accessible in cardiac progenitors. Deletion of GaCNE1 near hand2 resulted in reduced hand2 expression and laterality defects, including cardiac patterning defects. The remaining two GaCNE contain hits for ultra-rare variants in patients with congenital heart disease. One such region near TBX20 interacts with the TBX20 promoter in the human mesoderm. We are actively testing the impacts of patient variants in these GaCNEs.

Collectively, we identified deeply conserved GATA-dependent cardiac CREs, which likely contribute to cardiac development and disease. We propose a broadly applicable framework for identifying functional and disease-associated CREs by combining phenotypic perturbation and comparative genomics.


Mengyi SONG (Toronto, Canada), Anna PRENTICE, Xuefei YUAN, Huayun HOU, Casey CARLISLE, Eleanor DUNCAN, Cherith SOMERVILLE, Simon MONIS, Raymond KIM, Rebekah JOBLING, Ian SCOTT, Michael WILSON
08:45 - 10:45 #29553 - 013. FOXF1 mediates Hedgehog signaling-dependent heterochronic control of cardiac development.
013. FOXF1 mediates Hedgehog signaling-dependent heterochronic control of cardiac development.

Coordination of developmental timing is critical to embryogenesis. Dysregulated development results in birth defects, the leading cause of neonatal death. We have recently demonstrated that Hedgehog signaling is a heterochronic regulator of cardiac development. Specifically, Hedgehog signaling is required in the second heart field (SHF) to prevent precocious cardiomyocyte differentiation and congenital heart disease. We have identified the forkhead transcription factor FOXF1 as a Hedgehog target in the SHF and a mediator of Hedgehog-dependent heterochronic control of differentiation. Removal of FOXF1 from the SHF causes loss of SHF cardiac progenitors accompanied by inappropriate myocardialization of this tissue. To explore the mechanism by which FOXF1 controls developmental timing, we generated mESCs harboring inducible FOXF1. Overexpression of FOXF1 in differentiating cardiac progenitors delays cardiomyocyte gene expression and prolongs expression of progenitor markers. Principal component analysis of time course RNA-seq indicates deployment of FOXF1 prevents progress along the cardiac differentiation trajectory. Integration of FOXF1 ChIP-seq, ATAC-seq, and H3K27Ac ChIP-seq suggests FOXF1 acts as a pioneer factor to regulate a cardiac differentiation gene network: FOXF1 binds to and establishes accessibility of enhancers of cardiac differentiation genes that subsequently gain H3K27Ac after FOXF1 removal. Together, these studies indicate FOXF1 is a functional regulator of Hedgehog signaling-dependent heterochronic control of cardiomyocyte differentiation in the SHF. Current results suggest a model in which signal-dependent expression of FOXF1 in the SHF may prime, but temporarily prevent the onset of, the cardiac differentiation gene regulatory network.

 

Funding sources:

University of Chicago Cardiovascular Sciences Training Program

NIH NHLBI R01 HL147571

NIH NHLBI T32 HL007381


Joshua W. M. THEISEN (CHICAGO, USA), Ariel RYDEEN, Jeffrey D. STEIMLE, Megan ROWTON, Carlos PEREZ-CERVANTES, Xinan Holly YANG, Jessica JACOBS-LI, Suzy HUR, Alex M. HOOVER, Sunny Sun-Kin CHAN, Michael KYBA, Ivan P. MOSKOWITZ
08:45 - 10:45 #30328 - 014. Conserved transcriptional mechanisms directing Nr2f1 expression in vertebrate atria.
014. Conserved transcriptional mechanisms directing Nr2f1 expression in vertebrate atria.

Proper atrial chamber development is critical for the heart to function normally throughout vertebrate life. While Nr2f1 and Nr2f2 transcription factors (TFs) have conserved requirements promoting vertebrate atrial cardiomyocyte (AC) differentiation, the regulatory logic directing their expression in vertebrate ACs remains obscure. To elucidate mechanisms controlling Nr2f expression within vertebrate ACs, we examined the ~1.8 kb promoter of the zebrafish nr2f1a did not show specific expression in the ACs. Therefore, we explored conserved CRMs through comparing accessible chromatin in isolated ACs using ATAC-seq surrounding 730 kb of the zebrafish nr2f1a locus to vertebrate Nr2f1/2 loci using VISTA and manual alignments. Exploring the expression of CRMs using GFP transgenic reporters, we identified a 280 bp enhancer 3’ to nr2f1a, termed 3’reg1, which is conserved in gnathostome Nr2f1 loci and directs expression in ACs. Transcription factor binding site analysis showed that within 3’reg1 there are conserved sites for FoxF and Tcf, a transcriptional repressor relieved by Wnt signaling, which are TFs within a gene regulatory network (GRN) controlling venous pole development in mice. Mutating each of the FoxF sites and the Tcf site, respectively, reduced and expanded 3’reg1 expression in ACs and throughout the heart, while overexpression of murine Foxf1 mRNA and abrogation of endogenous Tcf7l1a caused pan-cardiac expression of 3’reg1. Furthermore, FoxF1 mRNA and pharmacological activation of Wnt signaling were sufficient to increase Nr2f1a+ cell number in atrium and atrial differentiation, supporting that these factors regulate endogenous Nr2f1a atrial expression. Also, an epistatic approach showed that Tcf7l1a functionally interacts with Foxf1, limiting its role as activator. Thus, Wnt signaling and Foxf1 promote zebrafish nr2f1a expression in ACs through a conserved 3’ CRM, providing new insights into a conserved GRN directing vertebrate atrial development. (NIH, AHA)


Ugo COPPOLA (Cincinnati, USA), Joshua S. WAXMAN
08:45 - 10:45 #29496 - 015. Ribosomal protein genes as a novel class of Congenital Heart Disease candidates regulating cardiac growth and proliferation in concert with cardiac transcription factors.
015. Ribosomal protein genes as a novel class of Congenital Heart Disease candidates regulating cardiac growth and proliferation in concert with cardiac transcription factors.

Hypoplastic Left Heart Syndrome (HLHS) mainly characterized by an underdeveloped left ventricle and is the most severe Congenital Heart Disease (CHD). Its etiology is poorly understood but is likely oligogenic. Sequencing efforts identified thousands of putative human disease variants, however, establishing genotype-phenotype relationships remains challenging. We therefore perform high-throughput in vivo functional analyses of candidate genes using the fly heart, human iPSC-cardiomyocytes (hiPSC-CMs), and zebrafish to interrogate their potential contributions in CHD/HLHS.

Whole-genome-sequencing of HLHS proband-parent trios with poor clinical outcome and GO enrichment analysis of prioritized genes revealed an over-representation for ribosomal protein (RP) genes. In patient-derived iPSC-CM carrying an RPS15A variant proliferation was reduced compared to the parents. Knockdown of variant-carrying RP genes in generic hiPSC-CMs also reduced proliferation and triggered a transcriptomic response consistent with nucleolar stress. In adult flies, RP knockdown resulted in partial/complete heart loss or reduced contractility. Consistent with nucleolar stress, reducing RpS15Aa function increased the size of cardiomyocyte nucleoli. In zebrafish, rps15a knockdown led to reduced CM numbers and/or contractility, and defective heart looping. Probing for cardiac-specific RP functions, we found conserved, synergistic interactions between RPS15A and cardiac transcription factors tinman/Nkx2.7 and dorsocross/Tbx5a/TBX5 in Drosophila, zebrafish, and hiPSC-CMs. RPS15A knockdown-induced defects were significantly reversed (1) by p53 co-knockdown in hiPSC-CMs and zebrafish, or (2) by YAP/yorkie overexpression or myc co-knockdown in flies.

We conclude that RP genes play a critical role in cardiac growth/ CM proliferation, likely in conjunction with cardiogenic genes, thus representing a potential novel class of genetic effectors in CHD/HLHS.


Tanja NIELSEN (San Diego, USA), Anais KERVADEC, X-X. I ZENG, Analyne SCHROEDER, Jeanne L THEIS, Timothy M. OLSON, Karen OCORR, Alexandre COLAS, Georg VOGLER, Rolf BODMER
08:45 - 10:45 #29492 - 016. Modeling MYBPC3-related Hypertrophic Cardiomyopathy and Left Ventricular Non-Compaction in mice.
016. Modeling MYBPC3-related Hypertrophic Cardiomyopathy and Left Ventricular Non-Compaction in mice.

Familial cardiomyopathies are severe, incurable, genetic diseases with heterogenous phenotypes, as pedigrees may contain individuals showing features of more than one cardiomyopathy. This is the case for Hypertrophic cardiomyopathy (HCM) and Left ventricular Non-Compaction (LVNC), two distinct conditions that can occur simultaneously and share a common genetic basis. Several mutations affecting MYBPC3, encoding for MYOSIN binding protein C, have been identified in familial cases of mixed LVNC and HCM. Using CRISPR-Cas9 technology, we generated three mouse models carrying two different Mybpc3 nonsense and one missense variants, aiming to understand if the nature of the various mutations influences the cardiomyopathy phenotype. We found that mouse homozygous for both Mybpc3 nonsense variants (Mybpc3 P109Sfs*12 and Mybpc3 R887Afs*160) developed a transient hypertrabeculation phenotype during late fetal development that resulted into postnatal cardiac hypertrophy, while the missense variant has caused so far no functional or structural deficiency. We have transcriptionally characterized the hypertrabeculation to hypertrophy transition in order to identify the molecular mechanisms driving this apparent phenotypic conversion. We have used the Hey2 CreERT2 driver to analyze the contribution of the compact myocardium derivatives to the onset of the hypertrabeculation phenotype and the later thickening of the ventricular wall. We suggest that establishing if HCM and LVNC have a common genetic and developmental substrate will contribute to understanding disease etiology and the design of potential therapeutic treatments.


Alejandro SALGUERO-JIMÉNEZ (Madrid, Spain), María SABATER-MOLINA, Bin ZHOU, Juan R. GIMENO-BLANES, José Luis DE LA POMPA
Auditorium
10:45 Break
11:15

"Friday 13 May"

Added to your list of favorites
Deleted from your list of favorites
PS4
11:15 - 13:00

Session IV
Conduction System

Moderators: Vincent CHRISTOFFELS (PI) (Amsterdam, The Netherlands), Lucile MIQUEROL (PI) (MARSEILLE, France)
11:15 - 13:00 #29424 - 017. Hippo signaling pathway maintains sinoatrial node homeostasis.
017. Hippo signaling pathway maintains sinoatrial node homeostasis.

The sinoatrial node (SAN) functions as pacemaker of the heart to initiate and drive rhythmic heartbeats. The Hippo signaling pathway is a fundamental pathway for heart development and regeneration. Although abnormalities of Hippo pathway are associated with cardiac arrhythmias in human patients, yet its role in the SAN is unknown. We investigated key regulators of the Hippo pathway in the sinoatrial node by conditionally inactivating the Hippo signaling kinases Lats1 and Lats2 using the tamoxifen-inducible, cardiac conduction system (CCS)-specific Cre driver Hcn4CreERT2 and Lats1/2 conditional knockout (CKO) alleles. In addition, the Hippo signaling effectors Yap and Taz were conditionally inactivated in the sinoatrial node. To determine the function of the Hippo signaling pathway in the SAN and other CCS components, we conducted a series of physiological and molecular experiments like telemetry ECG recording, calcium imaging, CUT&Tag sequencing. We found that Lats1/2 inactivation caused severe sinoatrial node dysfunction. Compared to the controls, Lats1/2 CKO mutants exhibited dysregulated calcium handling and increased fibrosis in the sinoatrial node, indicating Lats1/2 function through both cell-autonomous and non-cell-autonomous mechanisms. Notably, the Lats1/2 CKO phenotype was rescued by genetic deletion of Yap and Taz in the CCS, and these rescued mice had normal sinus rhythm and reduced fibrosis of the sinoatrial node, indicating that Lats1/2 function through Yap and Taz. CUT&Tag sequencing data showed that Yap regulates genes critical for calcium homeostasis such as Ryr2 and genes encoding paracrine factors important in intercellular communication and fibrosis induction such as Tgf-β1/3. Consistently, Lats1/2 CKO mutants had decreased Ryr2 expression and increased Tgf-β1/3 expression compared with control mice. Together, we reveal for the first time that the canonical Hippo-Yap pathway has a pivotal role in functional homeostasis of the sinoatrial node.


Mingjie ZHENG (Houston, USA)
11:15 - 13:00 #30517 - 018. IRX5 transcription factor cooperates with the TBX5/GATA4/NKX2-5 complex to regulate human cardiomyocyte functions.
018. IRX5 transcription factor cooperates with the TBX5/GATA4/NKX2-5 complex to regulate human cardiomyocyte functions.

Introduction: Transcription factors (TFs) are key regulators of gene networks governing cardiac development and function, but the role of some TFs such as the Iroquois homeobox (IRX) remains largely unknown. Irx5 participates to ventricular repolarization gradient in mice and IRX5 mutations (Hamamy syndrome) cause defects in various organs including heart and limbs. Interestingly, TBX5 mutations (Holt-Oram syndrome) also lead to heart/limb defects. Since TBX5 is part of a TF complex with GATA4 and NKX2-5, we investigated a link between IRX5 and this TF complex.

Methods: To elucidate whether these TFs physically and/or functionally interact, we used co-immunoprecipitation and luciferase assays on HEK293 cells expressing either mutated or truncated IRX5 proteins. IRX5 ChIP-Seq data were obtained from cardiomyocytes derived from induced pluripotent stem cells generated from 2 healthy individuals and compared to already published ChIP-Seq data for TBX5, GATA4 and NKX2-5.

Results: IRX5 and TBX5, GATA4, NKX2-5 physically interacted by pairs, but also as a multiprotein complex. Using IRX5 mutated and truncated proteins, its homeodomain was identified as necessary for its protein-protein interactions. Luciferase assays with successive addition of different TFs increased SCN5A expression: NKX2-5 alone strongly (5.2 fold vs. no TF) activated SCN5A expression that was further potentiated by IRX5/GATA4 addition (9.8 fold vs. no TF), and the activation was maximal when TBX5 was present as well (12.2 fold vs. no TF). Finally, ChIP-Seq data analysis identified 2253 and 2990 genes bound by IRX5 alone and all 3 TBX5, GATA4 and NKX2-5 TFs, respectively. Interestingly, 848 genes were bound by all 4 TFs and associated to electrical activity and fibrosis signaling biological processes.

Conclusion: Our data show new physical and functional interactions between IRX5 and 3 key cardiac TFs (TBX5, GATA4 and NKX2-5), suggesting new regulatory roles for IRX5 in specific human cardiac functions.


Robin CANAC (Echirolles), Bastien CIMAROSTI, Aurore GIRARDEAU, Virginie FOREST, Cynthia FOURGEUX, Jeremie POSCHMANN, Guillaume LAMIRAULT, Patricia LEMARCHAND, Nathalie GABORIT
11:15 - 13:00 #30552 - 019. The cis-regulatory architecture of a gene desert controlling pleiotropic Shox2 expression and cardiac pacemaker development.
019. The cis-regulatory architecture of a gene desert controlling pleiotropic Shox2 expression and cardiac pacemaker development.

The Shox2 transcription factor is essential for sinoatrial node (SAN) pacemaker cell differentiation in mice and associated with SAN dysfunction and arrhythmias in humans. At the genomic level, Shox2 is flanked by a gene desert, an extensive non-coding region spanning more than 500 kilobases. Despite their proposed critical roles in development, the enhancer architecture and biological functions of most gene deserts in mammalian genomes remain unknown. Here, by using a combination of epigenomic profiling, transgenic reporter assays and CRISPR-Cas9 genome editing, we uncover the transcriptional in vivo enhancer landscape within the Shox2 gene desert, which is essential for pleiotropic Shox2 expressionIn particular, we find that the gene desert controls Shox2 in the SAN and consequently is required for embryonic survivalUsing single-cell multiomics across key stages of mouse heart developmentwe identify a network of putative SAN-enhancers correlating with Shox2 expression in single nuclei and exhibiting histone acetylation marks in human fetal cardiac compartments. Genomic deletion analysis confirmed the functional requirement of one of these enhancers for robust expression of cardiac Shox2 in vivoIn addition, while the Shox2 regulatory landscape is partitioned into largely tissue-invariant chromatin architecture, region capture Hi-C uncovered an unexpected cardiac-specific contact domain within the gene desert, potentially acting as a topological mechanism for enhancer attenuation. In summary, our results serve as a blueprint to investigate gene desert function in cardiac development and identify the Shox2 gene desert as a robust cis-regulatory hub indispensable for pleiotropic patterning, cardiac pacemaker differentiation and embryogenesis.


Marco OSTERWALDER (Bern, Switzerland), Samuel ABASSAH-OPPONG, Brandon J. MANNION, Raquel ROUCO, Matteo ZOIA, Virginie TISSIERES, Virginia ROLAND, Iros BAROZZI, Diane E. DICKEL, Javier LOPEZ-RIOS, Guillaume ANDREY, Len A. PENNACCHIO, Axel VISEL, John COBB
11:15 - 13:00 #29557 - 020. A multi-model system approach identifies genetic interactions underlying Atrial Fibrillation Susceptibility.
020. A multi-model system approach identifies genetic interactions underlying Atrial Fibrillation Susceptibility.

Atrial fibrillation (AF), the most common heart rhythm disorder, is reaching epidemic proportions in aging populations and there is no cure. Over 200 genetic variants are associated with AF susceptibility, suggesting that the underlying cause is multifactorial. It is not understood how aging, genetic predispositions, and environmental factors synergize to promote arrhythmia, nor which gene regulatory networks initiate and maintain AF. Resolving complex interactions is difficult in mammalian systems, but approachable in the genetically tractable Drosophila cardiac-aging model and in hIPSC-atrial-like cardiomyocytes (ACM). Both models allow for high-throughput quantification of functional effects in response to combinatorial gene knockdown (KD). High-speed imaging permits quantification of cardiac parameters including action potential duration and arrhythmicity in ACMs, and contraction intervals and arrhythmicity in flies. Screening candidate genes in both platforms has identified seven corroborating hits. Network analysis linked these genes to ion channels, such as atrial-specific K+ channel Shaker (Sh), key developmental transcription factors Dorsocross (Doc) and Pannier (Pnr), as well as structural components such as Sarcolamban (SclA). Single genetic insult rarely produces robust arrhythmicity in our models, but we see increases in arrhythmicity when incorporating age and/or testing interactions between genes in this network. KD of Doc, a T-box transcription factor, in a Sh heterozygote background produces robust arrhythmicity, even at younger ages. In addition to commonly studied electrosensitive ion channel candidates, we took an alternative approach with Piezo, a mechanosensitive channel.  Piezo KD significantly increased arrhythmicity and long fibrillatory-like events, especially in older flies. Identifying interactions of genetic insult, age, and external factors that contribute to AF susceptibility will guide novel experimental and therapeutic strategies.


James KEZOS (La Jolla, USA), Anais KERVADEC, Alexandre COLAS, Philippe CHEVALIER, Karen OCORR
11:15 - 13:00 #30652 - 021. The zebrafish grime mutant uncovers an evolutionarily conserved role for Tmem161b in the control of cardiac rhythm.
021. The zebrafish grime mutant uncovers an evolutionarily conserved role for Tmem161b in the control of cardiac rhythm.

The establishment of cardiac function in the developing embryo is essential to ensure blood flow and, therefore, growth and survival of the animal. The molecular mechanisms controlling normal cardiac rhythm remain to be fully elucidated. From a forward genetic screen, we identified a unique mutant, grime, that displayed a specific cardiac arrhythmia phenotype. We show that loss-of-function mutations in tmem161b are responsible for the phenotype, identifying Tmem161b as a regulator of cardiac rhythm in zebrafish. To examine the evolutionary conservation of this function, we generated knockout mice for Tmem161b. Tmem161b knockout mice are neonatal lethal and cardiomyocytes exhibit arrhythmic calcium oscillations. Mechanistically, we find that Tmem161b is expressed in excitable cells and live imaging shows it is required for action potential repolarisation in the developing heart. Electrophysiology on isolated cardiomyocytes demonstrates that Tmem161b is essential to inhibit Ca2+ and K+ currents in cardiomyocytes. Importantly, Tmem161b haploinsufficiency leads to cardiac rhythm phenotypes, implicating it as a candidate gene in heritable cardiac arrhythmia. Overall, this data describes Tmem161b as a highly conserved and novel regulator of cardiac rhythm that functions to modulate ion channel activity in zebrafish and mice.


Kelly SMITH (Melbourne, Australia)
Auditorium
13:00 Lunch Young investigators Career Lunch with Didier Stainier, Brian Black and Rolf Bodmer
14:00

"Friday 13 May"

Added to your list of favorites
Deleted from your list of favorites
PS5
14:00 - 15:50

Session V
Single Ventricle

Moderators: Anthony FIRULLI (Principal Investigator) (Indianapolis, USA), José Luis DE LA POMPA (Researcher. Group Leader) (Madrid, Spain)
14:00 - 14:10 Session introduction by Additional Ventures. Kaitlin DAVIS (Program Manager) (Speaker, Palo Alto, USA)
14:00 - 15:50 #30531 - 022. The role of RNA-binding protein QKI in ventricular wall development.
022. The role of RNA-binding protein QKI in ventricular wall development.

QKI is a unique RNA-binding protein within hnRNP K-homology domain family. Recently, we have found a rare de novo mutation of QKI in a Hypoplastic Left Heart Syndrome (HLHS) patient with an in-frame 88-amino acid deletion that effectively removes the critical Src-PTK binding site. By analyzing a QkiLacZ reporter mouse line, QKI expression is found in the developing heart as early as E7.5 and remained in both cardiomyocytes and endocardial cells. This expression pattern is well maintained in the adult mouse heart. QkiLacZ/LacZ mutant embryos die around E10.5 and show multiple developmental defects, including severe cardiac developmental defects with hypoplastic ventricular wall and collapsed endocardial structure. To determine the role of QKI in human cardiac development, we have generated mutant human embryonic stem cells (hESCs) that are deficient in QKI (QKIdel) using CRISPR/Cas9 genomic editing technology. hESCs-QKIdel maintain normal self-renewal activity and pluripotency. Transcriptomic analysis at single-cell resolution (scRNA-seq) further demonstrates that hESCs-QKIdel can efficiently differentiate into cardioprogenitor cells. However, these mutant cardioprogenitors have a slightly reduced level of proliferative activity and fail to produce functional cardiomyocytes. Bulk RNA-seq and replicate multivariate analysis of transcript splicing (rMATS) demonstrates dramatically altered pre-mRNA splicing events in key genes involved in myofibrillogenesis. More interestingly, endocardial cells derived from hESCs-QKIdel also exhibit a significant defect in proliferative activity, which is independent of cardiomyocyte defect, confirming that QKI is involved in cardiogenic events likely via both developing cardiomyocytes and endocardial cells. This finding was further confirmed by a series of conditional Qki knockout models, including cardiomyocyte- and endocardial-specific knockouts.


Ying LIU (Indianapolis, USA), Weinian SHOU
14:00 - 15:50 #30578 - 023. The novel MYH6-E1584K tail domain variant associated with hypoplastic left heart syndrome impairs cardiomyocyte relaxation in vitro.
023. The novel MYH6-E1584K tail domain variant associated with hypoplastic left heart syndrome impairs cardiomyocyte relaxation in vitro.

Hypoplastic left heart syndrome (HLHS) is a complex form of congenital heart disease (CHD) characterized by hypoplasia of the left ventricle and proximal aorta, as well as stenosis or atresia of the mitral and/or aortic valves. In a cohort of 190 HLHS patients, we previously identified 21 rare variants in the gene encoding for α-myosin heavy chain (MYH6α-MHC), with variant carriers exhibiting reduced cardiac transplant-free survival. These variants span the entire MYH6 gene and affect every functional domain. Pathological variants in MHC head domains directly alter force generationyet the mechanisms by which MHC tail domain variants cause contractile defects are less understood. In one HLHS family, we identified a novel MYH6-E1584K tail domain variant, located in the third skip residue of the α-MHC tail. Skip residues are a structural feature unique to the sarcomeric myosins; no previous disease-associated variants have been identified in any of the cardiac MHC skip residues.

In our double-blind analysis, patient-specific induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) with MYH6-E1584K showed significant sarcomere disarray compared to MYH6-WT iPSC-CMs. MYH7 (b-MHC) was upregulated in both atrial tissue from an HLHS patient carrying MYH6-E1584K and MYH6-E1584K iPSC-CMs. Bulk and single-cell functional analyses of patient-specific MYH6-E1584K iPSC-CMs demonstrated elongated relaxation times at differentiation days 25-30, with no apparent impact on contraction time or overall contraction rate. Notably, this is different than our previously reported findings of iPSC-CMs carrying the head domain variant MYH6-R443P, which exhibited decreased contraction velocity. This data suggests the cellular mechanism by which MYH6 variants cause HLHS differs based on the functional domain in which the specific variant lies. Furthermore, these findings may provide a framework upon which other MHC tail domain variants can be studied.


Melissa ANFINSON, Melissa ANFINSON (Milwaukee, WI, USA), Suma THAREJA, Ryan BROWN, Matthew CAVANAUGH, Karl STAMM, Huan-Ling LIANG, Peter LAMBERTON, Jackson RADANDT, Min-Su KIM, Michael MITCHELL, Robert FITTS, Aoy TOMITA-MITCHELL
14:00 - 15:50 #29794 - 024. Torsion of the heart tube by shortage of progenitor cells: identification of Greb1l as a genetic determinant of criss-cross hearts in mice.
024. Torsion of the heart tube by shortage of progenitor cells: identification of Greb1l as a genetic determinant of criss-cross hearts in mice.

Criss-cross heart is a rare congenital heart defect (1/125 000) defined by abnormal twisting of the atrioventricular valves and often associated with malpositioned supero-inferior ventricles. However, the origin of this disease has remained unknown. Here, we identify Greb1l mutants as the first mouse model of criss-cross heart. Based on 3D quantifications of shape changes, we demonstrate that torsion of the atrioventricular canal occurs together with supero-inferior ventricles at E10.5, after heart looping. Mutants phenocopy specific features of partial deficiency in retinoic acid signalling, suggesting that GREB1L is a novel modulator of this signalling. Spatio-temporal gene mapping and cross-correlated transcriptomic analyses further reveal the role of Greb1l in maintaining a pool of precursor cells during heart tube elongation, by controlling ribosome biogenesis and cell differentiation. Growth arrest and malposition of the outflow tract are predictive of abnormal tube remodelling in mutants. Our work on a rare cardiac malformation opens novel perspectives on the origin of a broader spectrum of congenital defects associated with GREB1L in humans.


Segolene BERNHEIM (Paris), Adrien BORGEL, Jean-Francois LE GARREC, Emeline PERTHAME, Audrey DESGRANGE, Wojciech KREZEL, Francesca RAIMONDI, Damien BONNET, Lucile HOUYEL, Sigolène MEILHAC
14:00 - 15:50 #30508 - 025. Myocardial-intrinsic defects underlie an Rbfox-mediated zebrafish model of hypoplastic left heart syndrome.
025. Myocardial-intrinsic defects underlie an Rbfox-mediated zebrafish model of hypoplastic left heart syndrome.

Hypoplastic left heart syndrome (HLHS) is characterized by underdevelopment of left sided structures including the ventricle, valves, and aorta. Although the mechanisms of disease pathogenesis remain elusive due to a paucity of candidate genes and animal models, prevailing paradigm suggests that HLHS is a multigenic disease of co-occurring phenotypes. Here, we report that zebrafish lacking two orthologs of the RNA binding protein RBFOX2, a gene previously linked to HLHS in humans, display cardiovascular defects overlapping those in HLHS patients. In contrast to current paradigm, we demonstrate that co-existing ventricular, valve, and aortic deficiencies in rbfox mutant zebrafish arise secondary to impaired myocardial function as all three phenotypes are rescued when Rbfox is expressed specifically in the myocardium. On a molecular and cellular level, we find diminished expression and alternative splicing of sarcomere and mitochondrial components in rbfox-deficient hearts that compromise sarcomere assembly and mitochondrial respiration, respectively. Injection of human RBFOX2 mRNA restores ventricular structure and function in rbfox mutant zebrafish, while HLHS-linked RBFOX2 variants fail to rescue. Taken together, our data suggest that mutations in RBFOX2 are causal for HLHS pathogenesis and provide a complimentary paradigm for HLHS emergence where co-existing ventricular, valve, and aortic deficiencies have a monogenic etiology caused by myocardial dysfunction.


Mengmeng HUANG (Boston, USA), Alexander AKERBERG, Xiaoran ZHANG, Haejin YOON, Shakchhi JOSHI, Christopher NGUYEN, William PU, Marcia HAIGIS, Geoffrey BURNS, Caroline BURNS
14:00 - 15:50 #30115 - 026. The role of embryonic senescence in cardiac trabeculation and compaction.
026. The role of embryonic senescence in cardiac trabeculation and compaction.

Embryonic senescence is a novel process uncovered over the last decade and found as essential for proper embryonic development. We investigated whether this cell process could occur during heart development. We detected senescent cells at different stages of mouse cardiac development. γH2AX+ and then p21+ cells were enriched in trabeculae and the number of senescent cells transiently increased from early to late stages of trabeculation/compaction.  

We performed single cell RNA-sequencing of high tomato+ trabeculae myocytes dissociated  from  E13.5 and E16.5 hearts from embryos generated by breeding SmaCreERT2  with Rosa26tdtomato mice. We identified several clusters of senescent cells in trabeculae myocytes. We uncovered news genes as well as specific cell processes and signaling pathways involved in cardiac senescence.

Next, we used drugs that inhibit (Navitoclax) or exacerbate (Palbociclib) senescence to look at its impact on cardiac trabeculation and or compaction. High Resolution Episcopic Microscopy combined with Fractal analysis of embryonic hearts show that senescence has a major impact on ventricular compaction.

We are using mice with cardiac conditional deletion of VHL, which feature impaired metabolic switch at mid-gestation. It will allow us to identify whether O2, mitochondrial ROS and metabolism underlie trabeculae myocyte senescence.

Embryonic cardiac senescence is likely at the origin of ventricular compaction. If dysregulated, the cell process may be at the origin, of cardiac congenital diseases such as hypoplastic left ventricle and non-compaction syndrome often observed in rare metabolic diseases.


Audrey IBRE (Marseille), Michel PUCEAT
Auditorium
15:50 Break Auditorium
16:20

"Friday 13 May"

Added to your list of favorites
Deleted from your list of favorites
PS6
16:20 - 18:15

Session VI
Regeneration I - Inflammation and Signalling

Moderators: James MARTIN (professor) (houston texas, USA), Francesca ROCHAIS (Principal Investigator - Group Leader) (Marseille, France)
16:20 - 16:30 Session introduction by The Leducq Foundation. David TANCREDI (Speaker, France)
16:20 - 18:15 #29461 - 027. Characterising differences between the regenerative and non-regenerative immune response in Astyanax mexicanus.
027. Characterising differences between the regenerative and non-regenerative immune response in Astyanax mexicanus.

The human heart cannot regenerate following myocardial infarction and instead forms a fibrotic scar that impairs cardiac function and can lead to heart failure. Cardiac regeneration aims to treat the injured heart by stimulating the heart to repair itself. The Astyanax mexicanus is a uniquely suited cardiac regeneration model as it comprises two closely related populations: (1) ‘regenerative’ surface (SF) populations and (2) the ‘non-regenerative’ Pachόn (PF) cave population. The immune response to injury is known to be a key regulator of successful regeneration. However, how this response differs between SF and PF populations is unknown. To fully characterise the A. mexicanus immune response, single cell RNA-sequencing, differential gene expression analysis (DGE) and in situ hybridisation were used to determine the immune cell populations present in the heart at 1, 3, 7, 14 and 30 days post-cryoinjury (dpci). We found striking spatiotemporal differences in PF and SF myeloid dynamics. Immediately after injury, the PF show a stronger response to injury with a significantly greater influx of neutrophils into the wound. However, by 7dpci, this inflammatory response is resolved and DGE shows that PF neutrophils have returned to their uninjured state. In contrast, SF show a prolonged neutrophil response at 7 and 14dpci, characterised by a transcriptionally unique TNFα-IL-1β-NFκB2+ population. Future inhibitor studies will aim to determine the role of these late-stage inflammatory neutrophils in successful regeneration and explore whether this difference in immune response between successful regeneration and scarring could be a future novel immunomodulatory therapeutic target.


Helen POTTS (Oxford, United Kingdom), Madeleine LEMIEUX, William STOCKDALE, Esra SENGUL, Robin CHOUDHURY, Mathilda MOMMERSTEEG
16:20 - 18:15 #29530 - 028. Role of autophagy during cell competition in heart development and regeneration.
028. Role of autophagy during cell competition in heart development and regeneration.

Cell Competition (CC) is the process by which viable cells are eliminated from tissues by comparison with neighboring cells. Myc overexpression in a mosaic fashion induces CC in heart, a mechanism by which Myc-high cardiomyocytes actively eliminate neighboring cardiomyocytes with lower Myc levels. Our current aim is to explore the role of metabolism and autophagy on CC during heart development and regeneration.

Our results show that autophagy is activated during CC in embryonic hearts in both wildtype and Myc overexpressing neighbouring cardiomyocytes (iMOS-Myc). Genetic autophagy or mitochondrial fission downregulation by Atg7 deletion or Oma1 deletion respectively protects wildtype cardiomyocytes from apoptotic cell death owing to Myc dependent-CC. More interestingly, hypoxia, which induces autophagy in wildtype hearts, completely abolishes this lysosomal degradative process and CC in Myc-overexpressed hearts. Moreover, blocking the metabolic communication between cells by inhibiting monocarboxylate transporters (MCTs) prevents the reduction of wildtype cardiomyocytes and autophagy activation in Myc-overexpressed hearts. Together, these results suggest that some metabolites that are released through MCTs and whose levels can vary upon hypoxic conditions are implicated in mitophagy regulation during Myc- dependent CC.

On the other hand, we are studying the involvement of autophagy-dependent CC during cardiac regeneration. For such purpose, we have developed an inducible-traceable strategy for restricting autophagy by generating mosaic hearts with different levels of Atg7. Thus, the fitness comparison between cardiomyocytes with differences in autophagy induces the reduction of Atg7+/- cardiomyocytes population in a ROS dependent manner. Intriguingly, this effect only happens during the regenerative stage of the neonatal heart, suggesting potential implications of autophagy-dependent cell competition for cardiac regeneration.


Lorena ESTEBAN-MARTÍNEZ (Madrid, Spain), Rocío SIERRA, Miguel TORRES
16:20 - 18:15 #30331 - 029. Spatial transcriptomics reveal a colocalized cellular triad required for heart renewal.
029. Spatial transcriptomics reveal a colocalized cellular triad required for heart renewal.

The heart is a poorly renewable organ, meaning that after injury tissue architecture fails to recover. This presents a major challenge in treatment of ischemic heart disease, the leading cause of death worldwide. To date, most studies have focused on cardiomyocytes. However, few studies have examined cardiomyocyte-extrinsic mechanisms, such as innate immunity in heart renewal. Here, we investigated cellular spatial relationships in control and a renewal-competent model in multiple cell-types to gain insight into inefficient mammalian heart renewal. Recognizing that the heart is composed of an integrated multicellular network, we used spatial transcriptomics combined with single cell RNA sequencing to examine organ wide cellular interactions in control and YAP5SA hearts. YAP5SA is a constitutively active form of the Hippo signaling effector YAP, and cardiomyocyte-specific expression of YAP5SA has been shown to be a robust model of adult cardiac proliferation. We found a proliferation competent cardiomyocyte population in control hearts distinguished by cytoskeletal and metabolic character; in YAP5SA hearts, almost all of the cardiomyocytes had this transcriptional signature with many undergoing proliferation and sarcomere disassembly. In control hearts, we observed colocalization of these proliferation competent cardiomyocytes with fibroblasts expressing complement genes, which regulate innate immunity. In YAP5SA hearts, we also found complement-expressing M2 polarized macrophages co-localized with the fibroblasts and cardiomyocytes. Clodronate-mediated ablation of all macrophages or genetic deletion of the M2 macrophages resulted in drastically reduced cardiomyocyte cell activity and sarcomere disassembly. We characterized a pro-renewal microenvironment composed of co-localized proliferation-competent cardiomyocytes, and complement-expressing fibroblasts and macrophages, supporting a novel role for innate immunity in the regulation of cardiomyocyte proliferation.


Gang (Rich) LI (Houston, USA), Francisco GRISANTI, Xiao LI, Yuka MORIKAWA, Fansen MENG, Jong KIM, Bing XIE, Shijie LIU, Md Abul Hassan SAMEE, James MARTIN
16:20 - 18:15 #30444 - 030. The macrophage regenerative response.
030. The macrophage regenerative response.

Macrophages are key regulators of regeneration and evidence for their involvement in cardiac regeneration has been demonstrated in a variety of different species. We have performed scRNA-seq of regenerating zebrafish hearts and our data has revealed a number of interesting features associated with macrophages during cardiac regeneration. Firstly,  in regenerating neonatal mouse hearts there is a rapid influx of Ccr2 expressing macrophages 1 day post MI coincident with elevated Tnfa and Il1b expression. Furthermore, at 3 days post MI, despite being more numerous, macrophages expressed lower levels of Tnfa and Il1b compared to sham operated animals. We observed a similar increase in macrophage cell number but reduction in tnfa and il1b expression within these cells in zebrafish hearts 3 days after injury. These data suggest that the inflammatory response of macrophages in regenerating adult zebrafish hearts is more reminiscent of the regenerating neonatal mouse macrophage response, however future studies at earlier timepoints will be required to confirm these observations. Our data also indicates that Mmp14b is particularly enriched in macrophages which appear at 3dpa. Furthermore, we observed that inhibiting the collagenolytic activity of Mmp14 resulted in defective migration of macrophages into the injury site and a subsequent failure to regenerate the myocardium, leading to the formation of large collagen/fibrin scar. Our scRNA-seq analysis of Mmp14 inhibited macrophages indicated that even at this early time point the expression of a number of genes associated with the regenerative response have become misregulated.


Chris JOPLING (Montpellier)
16:20 - 18:15 #30474 - 031. Re-activation of HIF signalling in the epicardium improves heart regeneration after myocardial infarction.
031. Re-activation of HIF signalling in the epicardium improves heart regeneration after myocardial infarction.

In the mouse cardiac regenerative capacity is maintained for the first week after birth but lost thereafter. Reactivation of this process holds great therapeutic potential, however, the molecular pathways underlying neonatal heart regeneration remain elusive. Here, we explore a role for the hypoxia-inducible factor (HIF) family of transcription factors (HIF-1α and HIF-2α) on the regulation of epicardial activity which is essential for the cardiac response to injury.

Expression of HIF-1α and HIF-2α in the epicardium was significant at early stages of heart development and gradually decreased throughout gestation. Postnatally, single cell RNA-sequencing associated with GO term analysis showed an enrichment of hypoxia-related pathways in P1 compared to P7 hearts, largely in the epicardial cell population.  Accordingly, expression of Phd2, encoding the main suppressor of HIF signalling, was enriched in P7 epicardial cells. Notably, the increase in Phd2 levels coincided with a clear reduction in the expression of Wilms’ tumour 1 (WT1) in P7 versus P1 hearts, a key player in the epicardial activation. In vitro studies confirmed regulation of WT1 expression and epicardial migration following l modulation of HIF signalling. Finally, in vivo pharmacological inhibition of PHD enzymes in P7 infarcted hearts led to prolonged epicardial activation, increased vascularisation, augmented injury resolution and preserved cardiac function up to 3 weeks after injury.

Together, these findings reveal that modulation of HIF signalling can regulate epicardial activity and extend heart regenerative capacity beyond the 7-day window. Pharmacological stabilisation of HIF signalling may represent a viable therapeutic strategy for treating adult ischaemic heart disease.


Elisabetta GAMEN (Oxford, United Kingdom), Eleonor L. PRICE, Daniela PEZZOLLA, Carla DE VILLIERS, Mala GUNADASA-ROHLING, Rafik SALAMA, David R MOLE, Tammie BISHOP, Chris W PUGH, Robin P CHOUDHURY, Carolyn A CARR, Joaquim M VIEIRA, Paul R RILEY
Auditorium
18:15

"Friday 13 May"

Added to your list of favorites
Deleted from your list of favorites
KS1
18:15 - 19:00

Keynote speaker I

Chairperson: Michel PUCEAT (PI) (Chairperson, marseille, France)
Speaker: Giacomo CAVALLI (Keynote speaker) (Speaker, Montpellier Cedex 5, France)
Auditorium
19:00

"Friday 13 May"

Added to your list of favorites
Deleted from your list of favorites
PO2
19:00 - 20:45

Poster session II (odd number posters)

19:00 - 20:45 #29347 - 049. The MyD88 signaling axis regulates the inflammatory response during adult heart regeneration in zebrafish.
049. The MyD88 signaling axis regulates the inflammatory response during adult heart regeneration in zebrafish.

Inflammation is triggered immediately after cardiac injury, and it has been hypothesized to promote regeneration in regenerative models such as zebrafish.  Myeloid differentiation factor 88 (MyD88) is a central adaptor molecule for the Toll-like receptor and interleukin-1 receptor signaling pathways, both of which are essential regulators of inflammation.  However, how specific molecules, such as MyD88, regulate inflammation after cardiac injury remains unclear.  In this project, we are testing the hypothesis that the MyD88 signaling axis plays a central role in the initiation of the inflammatory response after injury and that this early response is required for cardiac regeneration in zebrafish.  Our data indicate that zebrafish lacking functional MyD88 display features linked to impaired cardiac regeneration potential, including reduced immune cell recruitment to the injured tissue and reduced coronary endothelial cell proliferation at the border zone.  Additionally, transcriptomic analysis reveals that myd88 mutants exhibit a downregulation of immune-related processes after cardiac cryoinjury.  To understand the mechanisms activated downstream of MyD88, we identified effectors of the MyD88 signaling pathway and are studying their role with gain- and loss-of-function tools.  To determine the cell types in which MyD88 signaling plays a role during cardiac regeneration, we are using a transgenic approach to block MyD88 function in a cell type-specific manner.  This project will provide insights into the cellular and molecular mechanisms involved in the inflammatory response during zebrafish cardiac regeneration.  By modulating specific immune components and their response to injury, we could potentially improve the regeneration outcome in mammalian models including in humans.


Pinelopi GOUMENAKI (Bad Nauheim, Germany), Stefan GÜNTHER, Didier STAINIER
19:00 - 20:45 #29489 - 051. Epicardial influence on healing of myocardial cryoinjury in embryo.
051. Epicardial influence on healing of myocardial cryoinjury in embryo.

The embryonic epicardium undergoes epithelial-to-mesenchymal transformation, contributing cells for formation of coronary vasculature. It is also a source of paracrine cues that are essential for fetal cardiac growth, coronary vessel patterning, and regenerative heart repair.

We have modified an established model of early embryonic transmural myocardial cryoinjury (resulting in incomplete repair by smooth muscle) by performing it at the stage, where the heart is already covered by the epicardium. We hypothesized that in the presence of the epicardium, the mechanisms of reparation will be different, and a complete regeneration will occur similar to adult fish or mammalian neonatal models.

Myocardial injury was induced by cryoprobe on ventricle of avian embryos (chicken and quail) at Hamburger-Hamilton (HH) stage 21, which were then re-incubated until HH31. Myocardial regeneration was analyzed electrophysiologically by epicardial optical mapping, and morphologically by immunohistochemical markers for endothelium (QH1), myocytes (MF20), and smooth muscle actin (SMA).

Functional analysis showed changes in epicardial signal spreading in the cryoinjured area. There were profound changes in the epicardium, which was thickened, detached and wrinkled in the affected, but also in the remote area. Numerous newly formed microvessels (QH1, SMA) and myocytes (MF20-positive) were present in the subepicardial region, confirming the regenerative capability.

In conclusion, embryonic myocardial cryoinjury at trabeculated stage induces epicardial activation with enhanced vascularization and myocardial regeneration. This could be potentially used for devising novel regenerative strategies in humans that could use epicardial activation to repair myocardial infraction.


Kristýna NEFFEOVÁ (Praha, Czech Republic), Zábrodská EVA, Barbora ŠAŇKOVÁ, David SEDMERA, Hana KOLESOVÁ
19:00 - 20:45 #29604 - 053. Investigating the role of shroom3 in cardiac function and regeneration.
053. Investigating the role of shroom3 in cardiac function and regeneration.

Shroom3 is a member of a small family of proteins that regulate cell shape by coordinating cell signaling and cytoskeletal architecture. Shroom3 is comprised of a PDZ domain that facilitates localization, and two ASD domains that mediate actin and myosin dynamics. Through mouse and human studies, Shroom3 mutations have been implicated in congenital heart defects, suggesting that Shroom3 may be important in cardiac development. In the heart, shroom3 is primarily expressed in cardiomyocytes (CMs). Using CRISPR/Cas9 technology, we generated a global homozygous null mutation (shroom3-/-) in zebrafish that survive to adulthood in contrast to the embryonic lethal gene-trap mutagenesis observed in mice. Differential gene expression analysis, ascertained by from wildtype versus shroom3-/- zebrafish whole heart RNAseq, revealed significant changes in several signaling pathways known to regulate cardiac regenerative capacity including Hippo, PI3K-Akt, HIF-1, and ErbB in addition to metabolic alterations indicative of heart failure development. Although external heart morphometric analysis revealed no obvious differences, shroom3-/- maturation is delayed overall compared to wildtypes. Functional cardiovascular assessment through an intermittent-flow variable swim tube fitness test illustrated that mutants are unable to metabolize oxygen efficiently and fatigue faster than wildtype counterparts. These findings were further supported via high frequency echocardiography which showed that shroom3-/- had a statistically lower stroke volume, ejection fraction, and fractional shortening. Finally, an impaired regenerative response was observed in mutants at both 7 and 30 days post ventricular cryoinjury. These preliminary findings suggest that Shroom3 likely contributes to cardiac function and response to injury in zebrafish.


Ayana JAMAL (Milwaukee, WI, USA), Erin BENTLEY, Brooke JEFFERY, Caitlin O'MEARA, Brian LINK
19:00 - 20:45 #30316 - 055. Investigation of the role of JUNB-interactants during direct cardiac reprogramming.
055. Investigation of the role of JUNB-interactants during direct cardiac reprogramming.

The characterization of novel mechanisms safeguarding cell fate identity in differentiated cells is essential for 1) our understanding of how differentiation is maintained in healthy tissues, and 2) the development of new strategies to improve therapeutic reprogramming. In this context, we recently identified a set of four transcription factors (TFs) that collectively promote cell fate stability and restrict direct lineage reprogramming in differentiated cells. Among those factors, JUNB was found to oppose reprogramming by limiting reprogramming TF (i.e MEF2C) ability to access its target DNA and remodel chromatin. AP-1 TFs, such as JUNB, are known to mediate their function via homo and hetero-dimerization with other proteins, thus we hypothesized that yet to be identified JUNB-interacting proteins might cooperate with JUNB to regulate chromatin accessibility and promote cell fate stability. Here we describe the identification and characterization of H1F0 as a novel JUNB interactant mediating cell fate stability and opposing cardiac reprogramming.


Yu-Ling CHANG (San Diego, USA), Michaela LYNOTT, Christopher LEE
19:00 - 20:45 #30392 - 057. Regeneration of the cardiac conduction system.
057. Regeneration of the cardiac conduction system.

Arrhythmias are a hallmark of myocardial infarction (MI) and contribute to poor prognosis. Increasing evidence implicates the ventricular conduction system (VCS) in generating arrhythmias. How cardiac injury affects the global architecture and cellular composition of the VCS, and whether the VCS can endogenously regenerate or repair itself, have not been systematically described. The neonatal mouse heart can regenerate following left anterior descending coronary artery ligation at postnatal day 1 (P1). The P1 heart replaces lost cardiomyocytes, removes temporary scarring, and recovers full function. This transient regeneration capacity is lost during the first week, such that P7 hearts fail to regenerate myocardium and instead maintain a permanent fibrotic scar.

We used the knock-in reporter line Cx40-eGFP to investigate VCS changes after injury in P1 versus P7 mice. By developing advanced tissue-clearing and wholemount imaging, we determined the 3D anatomy of the postnatal VCS. We observed loss of VCS cells near the injury site in infarcted P1 and P7 hearts, resulting in discontinuity within the Purkinje network. Longitudinal analysis showed an increased density of connexin-40-positive cells surrounding the injury zone at P1. We are currently using single-cell transcriptomic analysis to identify molecular pathways associated with VCS response to injury, (re-) growth and function. Collectively, our work identifies specific morphological changes in the Purkinje network following MI across the regenerative window, with evidence of recovery at early neonatal stages. These findings provide significant insights into VCS pathophysiology during heart injury and may identify molecular targets to maintain synchronous contraction in infarcted adult hearts.


Judy SAYERS (Oxford, United Kingdom), Xin SUN, Carla DE VILLIERS, Tatjana SAUKA-SPENGLER, Paul RILEY
19:00 - 20:45 #30435 - 059. Bmp- and Fgf signaling distinctly modulate chicken and mouse proepicardium cell fate.
059. Bmp- and Fgf signaling distinctly modulate chicken and mouse proepicardium cell fate.

Bmp and Fgf signaling are widely involved in multiple aspects of embryonic development. Non-coding RNAs, such as microRNAs, have also been recently reported to play essential roles during embryonic development. We previously demonstrated that microRNAs are capable of modulating cell fate decision during proepicardial/ septum transversum (PE/ST) development in chicken, since over-expression of miR-23 blocked while miR-125, miR-146, miR-223 and miR-195 enhanced PE/ST-derived cardiomyogenesis, respectively. Importantly, regulation of these microRNAs is distinct modulated by Bmp and Fgf administration. In this study, we aim to dissect the functional role of Bmp and Fgf signaling during mouse ST/PE development, their implication regulating these microRNAs and their impact on lineage determination. Mouse PE/ST explants and epicardial/endocardial cell cultures were distinctly administrated Bmp and Fgf family members. qPCR analyses of microRNAs, cardiomyogenic, fibrogenic differentiation markers as well as key elements directly epithelial to mesenchymal transition were evaluated. Our data demonstrate that neither Bmp2/Bmp4 nor Fgf2/Fgf8 signaling is capable of inducing cardiomyogenesis, fibrogenesis or inducing EMT in mouse ST/PE explants, yet upregulation of miR-146 and miR-195 is observed. RNAseq analyses in mouse PE/ST and embryonic epicardium identified novel Bmp and Fgf family members that might be involved in such cell fate differences, such as Bmp6, Bmp7, Bmp19, Fgf5, Fgf7 and Fgf10. Administration of these Bmp and Fgf family members in mouse PE/ST lead to limited modulation of EMT induction and cardiomyogenic and/or fibrogenic differentiation. Thus, our data support the notion of species-specific differences regulating PE/ST cardiomyogenic lineage commitment.


Maria Del Mar MUÑOZ-GALLARDO, Carlos GARCIA-PADILLA, Francisco HERNANDEZ-TORRES, Estefania LOZANO-VELASCO, Angel DUEÑAS, Isabel GARCIA-VALENCIA, Lledo PALENCIA-VICENT, Amelia ARANEGA, Diego FRANCO (JAEN, Spain)
19:00 - 20:45 #30443 - 061. The Wilms’ tumor suppressor gene is expressed in adult cardiomyocytes and it regulates myocardial metabolism and response to damage.
061. The Wilms’ tumor suppressor gene is expressed in adult cardiomyocytes and it regulates myocardial metabolism and response to damage.

The Wilms tumor suppressor gene (Wt1) encodes a C2H2-type zinc-finger transcription factor that participates in transcriptional regulation, RNA metabolism and protein-protein interactions. WT1 is critically involved in the development of several organs, including kidneys and gonads, spleen, adrenals, liver, and diaphragm (Hastie, 2017). WT1 is highly expressed in the embryonic epicardium where it regulates a process of epicardial-mesenchymal transformation and the development of the epicardial-derived cells.

We have recently shown evidence of a transient Wt1 expression in about 25% of cardiomyocytes of mouse embryos. Conditional deletion of this expression in the cardiac troponin T lineage caused abnormal sinus venosus and atrium development, thin ventricular myocardium and, in some cases, interventricular septum and cardiac wall defects (Díaz del Moral et al., Front Cell Dev Biol. 2021;9:683861).

We aimed to know if Wt1 is also expressed in adult cardiomyocytes and what could be the consequences of its conditional deletion for cardiac homeostasis and/or in the response to damage induced by isoproterenol and doxorubicin treatments.  For conditional deletion of Wt1 in cardiomyocytes, we generated tamoxifen inducible Wt1 mutants by crossing aMHCMerCreMer mice with homozygous Wt1 conditional mice, where the first exon of Wt1 is flanked by loxP sites.

We have found experimental evidence of a low expression of Wt1 in postnatal murine cardiomyocytes, using reporter and lineage tracing models as well as qPCR. Our preliminary data suggest that conditional deletion of Wt1 in cardiomyocytes induces interstitial fibrosis, increased oxidative stress markers, altered metabolism and mitochondrial dysfunction in Wt1-deficient cardiomyocytes. In addition, conditional deletion of Wt1 in adult cardiomyocytes increases the damage induced by doxorubicin and isoproterenol treatments. These findings suggest a novel role of Wt1 in myocardial physiology and protection against damage.

 

 


Sandra DÍAZ DEL MORAL, Kay-Dietrich WAGNER, Nicole WAGNER, Maha BENAOUICHA, Ramón MUÑOZ-CHÁPULI, Rita CARMONA (Malaga, Spain)
19:00 - 20:45 #30462 - 063. Redifferentiated cardiomyocytes retain residual dedifferentiation signatures and are protected against ischaemic injury.
063. Redifferentiated cardiomyocytes retain residual dedifferentiation signatures and are protected against ischaemic injury.

Cardiomyocyte renewal by dedifferentiation and proliferation has fuelled the field of regenerative cardiology in recent years, while the reverse process of redifferentiation remains largely unexplored. Redifferentiation is characterised by the restoration of function that is lost during dedifferentiation and is key to the healing process following injury. Previously, we showed that ERBB2-mediated heart regeneration has these two distinct phases: dedifferentiation, followed by redifferentiation. Here, using temporal RNAseq and proteomics, we survey the landscape of the dedifferentiation-redifferentiation process in the adult mouse heart. We find well characterised dedifferentiation pathways, such as reduced oxphos, increased proliferation and increased EMT-like features, largely return to normal, though surprisignly, elements of residual dedifferentiation remain, even after contractile function is restored. These hearts appeared rejuvenated and showed robust resistance to ischaemic injury. We find that redifferentiation is driven by negative feedback signalling, notably through LATS1/2 Hippo pathway activity. Disabling LATS1/2 in dedifferentiated cardiomyocytes augments dedifferentiation in vitro and prevents redifferentiation in vivo. Taken together, our data reveal the non-trivial nature of redifferentiation, whereby elements of dedifferentiation linger in a surprisingly beneficial manner. This cycle of dedifferentiation-redifferentiation protects against future insult, in what could become a novel prophylactic treatment against ischemic heart disease for at-risk patients.


Avraham SHAKKED (Rechovot, Israel)
19:00 - 20:45 #30485 - 065. Specific features of cardiac fibroblasts underlie adult zebrafish heart regeneration.
065. Specific features of cardiac fibroblasts underlie adult zebrafish heart regeneration.

Objectives

There is increasing evidence of the importance of interstitial cells in cardiovascular development and disease. In contrast to the adult mammalian heart, the adult zebrafish heart has the ability to regenerate. However, little is known about how interstitial cell populations contributing to this. We aim to better characterise interstitial cells with an emphasis on cardiac fibroblasts, during the regeneration process.

 

Methods

Using immunohistological analysis of the zebrafish heart from multiple transgenic reporter lines, we were able to quantify the various cell types that comprise the adult zebrafish heart. In addition, we performed single-cell RNA sequencing (scRNA-seq) on uninjured and amputated hearts at various time points in order to uncover the transcriptional properties of cardiac fibroblasts, as well as other interstitial cell populations that support adult zebrafish heart regeneration. The data was cross-referenced to published scRNA-seq data from murine studies to identify features of zebrafish cardiac fibroblasts.

 

Results

We were able to establish that, as in the adult mouse heart, endothelial cells and cardiomyocytes where the main cell types. However, fibroblasts where far less abundant in the adult zebrafish heart. Indeed, they represented 1-2% versus 11% of total cells in adult zebrafish versus mouse heart, respectively. ScRNA-seq analysis revealed features of interstitial cells that may support regeneration. Notably, in zebrafish, we found very limited fibroblast to αSMA+-myofibroblast transition as compared with injured mouse heart. By pharmacologically blocking regeneration, we found that αSMA+ expression was upregulated, but not in fibroblasts expressing high levels of collagen.

 

Conclusion

Our study suggests that features of myofibroblast transition, such as increased aSMA expression, are less pronounced in zebrafish cardiac fibroblasts as compared to fibroblasts in regenerating neonatal or infarcted adult mouse heart.


Alenca HARRINGTON (Montpellier), Laura ROLLAND, Adèle FAUCHERRE, Jourdano MANCILLA ARBAROA, Girisaran GANGATHARAN, Laurent GAMBA, Dany SEVERAC, Marine PRATLONG, Thomas MOORE-MORRIS, Jopling CHRIS
19:00 - 20:45 #30498 - 067. In vitro screening of small molecules to promote lymphangiogenesis and optimal heart repair following myocardial infarction.
067. In vitro screening of small molecules to promote lymphangiogenesis and optimal heart repair following myocardial infarction.

Myocardial infarction (MI) induces death of cardiac muscle and replacement by a non-contractile fibrotic scar, which ultimately leads to heart failure. Current treatments restore blood flow and assist with cardiac workload but none are regenerative, and clinical trials using stem-cell- based approaches have been disappointing. We have previously shown that the cardiac lymphatic system is activated following MI and that further stimulation with the lymphatic endothelial specific isoform of VEGF-C, VEGF-CC156S, resolves the immune response and improves cardiac function. Whilst this study demonstrates the potential of this approach, VEGF-CC156S is sub-optimal for clinical use. 

Our aim is to target the lymphatic vasculature to enhance the intrinsic regenerative potential of the infarcted adult heart using small molecules. To achieve this, we have established a sprouting assay using human lymphatic endothelial cells (hLEC), to mimic lymphangiogenesis in vitro and to provide the basis of a phenotypic screen. We have miniaturized this cell-based assay into a 384 well-plate format and have begun to screen focused libraries of compounds (epigenetic regulators, kinase inhibitors and stem cell modulators). Using automated imaging and post-hoc analyses we have selected, clustered and quantified hit molecules, which are currently being validated and further characterised through combined medicinal chemistry and preclinical models of adult cardiac injury.


Christophe RAVAUD (Oxford, United Kingdom), Sarah SIGAL, Xin SUN, Carole BATAILLE, Angela RUSSELL, Paul RILEY
19:00 - 20:45 #30509 - 069. Tracking atrioventricular valve regeneration in vivo.
069. Tracking atrioventricular valve regeneration in vivo.

Cardiac valve disease is a prevalent cause of morbidity and mortality worldwide, in which the cardiac valves that are normally ensuring unidirectional blood flow during the cardiac cycle, become defective. In severe cases, the diseased valve is surgically removed and replaced by decellularized implants, which generally rely on the repopulation by endogenous cells from the host. However, this recellularization process which ensures the stability and growth of the implant is greatly challenged in mammals due to the limited regenerative potential of their cardiac tissues.

We recently showed that adult zebrafish are capable of regenerating their cardiac valves. With the use of the nitroreductase-metronidazole genetic ablation system, we showed that valve-cell ablation in the adult zebrafish triggers the recruitment of endothelial cells and kidney marrow-derived cells, leading to the formation of a new functional valve. However, studying this process in histological sections of fixed tissues limits the assessment of the cellular contributions and rearrangements that take place during cardiac valve regeneration in vivo.

To overcome the limitations associated with adult specimens, we are now using zebrafish larvae to study the cellular dynamics and rearrangements involved in valve cell recruitment through live imaging. We have observed that also in larval stages, genetic ablation triggers a regenerative program leading to the recruitment of new valve cells, clearly visible at five days post-ablation. We also observed that this program is accompanied by an increase in the number of immune cells in the vicinity of the valve. We are now characterizing the different cellular sources and molecular pathways contributing to the formation of the new valve through live-imaging microscopy.

Overall, with this approach we expect to clarify the in vivo mechanisms of new valve cell recruitment after valve-cell ablation and determine the key factors promoting cardiac valve regeneration.


Ana Beatriz BARBOSA (Lisbon, Portugal), Inês CRISTO, Anabela BENSIMON-BRITO
19:00 - 20:45 #30516 - 071. Active participation of ventricular trabeculae to cardiac regeneration leads to Purkinje fiber hyperplasia.
071. Active participation of ventricular trabeculae to cardiac regeneration leads to Purkinje fiber hyperplasia.

The mouse heart can regenerate during a short time window after birth, which coincides with maturation of the ventricular myocardium and compaction of trabeculae. These transient invaginations at the inner surface of the developing heart give rise to both contractile cardiomyocytes (CM) and Purkinje fibers of the ventricular conductive system (VCS). Purkinje fibers are recruited throughout embryonic and fetal development in successive waves, including a major wave of around the time of birth. This depends on maximal Nkx2-5 levels and Nkx2-5 heterozygous mice present a hypoplastic VCS. Here we investigate the contribution of trabecular myocardium to the regeneration of contractile and conductive cardiac tissues after neonatal myocardial infarction (MI). 

Neonatal MI was performed by permanent ligation of the left coronary artery on one-day-old mice. Trabecular derivatives were genetically traced using Connexin40 (Gja5) driven inducible Cre recombinase, revealing an active participation and increased proliferation of trabecular-derived cells during cardiac repair. Moreover, subendocardial myocardium re-expresses Cx40 under ischemia, consistent with a switch towards an immature phenotype. Surprisingly, we observed that regenerated hearts possess a global and permanent hyperplasia of the distal VCS despite spatially restricted damage following MI. This MI induced hyperplasia is not found in Nkx2-5+/- mice, suggesting that the underlying mechanism is Nkx2.5 dependent. We hypothesize that VCS hyperplasia under conditions of regeneration is an extension of the normal perinatal recruitment wave.

Together our data uncover cellular mechanisms involved in postnatal regeneration that resemble normal maturation of the ventricular myocardium. Trabecular-derived CM dedifferentiate toward an immature bipotent phenotype. The proliferation of these cells helps in repopulating the myocardial ischemic zone but produces an excess of PF which leads to VCS hyperplasia.


Lucie BOULGAKOFF, Lucie BOULGAKOFF (Marseille), Rachel STURNY, Robert G KELLY, Lucile MIQUEROL
19:00 - 20:45 #30543 - 073. Interspecies comparison reveals novel driver of cardiac regeneration.
073. Interspecies comparison reveals novel driver of cardiac regeneration.

Myocardial infarction causes irreversible loss of cardiomyocytes and formation of a fibrotic scar, impairing cardiac function in patients. While the mammalian heart is incapable of regenerating the lost cardiomyocytes following injury, zebrafish can completely regenerate their heart through cell cycle re-entry of pre-existing cardiomyocytes. The molecular mechanisms underlying differences in regenerative capacity between species have yet to be unraveled. We employed spatial transcriptomics on mouse and zebrafish hearts post injury and compared the transcriptomes of their respective border zones to identify overlapping and diverging expression patterns. This dataset will be a helpful tool to identify drivers of cardiac regeneration. Here we will present the dataset and its validation. Furthermore, the dataset was used to select candidate genes for functional analysis in zebrafish using a knock-out approach of which the results will be presented here.


Mara BOUWMAN (Utrecht, The Netherlands), Dennis DE BAKKER, Hessel HONKOOP, Phong NGUYEN, Jeroen BAKKERS
19:00 - 20:45 #30546 - 075. Role of sympathetic reinnervation during zebrafish cardiac regeneration.
075. Role of sympathetic reinnervation during zebrafish cardiac regeneration.

Dysregulated sympathetic innervation pattern and activity are observed in a number of cardiovascular diseases including post myocardial infarction.  Upon cardiac injury, sympathetic nerves (SNs) in the neonatal mouse heart migrate into the injury site and ablation of SNs post-injury impairs neonatal mouse heart regeneration.  In contrast, SNs fail to migrate into the injury site and accumulate at the border zone in the post-infarction adult mouse heart and it fails to regenerate.  While these data suggest that SNs play an important role in heart regeneration, when and how the SNs migrate into the injury site and their role(s) in heart regeneration remain unknown.

Here using the zebrafish heart, which regenerates post-injury, we have analyzed when and how SNs migrate into the injured tissue.  Using various reporter lines and antibodies, we find that the zebrafish SNs migrate rapidly into the injured tissue.  A subset of nerves persists within the injured tissue for several days and the innervation pattern is re-established within 30 days post-injury.  This rapid response of SNs is accompanied by the disintegration of the pre-existing neural network.  Altogether our studies indicate that after cardiac injury, cardiac SNs are very dynamic.  Hence, analyzing the function of SNs after cardiac injury will significantly advance our understanding of their role(s) during organ regeneration and cardiac disease.


Mridula BALAKRISHNAN (Bad Nauheim, Germany), Yu Hsuan Carol YANG, Didier STAINIER
19:00 - 20:45 #30549 - 077. In vivo and In vitro Investigation of the Role of Estrogen on Regeneration Potential of Cardiac Progenitor Cells.
077. In vivo and In vitro Investigation of the Role of Estrogen on Regeneration Potential of Cardiac Progenitor Cells.

Clinical and preclinical studies reported insufficient cell engraftment and delayed cardiac functional recovery problems in stem cell-based therapy in heart failure. Therefore, pretreatment of stem cells with an exogenous factor may improve regenerative efficiency. Here we demonstrated the effect of estrogen administration on the regenerative capacity of CPCs with in vitro and in vivo experiments. Explant-derived CPCs were isolated from adult mouse hearts and molecular, functional, and electrophysiological changes in CPCs upon estrogen treatment were investigated. Estrogen treatment increases the expression of proliferation, pluripotency and stem cell markers like Abcg2 and Islet-1. The migration and vasculogenic potential of CPCs was also induced by estrogen. Considering the paracrine effect of transplanted progenitor cells, we have examined the changes in the exosomal miRNA profile of CPCs. We found that estrogen regulates exosomal miRNAs known as modulators in neo-angiogenesis and cell migration. These results provided molecular evidence that estrogen pretreatment improves the regenerative capacity of CPCs inducing their selfrenewal, potency and repairment mechanisms. We have also monitored ohmic behaviour for K-channels like inward rectifier K+-current and significant hyperpolarization-shift in inward currents upon estrogen treatment suggesting the establishment of the resting membrane potential and differentiation induced by estrogen treatment. We have observed the inductive role of estrogen on cardiomyocyte differentiation of CPCs. To support in vitro data, we have used mouse heart failure model established by isoproterenol injection. Molecular and electrophysiological changes after the intracardiac allogenic transplantation of estrogen pretreated CPCs were also demonstrated. All these results indicate that ex vivo pretreatment of CPCs with estrogen may propagate the transplantation efficiency.

 


Ceylan Verda BITIRIM (Ankara, Turkey), Kardelen GENC, Yusuf OLGAR, Zeynep Busra OZER, Ebru KOCAKAYA, Dunya AYDOS
19:00 - 20:45 #30568 - 079. Reptin suppresses cardiomyocyte proliferation during zebrafish heart development and regeneration.
079. Reptin suppresses cardiomyocyte proliferation during zebrafish heart development and regeneration.

Cardiomyocyte proliferation is a critical source of new myocardium during heart development and regeneration. Novel therapies designed to replace missing or lost cardiomyocytes in the context of congenital heart disease (CHD) or myocardial infarction (MI) represents the holy grail of cardiac regenerative medicine. To boost cardiomyocyte proliferation in either setting, critical regulators must be identified. We reported that Reptin (also known as Ruvbl2), a highly conserved and widely expressed AAA+ ATPase, is a potent suppressor of cardiomyocyte proliferation during heart development and regeneration. Specifically, reptin loss-of-function mutants display ventricular hyperplasia at 3 days post-fertilization (dpf), while cardiomyocyte-specific reptin overexpression causes ventricular hypoplasia. Moreover, heat-inducible reptin overexpression during adulthood also lowers the myocardial proliferation index and results in scarring. Building on these published data, I am currently investigating whether the suppressive activity of Reptin on cardiomyocyte proliferation is conserved in mice and dissecting the downstream molecular mechanisms mediating this function. To address the former, I created cardiomyocyte-specific Reptin knock-out mice by crossing cTnt:Cre;Reptinfl/+ X Reptinfl/fl. Of 38 liveborn pups, zero mutant animals were recovered. I found that the conditional mutants die by E12.5, demonstrating a strict requirement for murine heart development. I am currently investigating cardiomyocyte proliferation in E10.5 and E11.5 conditional knock-out embryos and analyzing differentially expressed genes identified by RNA-seq from wild-type and reptin mutant cardiomyocytes in zebrafish. 


Felicia WRANITZ (Boston, USA), C. Geoffrey BURNS, Caroline E. BURNS
19:00 - 20:45 #30570 - 081. A novel medium-throughput platform for target validation of regulators of cardiac regeneration in zebrafish larvae.
081. A novel medium-throughput platform for target validation of regulators of cardiac regeneration in zebrafish larvae.

Cardiovascular diseases remain the leading cause of death worldwide, and few effective treatment options are available. Myocardial injury, such as myocardial infarction, causes irreversible damage of the heart muscle and its replacement by scar, leading to a chronic decrease in heart function. In contrast to humans, the injured zebrafish heart muscle regenerates efficiently through robust proliferation of myocardial cells. Thus, the zebrafish presents a beneficial vertebrate model for studying genetic programs behind cardiac regeneration, which may be present, albeit dormant, in the adult human heart.

To this end, we established a novel platform for studying heart regeneration after cardiomyocyte ablation in zebrafish larvae. The specific ablation of cardiomyocytes is achieved through a transgenic construct inducing the expression of nitroreductase, a bacterial enzyme, in a pool of ventricular cardiomyocytes. Subsequent treatment with antibiotics induces cell death specifically in nitroreductase-expressing cells. In combination with automated 3D heart imaging, this platform can be used for medium-throughput screening of genes and compounds with presumed effects on regeneration. Our results confirm that we induce a robust loss of the targeted cardiomyocytes, which are replaced through the proliferation of remaining cardiomyocytes within 4 days post injury. Our results further show that treatment with known anti-regenerative molecules causes a significant delay in regeneration kinetics, providing a proof of principle for this platform in identifying anti-regenerative effects of genes and drugs. Using this platform, we aim to discover therapeutic targets and drugs that will allow us to activate the dormant regenerative potential of the human heart.


Katerina APOLINOVA (Barcelona, Spain), Sylvia DYBALLA, Benedetta COPPE, Carole JUNG, Nadia MERCADER HUBER, Vincenzo DI DONATO, Javier TERRIENTE
19:00 - 20:45 #30584 - 083. Effect of estrogen on the regenerative capacity of cardiac progenitor cells in ischemic rats.
083. Effect of estrogen on the regenerative capacity of cardiac progenitor cells in ischemic rats.

Cardiac progenitor cells (CPC), whose regenerative capacity is reduced due to cardiac damage, play a role in heart failure that develops after heart damage. Regenerative capacity of multipotent CPCs is being evaluated by clinical phase-1 studies. Due to the low rates of inoculation and differentiation, treatment modalities based on CPC transplantation need to be improved. Our aim in this study is to show how estrogen replacement affects cardiac regeneration and differentiation capacity of CPCs in female ischemic animals. Recent studies  have shown that estrogen has an active role in cardiac regeneration both in functional and molecular levels. In addition, in another study by our group, we showed that the effects of estrogen on CPC differentiation have a positive effect on the regenerative capacity of CPCs. Evaluation of estrogen-related changes in the regenerative capacity of female ischemic CPCs and healthy CPCs is important in order to better understand the role of estrogen in cardiac regeneration. To investigate this, we examined changes in endothelial, fibroblastic, and cardiac differentiation capacities of CPCs derived from healthy and ischemic heart explants upon estrogen replacement. Our results showed that estrogen has a positive effect on these parameters. We also examined estrogen-dependent changes in EMT markers (Vimentin, Snail, Slug, CDH1, and CDH2) affecting the engraftment rate. Our in vitro results showed a shift towards mesenchymal characteristics upon estrogen treatment. Our results with this study are a step towards a better understanding of the effects of estrogen on the regenerative capacity of the hearts of healthy or ischemic females.


Zeynep ÖZER, Zeynep ÖZER (Ankara, Turkey), Kardelen GENÇ, Bitirim CEYLAN VERDA, Akçalı KAMIL CAN
19:00 - 20:45 #30597 - 085. ROLE OF ESTROGEN IN THE FUNCTIONAL AND ELECTROPHYSIOLOGICAL CHARACTERİSTİCS OF SCA1+ CARDIAC PROGENITOR CELLS.
085. ROLE OF ESTROGEN IN THE FUNCTIONAL AND ELECTROPHYSIOLOGICAL CHARACTERİSTİCS OF SCA1+ CARDIAC PROGENITOR CELLS.

Cardiac progenitor cells (CPCs) are a diverse group of cells found in the adult heart that can develop into distinct cardiac lineages. CPC transplantation has poor stem cell engraftment and delayed cardiac functional recovery difficulties in stem cell-based heart failure treatment. Therefore, exogenous factor therapy of CPCs may boost regeneration efficiency. Estrogen is recognized to play a role in heart regeneration on both a functional and molecular level. In vitro experiments, we wanted to see how estrogen treatment affected Sca1+ regenerative ability. The Sca1+ CPC subpopulation was isolated exclusively from mouse cardiac explants. When compared to mRNA levels in heart tissue, the expression level of pluripotency markers in sorted Sca1+ cells is significantly higher. Estrogen also promotes colony formation and proliferation. In wound healing and angiogenesis experiments, estrogen therapy promoted migration and micro vascularization in Sca1+ cells. The expression of mesenchymal markers has also increased, implying that the outcomes are functional. We also looked for ohmic activity in K-channels, which was identical to the inward rectifier K+-current, as well as a considerable hyperpolarization shift in inward currents after estrogen therapy. These data show that estrogen therapy promotes the development of the resting membrane potential and differentiation. All of these findings suggest that preconditioning CPCs with estrogen ex vivo before transplantation may improve transplantation efficiency and could be employed as a therapy option in clinical trials.


Kardelen GENÇ (Ankara University Stem Cell Institute, Turkey), Ceylan Verda BITIRIM, Zeynep Büşra ÖZER, Ebru KOCAKAYA, Kamil Can AKÇALI
19:00 - 20:45 #30718 - 087. Sept7b is required for the subcellular organization of cardiomyocytes and cardiac function in zebrafish.
087. Sept7b is required for the subcellular organization of cardiomyocytes and cardiac function in zebrafish.

 Surjya Narayan Dash,1* Suneeta Narumanchi,2* Jere Paavola,2 Sanni Perttunen,2 Hong Wang,1 Päivi Lakkisto,2,3 Ilkka Tikkanen,2,4* and  Sanna Lehtonen1*

1Department of Pathology, University of Helsinki, Helsinki, Finland; 2Unit of Cardiovascular Research, Minerva Institute for Medical Research, Biomedicum Helsinki, Helsinki, Finland; 3Clinical Chemistry and Hematology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland; and 4Abdominal Center, Nephrology, Helsinki University Hospital, Helsinki, Finland

 * equal contribution

 

Actin is the most abundant and conserved protein in eucaryotic cells and involved in various cellular functions from cell division and migration to muscle contraction. Actin and myosin generate contractile force in cardiac muscle, and mutations in these proteins may lead to heart failure. Septins are small GTPases that associate with actin filaments and play an important role in cytoskeleton organization, however, their role in cardiomyocyte organization and function is not completely understood. We found that septin 7 is expressed in both embryonic and adult zebrafish heart. Knockdown of sept7b, the zebrafish ortholog of human septin 7, reduced F-actin and α-cardiac actin expression in heart and caused disorganization of actin filaments. Electron microscopy revealed disorganization of heart myofibrils and partial detachment from Z-discs in sept7b knockdown larvae. Functional studies showed that the knockdown larvae had reduced ventricular dimensions, contractility and cardiac output. Additionally, depletion of sept7b reduced the expression of retinaldehyde dehydrogenase 2 (raldh2), which catalyzes synthesis of retinoic acid (RA), necessary for heart morphogenesis. Furthermore, we found that sept7b and RA signaling pathways converge to regulate cardiac function. Together, these results identify an essential role for sept7b in zebrafish heart development and function.

 


Suneeta NARUMANCHI (Helsinki, Finland)
19:00 - 20:45 #29375 - 091. The extracellular matrix protein agrin is essential for epicardial epithelial-to-mesenchymal transition during heart development.
091. The extracellular matrix protein agrin is essential for epicardial epithelial-to-mesenchymal transition during heart development.

During embryonic heart development, epicardial cells residing within the outer layer of the heart undergo epithelial-mesenchymal transition (EMT) and migrate into the myocardium to support and stimulate organ growth and morphogenesis. Disruption of epicardial EMT results in aberrant heart formation and embryonic lethality. Despite being an essential process during development, the regulation of epicardial EMT is poorly understood.  Here we report EMT relative to the topography of the epicardial surface of the embryonic heart at high resolution using scanning electron microscopy (SEM). We identified high- and low-EMT regions within the mesothelial layer of the epicardium and an association of these regions with key components of the extracellular matrix (ECM).  The ECM basement membrane-associated proteoglycan agrin was found to localize in the epicardium in regions actively undergoing EMT. Deletion of agrin resulted in impaired EMT and compromised development of the epicardium, accompanied by down-regulation of the epicardial EMT regulator WT1. Agrin enhanced EMT in human embryonic stem cell-derived epicardial-like cells by decreasing b-catenin and promoting pFAK localization at focal adhesions. In addition, we observed that agrin promoted the aggregation of its receptor dystroglycan within the Golgi apparatus in murine epicardial cells. Loss of agrin resulted in dispersal of dystroglycan throughout the epicardial cells in embryos, disrupting basement membrane integrity and impairing EMT. Our results provide new insights into the role of the ECM in heart development and implicate agrin as a critical regulator of epicardial EMT.  


Xin SUN (oxford, United Kingdom), Sophia MALANDRAKI-MILLER, Tahnee KENNEDY, Elad BASSAT, Konstantinos KLAOURAKIS, Jia ZHAO, Eldad TZAHOR, Paul RILEY
19:00 - 20:45 #29455 - 093. Electrical remodeling of chick atrioventricular junction during postseptated embryonic development.
093. Electrical remodeling of chick atrioventricular junction during postseptated embryonic development.

Proper functional remodeling of atrio-ventricular junction (AVJ) leaving a sole myocardial connection in the annulus fibrosus (His bundle) is crucial for the post-septated heart development. Presence of myocardial accessory pathways (APs) bypassing the annulus fibrosus can lead to the ventricular pre-excitation and could lead to severe tachyarrhythmias. The aim of our study was to functionally characterize remodeling of AVJ and diminution of myocardial atrio-ventricular connection throughout chick cardiogenesis. At the embryonic day (ED) 14, 16 and 18, we used optical mapping of retrogradely perfused heart to determine atrio-ventricular delay and localize the APs. Measurements were performed during normal conditions and after atrioventricular node conduction slowing by adenosine injection. Hematoxylin-Eosin /Alcian Blue and Picrosirius Red staining were used for morphological validation of the APs. We observed a decreased proportion of hearts with electrically active APs from 60 % at ED 14 to 50 % at ED 16 and 20 % ED 18, respectively. At ED 14, the majority of the APs were detected during normal conditions and were distributed all around the AVJ. However, the most of the APs were detected after adenosine perfusion only at 16 and 18 ED, and were mostly right-sided. This was accompanied by a trend to decrease in atrio-ventricular delay during the studied period.  Progression of AVJ fibrotization with APs disappearance was confirmed by histology. We provide a view on functional AVJ remodeling throughout chick development. This could help to elucidate spatiotemporal characteristic of proper AVJ isolation and understanding of arrhythmias associated with presence of APs.


Eva ZABRODSKA (Prague, Czech Republic), Alena KVASILOVA, Kristyna NEFFEOVA, David SEDMERA, Veronika OLEJNICKOVA
19:00 - 20:45 #29480 - 095. Stratification of cardiac malformations in the heterotaxy syndrome based on embryonic mechanisms.
095. Stratification of cardiac malformations in the heterotaxy syndrome based on embryonic mechanisms.

Crucial for heart function is its partition into a left and right pump. Left-right asymmetry of cardiac morphogenesis begins with the rightward looping of the embryonic heart tube, which positions cardiac chambers relative to each other to establish the double blood flow. In humans, disturbances in this process have been associated with severe heart defects in the heterotaxy syndrome. We have previously demonstrated that the left signal Nodal is required in cardiac precursors for orienting and shaping the embryonic heart loop (Desgrange et al., 2020). Nodal mutants later develop an heterotaxy syndrome with a spectrum of heart defects. However, the heterogeneity of the heterotaxy phenotype has remained poorly understood. Since Nodal mutants at E9.5 display four categories of abnormal heart loops, we reasoned that heterotaxy may be stratified. For example, in the clinical nomenclature, the final position of the ventricles is considered to reflect the direction of the embryonic heart loop. However, this correlation has never been tested experimentally. To tackle the challenge of monitoring a specific phenotype at two stages of development in a single embryo, we developed a multimodality imaging pipeline, combined with advanced 3D image analyses. With this unique approach, we can identify heart loop features that are predictive of specific cardiac malformations. This study provides novel insights into the emergence of complex congenital defects in the heterotaxy syndrome.


Audrey DESGRANGE (Paris), Emeline PERTHAME, Mohamed EL BEHEIRY, Jean-Baptiste MASSON, Sigolène MEILHAC
19:00 - 20:45 #29499 - 097. Essential roles of cohesin protein Shugoshin 1 in mouse development.
097. Essential roles of cohesin protein Shugoshin 1 in mouse development.

Pediatric and adult patients with Chronic Atrial and Intestinal Dysrhythmia (CAID) have a recessive point mutation (A > G) in the first coding exon of the cohesin interacting protein Shugoshin 1 (SGO1). This leads to a lysine-to-glutamic acid change at the highly conserved amino acid 23 in SGO1 (p.Lys23Glut: SGO1-K23E). To investigate the role of Sgo1 in vivo, we generated Sgo1-K23E and Sgo1-flox mouse models. Affected Sgo1-K23E/K23E homozygous mouse mutants die embryonically and show global developmental delay, consistent with an early embryonic requirement for Sgo1. Non-affected Sgo1-K23E/K23E homozygous mouse mutants survive postnatally at reduced Mendelian ratios and in a genetic background-dependent manner, but develop normally with no overt signs of functional or structural defects. Likewise, early postnatal conditional ‘knock-in’ of Sgo1-K23E similarly does not affect cardiac or intestinal function, but, surprisingly, leads to lack of neonatal hair growth. Mesodermal, second heart field, and neural crest lineage-specific Sgo1 knock-out mutants show complete embryonic lethality and lineage-specific defects shortly upon activation of Cre expression. Together, our results indicate that mouse Sgo1-K23E induced defects are genetic background dependent and that Sgo1 is likely globally required throughout development.


Piet VAN VLIET (Montreal, Canada), Marie-Pier TREMBLAY, Severine LECLERC, Gregor ANDELFINGER
19:00 - 20:45 #29521 - 099. Beyond the exome: Linking the conserved non-coding genome to cardiac development.
099. Beyond the exome: Linking the conserved non-coding genome to cardiac development.

The genetic control of heart development has been extensively studied, partly due to the prevalence of congenital heart disease (CHD). CHDs primarily arise from mutations affecting gene dosage or altering spatial/temporal expression patterns, making non-coding elements that control these aspects of gene expression particularly important. However, the annotation and functional dissection of non-coding elements is lacking when compared to the coding genome.

Recently, our lab identified 8866 human regulatory elements deeply conserved between zebrafish and humans. Dubbed accessible conserved non-coding elements (aCNEs), these open chromatin regions are enriched for enhancers and are conserved at the DNA sequence level between zebrafish and humans. Due to the deep conservation of these regions, we hypothesize that a subset of aCNEs are conserved critical regulators of cardiac development and implicated in cardiac disease.

To identify regions with a likely role in cardiac development, I used publicly available ChromHMM data from human fetal hearts to identify aCNEs with chromatin signatures representing enhancer states. I then used published ChIPseq datasets to identify regions bound by known cardiac transcription factors (TF), including NKX2.5, GATA4/5/6, and TBX5. These criteria identified 329 regions, capturing previously identified cardiac enhancers, and predicting 258 novel cardiac enhancers.

Functional examination of 17 of these novel regions showed that 88% were able to drive reporter gene expression in the developing zebrafish heart. Strikingly, two of these regions contain ultra-rare SNVs in patients with familial Left-sided cardiac legions. Currently, we are working on linking upstream TFs and downstream target genes to aCNEs to gain molecular insight into how these regions function in cardiac development and how alterations may contribute to CHDs. Altogether, this work identified putative cardiac enhancers crucial in cardiac development and disease.


Casey CARLISLE (Toronto, Canada), Mengyi SONG, Xuefei YUAN, Cherith SOMERVILLE, Rebekah JOBLING, Raymond KIM, Michael WILSON, Ian SCOTT
19:00 - 20:45 #29554 - 101. Yap and Taz are required for neural crest-derived heart development.
101. Yap and Taz are required for neural crest-derived heart development.

Congenital heart defects (CHDs) are the most common human birth defect, occurring in 1% of newborns. Cardiac neural crest cells (cNCCs) are a migratory and multipotent cell population known to aid in the development of the cardiac outflow tract (OFT), valves, and interventricular septum. We aim to establish the contribution of Yap and Taz effectors of the fundamental Hippo signaling pathway in neural crest (NC)-derived cardiac development. Conditional knockout (CKO) mice were created using a cre-lox system, by Wnt1cre and Wnt1cre2SOR, NC-specific drivers.Yap+/-;Taz-/- CKO mutants produced various OFT and remodeling defects including ventricular septal defect (VSD), tetralogy of Fallot, double outlet right ventricle, and cardiac valve aberrations, varyingly between E14.5, E16.5, and E18.5. Interestingly, Yap+/-;Taz+/- double-heterozygous hearts exhibited external morphology similar to that of controls, but sectioning revealed mild VSD, along with cardiac valve leaflet irregularities, at E16.5 and E18.5. Cell apoptosis and proliferation were unchanged in Yap+/-;Taz-/- CKO mutants. Compelling preliminary data utilizing various methodologies including RNA-sequencing, Reverse Phase Protein Array (RPPA), Assay for Transposase-Accessible Chromatin-sequencing (ATAC-seq), and transwell migration assay, indicate a potential role for Yap/Taz in regulating NC migration. Preliminary migratory studies are followed by both OFT culture and live-imaging. Together, our data indicate that Yap/Taz play a critical role in proper cNCC-derived cardiac formation.


Shannon ERHARDT (Houston, USA), Mingjie ZHANG, Xiaolei ZHAO, Tram LE, Jun WANG
19:00 - 20:45 #30122 - 103. Inhibition of TGFb pathway prevents short body size and cardiac defects in Nipbl-deficient mice, a mouse model of Cornelia de Lange syndrome.
103. Inhibition of TGFb pathway prevents short body size and cardiac defects in Nipbl-deficient mice, a mouse model of Cornelia de Lange syndrome.

Cornelia de Lange syndrome (CdLS) is a rare genetic and developmental disorder affecting about 1:10 000/1:30:000 children. Cardiac defects are observed in more than 50% of patients. CdLS children feature septal defects, and outflow tract defects including the hypoplastic aorta, stenosis, or coarctation of great arteries as well as Tetralogy of Fallot. We found that Nipbl+/- mice on a C57Bl/6 genetic background featuring a significant decrease in Nipbl mRNAs as well as a decrease in the protein in the heart also featured a severe delay in embryonic and postnatal growth. High-Resolution Episcopic Microscopy analysis of the heart at birth revealed ventricular hypertrophy, ventricular septation defects associated with a persistent truncus arteriosus. The adult hearts then feature a severe aortic phenotype with an enlargement of the intima including senescent cells and stenosis resulting in an increase in aortic flux velocity and persistent left ventricular hypertrophy. The aortic phenotype was recapitulated when Nipbl was deleted in smooth muscle cells using SmaCreERT2 x Nipblfl/fl mice. Using proteomics and RNA-sequencing of the outflow tract, we identified a dysregulated TGFb and retinoic acid pathways in embryonic Nipbl+/- hearts as well as the presence of P21+  and pERK+ senescent cells as early as in E13.5 Nipbl+/- embryonic hearts.

Human CdLS patient iPS cells differentiated in smooth muscle cells also featured a severe senescence phenotype. The phenotype was reversed by an ALK5 inhibitor

Treatment of pregnant mice with an ALK5 inhibitor from E9.5 to E13.5 also rescued the cardiac phenotype as well as the body size of mice at birth. 


Celine HACHOUD, Faten CHAABANI (Marseille), Erwan WATRIN, Gregor ANDELFINGER, Michel PUCEAT
19:00 - 20:45 #30432 - 107. Down Syndrome Congenital Heart Defects arise from mitochondria and proliferation dysfunction.
107. Down Syndrome Congenital Heart Defects arise from mitochondria and proliferation dysfunction.

Down Syndrome (DS), caused by trisomy of human chromosome 21 (Hsa21), results in a range of phenotypes, including congenital heart defects (CHD). These phenotypes arise from increased dosage of one or more of the ~230 genes on Hsa21, however the gene(s) needed in three copies to cause CHD and the underlying mechanisms remain unknown.

The lab has generated mouse model for DS (Dp1Tyb) carrying a duplication of 23Mb on Mmu16, orthologous to Hsa21. These mice show the types of CHD seen in babies, such as ventricular septal defects and atrioventricular septal defects. The overall aim is to identify causative genes and understand the pathological mechanisms underlying CHD in DS.

Using our mapping-panel, we found that a critical gene for the CHD phenotype is Dyrk1a. Our data show that DS fetal hearts have decreased oxygen consumption and proliferation, suggesting that this is an important cause of the developmental defects. We show that three copies of Dyrk1a are required for CHD in Dp1Tyb. Three copies of Dyrk1a are also required for the mitochondrial and proliferation defects in embryonic cardiomyocytes.

Our preliminary analysis of the atrioventricular cushion (AVC) shows a reduction of cellular density as well as a reduction of NFATC1 signaling pathway, involved in valve morphogenesis. Thus, decreased NFAT signaling in the AVC might lead to decreased mesenchymal proliferation and cause the reduced AVC density in Dp1Tyb mice.

Taken together, we propose that CHD in DS arise in part from increased DYRK1A activity in cardiomyocytes leading to mitochondrial and proliferation dysfunction.


Rifdat AOIDI (London, United Kingdom), Eva LANA-ELONA, Dorota GIBBINS, Claudio BUSSI, Sheona WATSON-SCALES, Darryl HAYWARD, Ok-Ryul SONG, Maximiliano GUTIERREZ, Elizabeth M.c FISHER, Victor TYBULEWICZ
19:00 - 20:45 #30441 - 109. miRNA-mediated control of epicardium-derived cell migration during cardiogenesis and ischemic heart disease.
109. miRNA-mediated control of epicardium-derived cell migration during cardiogenesis and ischemic heart disease.

The epicardium is a layer of epithelial cells covering the surface of the heart that provides important cellular contributions for embryonic heart formation. In addition, seminal works during the last decade have pointed out that the adult epicardium is re-activated after heart damage contributing to cardiac remodeling. Several fate mapping and cell lineage studies have demonstrated that coronary vascular smooth muscle cells (cVSMC) and cardiac fibroblasts develop from the EPDCs in a multi-step process involving cell proliferation, epithelial-to-mesenchymal transition (EMT) and cell migration. However, the molecular signalling cascades regulating EPDC specification and migration are poorly understand. Here we show that miR-200b is expressed from E12.5 to E15.5 during heart development. FISH in situ hybridization analysis in Wt1CreYFP, embryos, as well as, qPCR of sorted cells, showed that miR-200b is present in a cell subpopulation of EPDCs at these stages of cardiogenesis. RNA-pull-down together with RNAseq analyses reveal that all miR-200b-targets in epicardial cells are related to cell migration. In vitro experiments of gain and loss of functions, by using EPDCs and whole-organ cultures from mouse embryos, evidenced that miR-200b modifies cell motility. Additionally, analysis in ventricles from infarcted adult mice, strongly demonstrate that miR-200b is upregulated from post-myocardial infarction day 3, peaking at day 7. Collectively, our data suggest that this miRNA might be a key molecule regulating epicardial cell lineage migration during cardiac development, as well as, in EPDC activation after myocardial infarction.


Cristina SANCHEZ-FERNANDEZ, Oscar OCAÑA, Francisco HERNÁNDEZ-TORRES, Rita CARMONA, Bellon NOELIA, Vanessa NUÑEZ, Laura ALONSO-HERRANZ, Mercedes RICOTE, Estefania LOZANO-VELASCO, Carlos GARCIA-PADILLA, Diego FRANCO, Jorge N DOMINGUEZ-MACIAS, Amelia E ARANEGA (Jaen, Spain)
19:00 - 20:45 #30465 - 111. Acute Myocardial Infarction Induces Neuronal Remodeling in Murine Superior Cervical Ganglia.
111. Acute Myocardial Infarction Induces Neuronal Remodeling in Murine Superior Cervical Ganglia.

A role for cardiac sympathetic hyperinnervation in arrhythmogenesis after myocardial infarction (MI) has increasingly been recognized. In humans and mice, the heart receives cervical as well as thoracic sympathetic contributions. In mice, superior cervical ganglia (SCG) have been shown to contribute significantly to myocardial sympathetic innervation of the left ventricular anterior wall. Of interest, the SCG is situated adjacent to the carotid body (CB), a small organ involved in oxygen and metabolic sensing. We investigated the remodeling of murine SCG, as well as the proximate CB, over time after MI. Murine SCGs were isolated from control mice, as well as 24 hours, 3 days, 7 days and 6 weeks after MI. SCGs were stained for autonomic nervous system markers (Tubb3, TH and ChAT), as well as for neurotrophic factors (BDNF and NGF) and their receptors. Quantification of the staining-intensity as well as the neuronal size was performed in the entire SCG and in the proximate CB. Our results show that ChAT and TH are co-expressed in SCG neuronal cells in control ganglia. After MI, neuronal remodelling occurs, with a significant increase in size of ganglionic cells and a decreased intensity of ChAT expression. This SCG remodeling was observed as early as 24 hours after infarction, with a peak at day 7, regressing within 6 weeks post-MI to basal levels. Of note, the most robust neuronal remodeling was observed at the region adjacent to the CB. An increase of neurotrophic factors (BDNF and NGF) was observed in the CB and neuronal cells, whereas the high affinity receptors for BDNF and NGF increased in the SCG after MI. These findings were concomitant with an increase in GAP43 expression indicating axonal outgrowth in the SCG. In conclusion, overt neuronal remodeling occurs after MI in the SCG as well as in the CB, suggesting an interaction of these 2 structures that might contribute to pathological cardiac hyperinnervation


Yang GE, Lieke VAN ROON (Leiden, The Netherlands), Janine M. VAN GILS, J. Conny VAN MUNSTEREN, Anke M. SMITS, Marie-José GOUMANS, Marco DERUITER, Monique R.m. JONGBLOED
19:00 - 20:45 #30472 - 113. Llgl1 promotes timely establishment of an apical laminin sheath around the developing ventricle and maintains ventricular wall integrity.
113. Llgl1 promotes timely establishment of an apical laminin sheath around the developing ventricle and maintains ventricular wall integrity.

During cardiac development the ventricle undergoes trabeculation, where a subset of cardiomyocytes delaminate from the myocardial wall and project into the heart lumen. At the onset of trabeculation the ventricular wall exhibits apicobasal polarity, where the exterior surface is apical and trabecular cardiomyocytes emerge basally. Despite recent advances, the mechanisms by which ventricular wall integrity is maintained as cardiomyocytes delaminate are still poorly understood.

Laminins are extracellular matrix proteins that are typically basally deposited, forming the basement membrane. We identified a dynamic basal-to-apical shift in laminin deposition in the developing ventricle: prior to trabeculation laminin localises to the luminal (basal) ventricular surface, but concomitant with initiation of trabeculation basal laminin is removed and instead deposited at the apical exterior of the ventricle. Analysis of the ventricular wall in laminin beta1a zebrafish mutants reveals an increased number of cardiomyocytes undergoing aberrant apical extrusion, suggesting apical laminin helps maintain myocardial wall integrity.

Llgl1 (Lethal(2) giant larvae protein homolog 1) is a component of the Scribble complex, which is important for maintaining apicobasal polarity including regulating Crumbs, an apical transmembrane protein required for ventricular wall organisation and trabeculation. We therefore hypothesised Llgl1 plays a role in ventricular wall polarisation, organisation, or trabeculation. Analysis of llgl1 zebrafish mutants revealed a delayed basal-apical laminin shift, alongside defects in Crumbs relocalisation. These defects are accompanied by an increase in ventricular cardiomyocytes undergoing apical extrusion. Together our data indicate that llgl1 maintains ventricular wall integrity during trabeculation through promoting timely establishment of an apical laminin sheath around the ventricle.


Eric POLLITT, Emily NOËL (Sheffield, United Kingdom)
19:00 - 20:45 #30478 - 115. Defining the role of the PIDDosome in ploidy control during heart development.
115. Defining the role of the PIDDosome in ploidy control during heart development.

The mammalian adult heart is a post-mitotic organ characterized mainly by polyploid cardiomyocytes (CMs). The post-mitotic status of adult CMs poses a clear limit to heart regeneration. Recent studies have revealed that the PIDDosome, a multi-protein complex consisting of Pidd1, Raidd, and caspase-2, controls p53 activation in response to supernumerary centrosome and limits scheduled as well as accidental polyploidization events. Since PIDDosome function in heart remains elusive, the overall goal of this project is to unveil the role of the PIDDosome in CM polyploidization, as barrier to tissue regeneration.

Flow cytometry-based analyses of adult CM nuclei showed that the absence of either Pidd1, Raidd, or caspase-2 results in an increase of tetraploid CM nuclei. Moreover, cardiac-specific deletion of caspase-2 resembles the ploidy phenotype observed in Casp2 knockout mice, suggesting that the increased CM ploidy is a cell autonomous process. Comparative analyses between different postnatal stages highlighted a gradual increase in tetraploid CM nuclei in hearts from Pidd1, Raidd or Casp2 knockout mice compared to wild-type mice at postnatal day 7 narrowing down the time of PIDDosome action. Previous studies have demonstrated that Pidd1 must be anchored to mother centriole via Ankrd26 in order to activate the PIDDosome pathway. Preliminary data from Ankrd26 knockout mice revealed that similar to Pidd1, Raidd and Casp2  knockout mice, the absence of Ankrd26 leads to an increase in tetraploid CM nuclei. On the other hand, in p53 knockout mice the ploidy is not statistically increased compared to wild-type animals, suggesting that the PIDDosome regulates CM ploidy in a p53-indipendent way.


Marina LEONE (Innsbruck, Austria), Felix EICHIN, Veronika NOGELLOVA, Valentina SLADKY, Vincent BRAUN, Andrew HOLLAND, Andreas VILLUNGER
19:00 - 20:45 #30493 - 117. Prenatal and postnatal models of altered hemodynamics in study of atrial structural and electrical remodeling.
117. Prenatal and postnatal models of altered hemodynamics in study of atrial structural and electrical remodeling.

The atria serve as a contractile blood reservoir of the cardiac pump. During embryogenesis, the atria and ventricles are soon distinguished from the persisting tubular segments (atrioventricular canal, outflow tract) by disappearance of the cardiac jelly and formation of the pectinate muscles and trabeculae, respectively. Unlike the well-characterized and frequently investigated ventricular myoarchitecture, the arrangement of muscular bundles in the atria is insufficiently studied. Here we present the effects of experimental hemodynamic alterations on embryonic and adult structural and electrical properties of the atria in a variety of animal models. Chick embryonic left atrial ligation (LAL) led to inhibition of pectinate muscle development in the excluded portion of the left auricle and appearance of ectopic pacemaking activity in that region. In rat volume overload model induced by aorto-caval fistula (ACF), the atrial weights were significantly increased (similar to ventricles), with more pronounced response in the right atrium; this increase was significantly attenuated by renal denervation. In another experimental rat model of pulmonary hypertension (SUGEN-hypoxia), an extreme rise in right atrial and ventricular pressures was observed, with accompanying profound structural remodeling of both ventricles as well as the atria. Analysis of these models will provide us with clues regarding the mechanisms governing the normal development of atrial form and function and their perturbations in overload-induced remodeling and arrhythmogenesis.


David SEDMERA (Prague, Czech Republic), Frantisek VOSTAREK, Matus MIKLOVIC, Alena KVASILOVA, Frantisek PAPOUSEK, Vojtech MELENOVSKY, Frantisek KOLAR
19:00 - 20:45 #30504 - 119. Loss of the dystrophin-glycoprotein complex in cardiomyocytes is tolerated in young adult mice during injury and homeostasis.
119. Loss of the dystrophin-glycoprotein complex in cardiomyocytes is tolerated in young adult mice during injury and homeostasis.

Muscular dystrophies (MD) are a group of diseases that cause progressive weakness and loss of muscle mass. Mutations in components of the dystrophin-glycoprotein complex (DGC) cause several types of MDs, such as Duchenne. There are no effective cures available for MD and current treatments only modestly alter disease progression. This is mostly attributed to the lack of understanding of the precise causal mechanisms. While both cardiac and skeletal muscles share many functional and molecular similarities, DGC function in both tissues is not identical. Thus, in both animal models and human patients, skeletal muscle dysfunction develops much faster than abnormalities in the heart. To better understand cardiac involvement in MD, we used a mouse model of cardiomyocyte-specific knockout (KO) of Dystroglycan- a main component of the DGC. We evaluated cardiac function and molecular changes in mice during homeostasis at different ages, and during acute injury settings. We show that KO of Dystroglycan in cardiomyocytes leads to a broader loss of other DGC proteins. However, normal cardiac function in mutant mice is not affected until the age of six months, after which it is drastically impaired. Surprisingly, a panel of acute heart injuries in young mutant mice do not reveal exacerbated damage as compared to WT. Yet, when stressing young mutant mice by β-adrenergic receptor stimulation we observe a marked deterioration in heart function. Late induction of Dystroglycan KO in both young and adult mice causes a temporary impairment in cardiac function, which returns to normal within a few weeks. Taken together we show that, as in the clinic, mice lacking the DGC can function quite well (if not stressed) as young adults suggesting the existence of compensatory mechanisms to cope with the loss of DGC in cardiomyocytes. Understanding these compensatory mechanisms might reveal novel targets for protection in both cardiac and skeletal muscle tissues affected in MDs.


Alex GENZELINAKH (Rehovot, Israel), Dina AWEIDA, Sharon HAVUSHA-LAUFER, Nachum NATHAN, Shenhav COHEN, Eldad TZAHOR
19:00 - 20:45 #30523 - 121. Dissecting the regulatory program controlling trapezius muscle development at the head trunk interface.
121. Dissecting the regulatory program controlling trapezius muscle development at the head trunk interface.

Branchiomeric head and neck muscles have a shared origin with second heart field cardiac progenitor cells in cardiopharyngeal mesoderm (CPM). Clonal analysis has identified a series of common skeletal and cardiac muscle lineages along the anterior-posterior axis of the developing pharynx. Progenitor cells giving rise to jaw opening muscles and the right ventricle, or facial expression muscles and the cardiac outflow tract, are found in anterior CPM. Posterior CPM gives rise to neck muscles, including the trapezius, and both arterial and venous pole myocardium. The retinoic acid (RA) signalling pathway is required for normal deployment of cardiac progenitor cells in posterior CPM and blocking RA signaling during a defined early time window leads to conotruncal and atrial septation defects. Here we show using pharmacological approaches that blocking RA signaling also results in a strikingly selective loss of the trapezius muscle, without affecting other branchiomeric or somite-derived trunk and limb muscles. This reveals that, although regulators such as the transcription factor and 22q11.2 deletion syndrome gene TBX1 are broadly required for branchiomeric myogenesis, regulatory subprograms differ in posterior and anterior CPM. RA is required to specify the trapezius anlagen upstream of expression of myogenic determination factors of the MyoD family. Lineage specific activation of a dominant negative RA receptor suggests that, unexpectedly, this effect is not mediated by direct RA signaling to trapezius progenitor cells in CPM, but indirectly through the somitic lineage. These findings suggest a model in which trapezius development is dependent on a signaling cascade between cell types across the head/trunk interface. Our results provide insights into the mechanisms driving myogenic development within CPM and contribute to a better understanding of muscle pathology and evolution.

 


Camille E. DUMAS (Marseille), Christopher DE BONO, Claudio CORTES, Estelle JULLIAN, Fabienne LESCROART, Stephane ZAFFRAN, Noritaka ADACHI, Robert G. KELLY
19:00 - 20:45 #30534 - 123. Can Wt1 control cardiomyocyte fate?
123. Can Wt1 control cardiomyocyte fate?

During development, the heart growths through addition of progenitor cells to the poles of the primordial heart tube.  In the zebrafish, wilms tumor 1 transcription factor a (wt1a) and b (wt1b) are expressed in the pericardium, at the venous pole of the heart. From this pericardial layer, the proepicardium emerges. Proepicardial cells are subsequently transferred to the myocardial surface and form the epicardium, covering the myocardium. We found that while wt1a/b expression is maintained in proepicaridal cells, it is downregulated in those pericardial cells contributing to cardiomyocytes from the developing heart. Sustained wt1 expression impaired cardiomyocyte maturation by reducing chromatin accessibility of specific genomic loci. Strikingly, a subset of wt1a/b-expressing cardiomyocytes changed their cell adhesion properties, delaminated from the myocardium and upregulated epicardial gene expression. Thus, wt1 acts as a break for cardiomyocyte differentiation and ectopic wt1 expression in cardiomyocytes can lead to their transdifferentiation into epicardium.


Ines MARQUES (Bern, Switzerland), Alexander ERNST, Prateek ARORA, Andrej VIANIN, Andrés SANZ-MOREJÓN, Uta NAUMANN, Adolfo ODRIOZOLA, Xavier LANGA, Laura ANDRÉS-DELGADO, Benoît ZUBER, Marco OSTERWALDER, Filipa SIMÕES, Christoph ENGLERT, Nadia MERCADER
19:00 - 20:45 #30540 - 125. TAB2 and TAK1 localize to primary cilia and play significant roles in cardiac development and disease.
125. TAB2 and TAK1 localize to primary cilia and play significant roles in cardiac development and disease.

Haploinsufficiency of TAB2 causes Congenital Heart Disease (CHD). TAB2 mediates non-canonical TGF-β signalling via interactions with TAK1 and TRAF6. Mutations of TAK1 cause Frontometaphyseal Dysplasia (FMD) and cardiospondylcarpofacial syndrome (CSCFS), rare multisystem syndromes, where CHD may appear in the clinical spectrum.

Whole exome sequencing data from a cohort of 1,471 syndromic CHD patients (sCHD), 2,405 patients with isolated CHD (iCHD) and 45,082 controls show increased burden of rare TAB2 and TAK1 variants in sCHD, but not in iCHD. We hypothesized that TAB2 and TAK1 are functionally important in heart development and addressed this experimentally in vitro and in vivo. Using in vitro models and embryonic tissues, we show that TAB2 and TAK1 are expressed in embryonic hearts and localise to primary cilia. Furthermore, mutation of Tak1 and Tab2 inhibits cardiomyogenesis of P19CL6 stem cells. In vivo zebrafish models show that mutation of tab2 and tak1 cause cardiac developmental defects. In addition, deep phenotyping of tak1 mutant zebrafish showed that several phenotypic features of FMD and CSCFS appear in the zebrafish model during early stages of development. This suggests a potential novel in vivo model for investigating FMD and CSCFS. In summary, our data suggests an important role for TAB2 and TAK1 in cardiac development and CHD, coordinated via the primary cilium.


Daniel Alexander BAIRD (Copenhagen, Denmark), Canan DOGANLI, Yeasmeen ALI, Enrique AUDAIN, Line JESSEN, Pauline TRUELSEN, Jeppe Theisen PEDERSEN, Malou Maria NIELSEN, Kateřina APOLÍNOVÁ, Vincenzo DI DONATO, Javier TERRIENTE, Marc-Phillip HITZ, Søren Tvorup CHRISTENSEN, Lars Allan LARSEN
19:00 - 20:45 #30542 - 127. Novel regulatory mechanism of hemogenic endocardium during cardiovascular development.
127. Novel regulatory mechanism of hemogenic endocardium during cardiovascular development.

     A subset of endocardial cells is hemogenic during early embryogenesis. Hemogenic endocardial cells are enriched in the cushion region and undergo endocardial-hematopoietic transition via Nkx2-5-dependent manner, suggesting that Drosophila tinman-dependent cardio-hematopoietic program is conserved in mammals. These hemogenic endocardial cells give rise to a subset of cardiac tissue macrophages that are essential for the cushion remodeling.

     To examine the regulatory network of Nkx2-5-dependent endocardial hematopoiesis, we analyzed scRNA-seq data from wildtype and Nkx2-5-null embryonic hearts. As expected, Nkx2-5-null hearts were devoid of clusters for hemogenic endocardium (e.g. Runx1+, Cd41+) and cushion endocardium (e.g. Twist1+, Msx1+). Interestingly, scRNA-seq analysis further revealed that genes related to Notch signaling pathway are significantly downregulated in Nkx2-5-null endocardium. To examine whether Notch signaling induces Nkx2-5-dependent endocardial hematopoiesis, we performed genetics experiments. Nkx2-5-null endocardium showed significant downregulation of Notch1+ endocardial cells, and NICD overexpression drastically activated hematopoiesis in the endocardium. Interestingly, impaired hematopoiesis and cushion defect in the Nkx2-5-null heart were both rescued by overexpression of NICD. Further gene regulatory network analysis identified that Dhrs3, which is known to catalyze the reduction of all-trans-retinaldehyde to all-trans-retinol and attenuates retinoic acid signaling, was a signature gene of the hemogenic endocardial cells downstream of Nkx2-5. Forced activation of NICD increased the number of not only Dhrs3+ hemogenic endocardial cells but also Dhrs3+ macrophages, suggesting a possible involvement of Dhrs3+ macrophages in cushion remodeling.

     This study demonstrated that the Nkx2-5/Notch signaling axis plays a pivotal role in endocardial-hematopoietic transition during early embryogenesis, thereby facilitating local tissue remodeling.


Norika LIU (Tokyo, Japan), Naofumi KAWAHIRA, Yasuhiro NAKASHIMA, Haruko NAKANO, Akiyasu IWASE, Yasunobu UCHIJIMA, Sean WU, Susumu MINAMISAWA, Hiroki KURIHARA, Atsushi NAKANO
19:00 - 20:45 #30551 - 129. Chromatin regulatory genes identified as modifiers of 22q11.2 deletion syndrome are also identified in sporadic congenital heart disease.
129. Chromatin regulatory genes identified as modifiers of 22q11.2 deletion syndrome are also identified in sporadic congenital heart disease.

Most cases of congenital heart disease (CHD) occur sporadically and are, in part, attributable to rare de novo variants in chromatin regulatory genes, shown to account for ~3% of sporadic CHD by the Pediatric Cardiac Genomics Consortium (PCGC). This increases to 20% in cases of CHD that co-occur with neurodevelopmental disabilities (NDDs) and/or extra-cardiac anomalies. We analyzed the exome from whole genome sequence in 456 cases with conotruncal heart defects (CTDs) versus 537 controls with no heart anomalies, all with 22q11.2 deletion syndrome (22q11.2DS), to identify genetic modifiers. 22q11.2DS is ideal for this study as it has variable phenotypic expressivity of CHD and NDDs, along with other congenital abnormalities. Among the most interesting genes found was a subset of chromatin regulators that account for 9.21% of the CTD patients. We cross-referenced our list of 40 chromatin regulators with 90 identified in sporadic CHD by the PCGC, finding significant overlap (n=12; p<3.16e-4) between the two groups. This is particularly compelling when considering that the identification of these variants was performed using different patient cohorts and statistical methods. Further, the chromatin regulatory genes in our cohort are overwhelmingly associated with genetic syndromes with increased incidence of CHD, NDDs, and extra-cardiac anomalies, making it of interest to determine whether they may also serve as modifiers for other syndromic anomalies. Thus, the data from our 22q11.2DS cohort reinforces the importance of chromatin regulatory genes as risk factors for sporadic CHD with syndromic features and suggests that modifiers of 22q11.2DS have inter-syndromic significance.


Daniella MILLER (NYC, USA), Yingjie ZHAO, International 22Q11.2Ds Brain Behavior CONSORTIUM, Bernice E. MORROW
19:00 - 20:45 #30555 - 131. Molecular regulation of great vessel formation and remodelling.
131. Molecular regulation of great vessel formation and remodelling.

The cardiovascular system, comprised of the heart and the blood vessels, distributes oxygen and nutrients to the body. Accordingly, defective cardiovascular development can cause congenital heart disease (CHD). We have previously elucidated the mechanisms by which the transmembrane protein NRP1 promotes the remodelling of the foetal cardiac outflow tract (OFT) into the base of the pulmonary artery and aorta. NRP1 also enables the remodelling of the primitive pharyngeal arch arteries (PAA) into the great vessels that distribute blood from the heart into the lungs and body. We are seeking to (a) define the temporal window in which NRP1 enables PAA morphogenesis, (b) elucidate which specific cell types require NRP1 for PAA morphogenesis and (c) identify the NRP1 ligand(s) that mediate PAA morphogenesis. Here we show that NRP1 acts in both endothelial cells from the second heart field to promote PAA formation and in the cardiac neural crest lineage to promote vascular smooth muscle cell differentiation on the PAA, thereby promoting two consecutive stages of PAA morphogenesis. We have also begun to compare the requirement of different NRP1 ligands for these consecutive stages of great vessel formation. Defining the molecular and cellular mechanisms by which NRP1 enables PAA morphogenesis will increase our understanding of the normal and abnormal development of the cardiovasculature. In the long run, this knowledge may help improve the early diagnosis and treatment of CHD that involve the aortic arch arteries.


Elena IOANNOU (London, United Kingdom), Ariadna GADOR NAVARRO-ARAGALL, Alice PLEIN, Laura DENTI, Peter SCAMBLER, Christiana RUHRBERG
19:00 - 20:45 #30562 - 133. WT1 expression in endothelial cells is required for physiological blood vessel formation.
133. WT1 expression in endothelial cells is required for physiological blood vessel formation.

Wt1 encodes a zinc finger protein whose best-known function is its role as a transcription factor. Although for many years WT1 has been considered as one of the main hallmarks of the embryonic epicardium, new pieces of evidence have demonstrated the expression of WT1 in coronary endothelial cells during the vascularization of the embryonic heart and following myocardial infarction. Here we have used Wt1 reporter and lineage tracing mouse models to fully characterize the expression of WT1 in the endothelium through all developmental stages of coronary formation. To investigate its role during coronary vessel formation we have generated an inducible endothelial-specific Wt1 KO mouse model (Wt1-KOΔEC). The analysis of these mutant mice has demonstrated that WT1 expression in coronary endothelial cells during coronary plexus formation is required for the correct development of the coronaries and the compact myocardium. The transcriptomic analysis of coronary endothelial cells from Wt1-KOΔEC mice demonstrates that WT1 works as a master regulator of crucial processes of endothelial cells. We next used the postnatal retina, to investigate whether Wt1 functions in endothelial cells are conserved among other vascular beds. We observed profound alterations in the retinal endothelial network of postnatal Wt1-KOΔEC mice, revealing a reduced endothelial migratory phenotype and severely reduced vascular complexity. Our results shed new light on the functions of WT1 in the endothelium and suggest interesting roles for WT1 during physiological blood vessel formation. We speculate that pharmacologic interventions of WT1 or WT1 downstream targets may be useful for stimulating coronary vascular growth.


Marina RAMIRO-PARETA (BARCELONA, Spain), Rosa PORTELLA-FORTUNY, Claudia MÜLLER-SÁNCHEZ, Manuel REINA, Francesc X. SORIANO, Ofelia M. MARTÍNEZ-ESTRADA
19:00 - 20:45 #30566 - 135. Mechanics of heart tube formation in Drosophila.
135. Mechanics of heart tube formation in Drosophila.

Heart development begins with the formation of a tube, as contralateral cardiac progenitors migrate and merge medially. Defective cell movement results in improper heart tube formation and congenital heart defects. However, the mechanisms of cell migration in early cardiogenesis remain unclear. In Drosophila, the embryonic heart is a linear structure composed of 52 pairs of bilateral cardiac precursors (cardioblasts, CB), that migrate dorsally and medially to form the heart tube. Using quantitative time-lapse microscopy, we found that CBs took periodic forward and backward steps as they migrated. The forward steps were greater in both amplitude and duration, resulting in net forward movement. Force generation by the molecular motor non-muscle myosin II is critical for cell movements. We found that myosin displayed an alternating pattern of localization between the leading and trailing ends of migrating CBs, forming oscillatory waves that traversed the cells. Live imaging revealed that the alternating myosin polarity was associated with alternating contractions of the leading and trailing edges. Mathematical modeling predicted that forward migration requires the presence of a boundary at the trailing edge of CBs that restricts backward movement. Consistent with this, we found a supracellular actin cable at the trailing edge of the CBs. Releasing the tension sustained by the supracellular actin cable increased the amplitude of the backward steps of CBs, thus reducing the speed of migration. Our results indicate that periodic cell shape changes coupled with a supracellular actin cable result in asymmetrical forces that guide cardioblast migration.


Negar BALAGHI (Toronto, Canada), Gonca ERDEMCI-TANDOGAN, Rodrigo FERNANDEZ-GONZALEZ
19:00 - 20:45 #30574 - 137. Large scale CRISPR-based screen for cell-type specific regulators of heart development.
137. Large scale CRISPR-based screen for cell-type specific regulators of heart development.

Congenital heart disease (CHD) remains the largest cause of birth defects worldwide, affecting about 1% of live births. Human genetics studies have identified genes potentially implicated in CHD, but the function of many of these genes in heart development and the specific cell types affected by their perturbation remain unknown. Furthermore, recent bulk and single-cell omics approaches have extensively characterized the molecular landscape of heart development, also identifying candidates expressed at various developmental stages. However, the functional relevance of many of these candidates remains largely unknown. The enormous search space for functionally important players in heart development and disease collectively created by these studies is challenging to approach with individual functional studies such as mouse knockout models. Here, we take advantage of the well-established system of hPSC differentiation to cardiac cell types, coupled with the recently established CRISPR-based perturbation method, CRISPR activation/inhibition, to perform a screen of 500 candidates potentially involved in the specification and differentiation of cardiac cell types. We will first establish our screen in atrial and ventricular cardiomyocytes, with the goal of expanding our approach to other cardiac cell types in the future. Taking advantage of the faithful recapitulation of key developmental stages in the hPSC system, we expect to uncover genes essential for the formation of differentiated cell types as well as their intermediary progenitor stages. Thus, by moving from expression to function at large scale, we hope to contribute to an enhanced understanding of cell-type specific molecular mechanisms of heart development and disease.


Tasneem EBRAHIM (New York, USA), Xiaoting ZHOU, David GONZALEZ, Felix RICHTER, Nan YANG, Nicole DUBOIS
19:00 - 20:45 #30581 - 139. The role of Sox9 in atrioventricular septation.
139. The role of Sox9 in atrioventricular septation.

The Second Heart Field (SHF) is a population of cardiac progenitor cells that contributes to the developing heart after the initial formation of the primary heart tube. At the arterial pole, the SHF contributes to the outflow tract (OFT), the right ventricle (RV), and the ventricular septum (VS). At the venous pole, the SHF contributes to some atrial myocardial structures and to the dorsal mesenchymal protrusion (DMP). The importance of the SHF for normal heart development is demonstrated in studies in which perturbation of gene expression in the SHF has shown to result congenital heart malformations, including atrioventricular septal defects (AVSDs).

The AV mesenchymal complex (AVMC) consists of four separate entities; the two major endocardially-derived AV cushions, the mesenchymal cap situated on the leading edge of the developing primary atrial septum, and the DMP. Proper development of the AVMC is essential for the development of the septal structures at the AV junction

It is well-known that the transcription factor Sox9 plays an important role in the development of the AV cushions and valve formation. Preliminary studies and analysis of data in published papers by others, led us to believe that Sox9 could play a role in SHF-dependent development of the AV valvuloseptal complex. To test this hypothesis, we generated a SHF-specific Sox9 knockout mouse and studied how depleting Sox9 from this cell population affected the formation of the components of the AVMC and the impact of this experimental approach on AV septation.  The results of the study show that SHF-derived Sox9 plays a critical role in atrial as well as ventricular septation.


Raymond DEEPE, Renelyn WOLTERS (GOOSE CREEK, USA), Jenna DRUMMOND, Andy WESSELS
19:00 - 20:45 #30587 - 141. Pitx2 regulates the building plan of the heart.
141. Pitx2 regulates the building plan of the heart.

Pitx2 is recognized as a late effector of laterality regulating cardiac asymmetric remodeling, however the role of the gene in the context of cardiac differentiation and morphogenesis is still mostly unexplored. Pitx2 null mice present severe congenital heart diseases (CHD) including atrial and ventricular septal defects, and double outlet right ventricle. We planned to assess the dynamics of Pitx2 contribution to heart development and the link between absence of Pitx2 and CHD onset. To this aim, we combined morphogenetic and molecular analysis of Pitx2 mutant embryonic hearts with pseudolineage tracing of Pitx2null cells. Here, we show that Pitx2 delineates a subpopulation of left cells which undergo directional expansion during cardiac development. This process drives concomitant ingression and progressive reallocation of the adjacent Pitx2–ve population inside the heart. Directional expansion of the Pitx2 cell population progressively coordinates all aspects of external and internal remodeling through cardiac development, thereby directing ventricular sliding, outflow tract spiraling and septation. Our results provide a novel framework to understand the role of laterality in the developing heart and the rationale for congenital heart disease onset in laterality mutants.


Michela MENEGOLLO, Marta MURGIA, Marina CAMPIONE (Padova, Italy)
19:00 - 20:45 #30593 - 143. Sinoatrial node dysfunction driven by a pathogenic TBX5-p.G125R missense mutation.
143. Sinoatrial node dysfunction driven by a pathogenic TBX5-p.G125R missense mutation.

A pathogenic variant p.G125R (c.373G>A) in the 5th exon of TBX5 (T-box transcription factor 5) induces sick sinus syndrome and sinus bradycardia, indicating dysfunction of the leading pacemaker of the heart, the sinoatrial node (SAN). While homozygous mice die in utero, heterozygous mice with this variant (Tbx5G125R/+) are morphologically unaffected but display longer and more variable RR intervals, atrial extra systoles, and susceptibility to atrial fibrillation, recapitulating the electrophysiological abnormalities seen in human carriers (van Ouwerkerk, 2022). Tbx5G125R/+ mice present with a prolonged sinus node recovery time and sinus bradycardia, indicating SAN dysfunction. Tbx5-p.G125R clearly influences the atrial phenotype, however, its effects on the SAN are unclear. Here, we have found 3221 differentially expressed genes (Padj <0.05) in the Tbx5G125R/+ SAN; 1432 were downregulated and 1789 were upregulated. Several SAN-specific markers, key SAN transcription factors, and bone morphogenetic protein signaling components involved in SAN identity and development are differentially expressed in the Tbx5G125R/+ SAN. Nevertheless, 3D reconstruction analyses of prenatal hearts indicated that SAN morphology is unaffected. However, fibroblasts are depleted in Tbx5G125R/+ atria, and fibroblast- and extracellular matrix composition-associated gene expression is further decreased in Tbx5G125R/+ SAN tissues, suggesting structural and compositional abnormalities. Altered expression of calcium handling genes and ion channels involved in normal SAN function further indicate that the Tbx5G125R/+ SAN is functionally affected. We propose that Tbx5-p.G125R has a broad effect on the regulation of key drivers of SAN development, structure, cellular composition, and electrophysiological function.


Lieve VAN DER MAAREL (Amsterdam, The Netherlands), Fernanda BOSADA, Laura STOOP, Bjarke JENSEN, Vincent CHRISTOFFELS
19:00 - 20:45 #30602 - 145. A novel pathogenic GATA6 variant identified in a family with persistent truncus arteriosus, childhood-onset diabetes mellitus and spontaneous intestinal perforation.
145. A novel pathogenic GATA6 variant identified in a family with persistent truncus arteriosus, childhood-onset diabetes mellitus and spontaneous intestinal perforation.

GATA6 is a member of the GATA family of transcription factors that play an important role during heart development. GATA6 variants are primarily associated with congenital heart disease (CHD) and pancreatic agenesis. However, the phenotypic spectrum has expanded, including additional congenital malformations affecting the biliary system, congenital diaphragmatic hernia and developmental delay. Here, we report a family where the father and child duo had persistent truncus arteriosus (PTA) and the proband had atrial septal defect (ASD). Notably, the father had childhood-onset diabetes mellitus and the affected child was diagnosed with spontaneous intestinal perforation (SIP) on day 2 after birth, which has not been previously associated with GATA6 mutation. Using exome sequencing, we identified a novel, heterozygous missense variant in GATA6 (c.1403G>A; p.Cys468Tyr) in affected members. The variant in our family is absent from the gnomAD and ExAC databases and causes a missense change predicted to be damaging by all in silico tools. Sanger sequencing confirmed the exome sequencing results and segregation of this variant. GATA6 p.Cys468Tyr mutation affects the highly conserved zinc-finger domain of GATA6 and impedes its ability to bind DNA. In vitro functional analysis showed that p.Cys468Tyr mutant protein exhibited significantly decreased transcriptional activity. Furthermore, p.Cys468Tyr mutant demonstrated an impaired localization pattern and protein aggregation. Our findings expanded the variant and phenotypic spectrum for GATA6. Our study highlights that GATA6 variants can cause not only cardiac and pancreatic malformations but also gastrointestinal abnormalities, including SIP. Further investigation is needed to define the mechanisms underlying the full phenotypic spectrum associated with pathogenic variation in GATA6.


Jun YASUHARA (Columbus, USA), Sathiyanarayanan MANIVANNAN, Uddalak MAJUMDAR, David GORDON, Katherine MYERS, Gloria ZENDER, Kim MCBRIDE, Peter WHITE, Vidu GARG
19:00 - 20:45 #30605 - 147. Coordinated regulation of the actomyosin cytoskeleton, cell shape, and chamber morphology in the zebrafish heart.
147. Coordinated regulation of the actomyosin cytoskeleton, cell shape, and chamber morphology in the zebrafish heart.

During cardiac morphogenesis, the linear heart tube expands to create cardiac chambers, each with a convex outer curvature (OC) and a concave inner curvature (IC). This stereotypical chamber shape facilitates the function of the embryonic heart, and errors in chamber morphogenesis are frequently associated with congenital heart disease. Using zebrafish as a model organism, our studies show that regional changes in cardiomyocyte morphologies underlie curvature formation: as the ventricle emerges, OC cells enlarge and elongate along their lateral axis, whereas IC cells remain relatively small and round while extending along their apicobasal axis. These divergent growth behaviors result in more squamous cells in the OC and more cuboidal cells in the IC. Coupled with these changes in cell morphology, we find distinct organization of the actomyosin cytoskeleton in each curvature: whereas F-actin and phospho-Myosin are distributed primarily along the basal and lateral cortices of developing OC cells, IC cells exhibit relative enrichment of this network along their apical cortex. These data suggest a link between actomyosin dynamics and patterns of cell shape change; indeed, we find that modulation of actin polymerization or myosin activity disrupts curvature-specific cell shapes. We therefore hypothesize that ventricular curvature formation involves the coordination of curvature-specific reorganization of the actomyosin network with acquisition of squamous and cuboidal cell morphologies in the OC and IC. Intriguingly, we find that the T-box transcription factor Tbx5a is required for both of these processes. Ongoing studies aim to connect the effector genes downstream of Tbx5a with the regulation of actomyosin organization and the attainment of OC and IC cell morphologies. Altogether, our work provides a new model for the control of chamber morphogenesis by localized cytoskeletal dynamics that create patterns of cardiomyocyte cell shapes.


Dena LEERBERG, Gabriel AVILLION, Deborah YELON (La Jolla, USA)
19:00 - 20:45 #30617 - 149. TBX1-VEGFR3 interaction in cardiac development.
149. TBX1-VEGFR3 interaction in cardiac development.

Background/Introduction

Tbx1 is the major developmental gene involved in 22q11.2 deletion syndrome (22q11.2DS), the most common known genetic cause of congenital heart disease (CHD). The clinical phenotype is well recapitulated in Tbx1 mouse mutants, including cardiovascular abnormalities affecting the aortic arch, ventricular septum and cardiac outflow tract (OFT).

Intriguingly, clinical studies have linked VEGFR3, a gene that is regulated by TBX1, to CHD; rare variants in VEGFR3 predispose to OFT malformations, including Tetralogy of Fallot (TOF), the most common CHD found in 22q11.2DS patients. We have shown that in mice, TBX1 regulates Vegfr3 in endothelial cells. Furthermore, these genes interact strongly during cardiac lymphangiogenesis ensuring the correct number, morphology and growth of cardiac lymphatic vessels. Thus, Tbx1-Vegfr3 compound mutants are, potentially, a good model for genetic studies to reveal the site (tissue) and timing of the critical interaction in OFT development. 

Purpose

We hypothesize that TBX1 and VEGFR3 play essential roles in endothelial-mesenchymal transition (EMT), a key process in OFT formation. To test this, we performed a histological analysis of hearts of conditional Vegfr3 homozygous (Tbx1Cre/+; Vegfr3flox/flox) embryos at E18.5.

Results and Conclusions

The results shown that Tbx1-driven inactivation of Vegfr3 causes a variety of intracardiac anomalies, including morphogenesis defects of the OFT. Our preliminary analysis provides proof of a genetic interaction between Tbx1 and Vegfr3 in cardiac morphogenesis and confirms the importance of Vegfr3 for cardiac development in the mouse. Future studies, will use time- and tissue-specific gene mutation in order to refine our genetic study.


Stefania MARTUCCIELLO (Fisciano (Sa), Italy), Sara CIOFFI, Mariangela CAVALLARO, Bilio MARCHESA, Antonio BALDINI, Elizabeth ILLINGWORTH
19:00 - 20:45 #30620 - 151. I- and A-band titin-truncating variants affect heart function and cardiac disease progression differently in heterozygous zebrafish mutants.
151. I- and A-band titin-truncating variants affect heart function and cardiac disease progression differently in heterozygous zebrafish mutants.

Allelic heterogeneity in titin truncating variants (TTNtv) causes variable phenotypes and is associated with dilated cardiomyopathy as well as atrial fibrillation. However, limited knowledge is available regarding the cellular and molecular mechanisms behind the various disease manifestations. In order to improve our understanding of the cardiac pathology in patients carrying TTNtv, we investigated two different zebrafish models carrying an I- and A-band TTNtv, respectively. Using electrocardiography and echocardiography, we characterized the phenotype of adult heterozygous carriers of the TTNtv. The I-band truncated mutant was primarily affected in ventricular structure and function, while the A-band truncated mutant had electrical disturbances in the atrium already from early adulthood. Therefore, we set out to investigate whether there were any signs of cardiac dysfunction at the larval stage. Using in vivo calcium imaging, we found atrial calcium mishandling in three-week-old TTNtv-A heterozygous mutants while TTNtv-I heterozygous mutants displayed no defects. Additional findings were present as early as 5 days-post-fertilization, including increased sarcomere length in TTNtv-A heterozygous mutants and decreased ERK signaling only in TTNtv-I heterozygous mutants. These data show that TTNtv have molecular and physiological implications that affect the cardiac chambers differently depending on the position of the mutation.


Marie BØLLING KONGSTED (Copenhagen, Denmark), Rashmi PRIYA, Didier YR STAINIER, Pia RENGTVED LUNDEGAARD
19:00 - 20:45 #30623 - 153. Nr2f1a maintains nkx2.5 expression to repress sinoatrial node identity within venous atrial cardiomyocytes.
153. Nr2f1a maintains nkx2.5 expression to repress sinoatrial node identity within venous atrial cardiomyocytes.

Nr2f transcription factors are conserved regulators of vertebrate atrial development, yet the mechanisms by which these proteins function within atrial cardiomyocytes (ACs) are still not completely understood. To investigate the consequences of Nr2f loss within the atrium, we performed transcriptomic analysis on isolated ACs from wild-type (WT) and nr2f1a mutant zebrafish at 48 hours post-fertilization (hpf). Interestingly, our results revealed altered expression of core genes, including increased tbx3a and decreased nkx2.5, which respectively promote and repress sinoatrial node (SAN) differentiation. Subsequent examination of nr2f1a mutant atria from 48 to 96 hpf showed a progressive expansion of SAN markers into the atrium and concurrent regression of Nkx2.5 from the venous pole, with analysis of heart rate and action potential duration suggesting that nr2f1a mutant ACs functionally adopt pacemaker identity. Genetic epistasis using a heat-shock inducible Nkx2.5-EGFP transgene revealed that overexpression of Nkx2.5 can repress the SAN expansion in nr2f1a mutant hearts. Furthermore, profiling of chromatin accessibility in isolated ACs identified a putative nkx2.5 enhancer harboring an Nr2f binding site, which we found is expressed in the ACs adjacent to the SAN in transgenic embryos. Expression of the putative enhancer is lost in nr2f1a mutants, further supporting that Nr2f1a may directly maintain a border of atrial nkx2.5 expression. Altogether, our results reveal a novel requirement for Nr2f transcription factors in maintaining AC identity at the expense of SAN identity by maintaining Nkx2.5 expression in vertebrate hearts, which may provide insight into the etiology of arrhythmias and CHDs associated with human NR2F2 mutations.


Kendall MARTIN (Cincinnati, USA), Padmapriyadarshini RAVISANKAR, Manu BEERENS, Calum MACRAE, Joshua WAXMAN
19:00 - 20:45 #30633 - 155. Investigating a Novel Genetic Cause of Cardiomyopathy.
155. Investigating a Novel Genetic Cause of Cardiomyopathy.

Cardiomyopathy is a unified term for heterogeneous, progressive diseases of the myocardium that result in an enlarged, thickened or stiffened pathophysiology which, ultimately, disrupts the normal function of the heart.

A homozygous pathogenic variant, p.Arg253Trp SLC5A6, has been identified in a family whereby two siblings presented with dilated cardiomyopathy. SLC5A6 encodes the Sodium-dependent Multivitamin Transporter, a transmembrane protein that is crucial for facilitating the active transport of three vitamins: biotin, pantothenic acid and lipoic acid; all of which are critical, organic enzyme cofactors required for energy metabolism in the mitochondria.

Energy metabolism is a fundamental process in cardiac maintenance and, as the most metabolically demanding organ in the body, the heart requires a high ATP turn over to achieve efficient contractile function of the myocardium. To date, there is little known about the expression of SLC5A6 in the heart therefore, transgenic mouse models have been generated and various immunohistochemical, RNA and protein techniques have been performed in order to investigate its role.

A conditional deletion of Slc5a6 in cardiomyocytes results in the development of cardiomyopathy in mice and electron microscopy of Slc5a6 cardiac-specific knockout mice hearts showed mitochondrial degradation.
It is thus predicted that defects in Slc5a6 will prevent the transport of mitochondrial enzyme cofactors and underpin the progression to cardiomyopathy. Therefore, this project not only aims to establish the role of SLC5A6 in the heart but to investigate whether vitamin supplementation to Slc5a6 cardiac-specific knockout mice will delay progression to cardiomyopathy.


Millie FULLERTON (Newcastle, United Kingdom), Simon BAMFORTH, Helen PHILLIPS
19:00 - 20:45 #30639 - 157. Extracellular Matrix Proteoglycan Prelp modulates laminin mediated cardiac morphogenesis.
157. Extracellular Matrix Proteoglycan Prelp modulates laminin mediated cardiac morphogenesis.

The Cardiac Jelly (CJ) is a specialized extracellular matrix (ECM) found between the myocardial and endocardial layers in the embryonic heart.  Developmental processes such as cardiac looping and ballooning depend on remodeling and maturation of the CJ by small leucine-rich proteoglycans such as Proline Arginine Rich Ends Leucine-rich repeats Protein (Prelp).  Prelp interacts with large ECM proteins such as Laminins and Collagens and is hypothesized to act as an anchor between the Basal Lamina (BL) and Reticular Lamina (RL).  Using imaging of live and fixed samples, we found that zebrafish prelp is required for the assembly of the cardiac BL as assessed by Laminin localization.  Loss of prelp leads to an increased number of cardiomyocytes, enlarged atrium and weak cardiac contractilityMyocardium-specific rescue of prelp expression rescues the atrial chamber morphology defect.  Injecting mRNA encoding amino-terminal regions of Prelp in prelp mutants partially rescues the BL Laminin localization defect and improves cardiac function.  Transcriptomic analysis of prelp mutant hearts revealed the downregulation of basement membrane components such as Tenascin-C (tnc) and Fibronectin leucine-rich transmembrane protein 1b (flrt1b) which upon mRNA injections into prelp mutants rescued the BL Laminin localization defects.  Altogether, these data show the importance of small leucine rich proteoglycans in the BL in modulating atrium morphogenesis and myocardial-ECM interactions during early cardiac development.


Srinath RAMKUMAR (Bad Nauheim, Germany), Anabela BENSIMON-BRITO, Stefan GUENTHER, Didier STAINIER
19:00 - 20:45 #30642 - 159. Zfp516 deletion leads to upregulation of neuronal programme in cardiomyocytes, bradycardia, and outflow tract defects.
159. Zfp516 deletion leads to upregulation of neuronal programme in cardiomyocytes, bradycardia, and outflow tract defects.

Insights into the functions of a transcription factor ZFP516 in embryonic development are limited, except for its role in brown fat formation and regulation of stemness in embryonic stem cells. Germline homozygous loss of Zfp516 leads to neonatal lethality in mice, however, the cause of this lethality is not currently known. Here we find a role for ZFP516 in mouse embryonic heart development. Zfp516 homozygous loss results in outflow tract abnormalities at E14.5 such as the overriding aorta and in pulmonary or aortic valve phenotypes with full penetrance. Other cardiac phenotypes such as atrioventricular septal defect, perimembranous or muscular ventricular septal defects, or dorsal mesenchymal protrusion absence were observed in at least half of the E14.5 Zfp516 -/- embryos. At a molecular level we detected an upregulation of genes important in neurogenesis and synaptogenesis in murine hearts of Zfp516 -/- embryos at E13.5 with Nrn1 showing 15 fold increase in mRNA expression that was localized to a compact myocardium. At a functional level echocardiogram assessment of E13.5 murine hearts showed bradycardia with signs of atrioventricular block in Zfp516 -/- embryos, suggesting that there might be deficiencies in the function of the cardiac conducting system a day before structural defects were detected. Moreover, an investigation of lineage-specific requirements for ZFP516 in normal heart development showed a role in both the Sox10-expressing migrating neural crest and the Isl1-expressing second heart field lineages. Finally, germline analysis of >400,000 humans revealed loss of function mutations of the human ortholog are under negative selection suggesting an important role of the gene in the human development.


Joanna PRZEWROCKA (London, United Kingdom), Rifdat AOIDI, Hannah VANYAI, Stefan GEYER, Fabrice PRIN, Stefan BOEING, Deborah SCHNEIDER-LUFTMAN, Eva LANA-ELOLA, Victor TYBULEWICZ, Su Kit CHEW, Wolfgang WENINGER, Jannine CODY, Nnennaya KANU, Eva GRÖNROOS, Tim MOHUN, Charles SWANTON
19:00 - 20:45 #30647 - 161. Endocardial identity is established during early somitogenesis by Bmp signalling acting upstream of npas4l and etv2/etsrp.
161. Endocardial identity is established during early somitogenesis by Bmp signalling acting upstream of npas4l and etv2/etsrp.

The endocardium plays important roles in the development and function of the

vertebrate heart, however few molecular markers of this tissue have been identified

and little is known about what regulates its differentiation. We describe here the

Gt(SAGFF27C); Tg(4xUAS:egfp) line as a marker of endocardial development in

zebrafish. Transcriptomic comparison between endocardium and pan-endothelium

confirms molecular distinction between these populations and time-course analysis

suggests differentiation as early as 8 somites. To investigate what regulates

endocardial identity, we employed npas4l/cloche, etv2/etsrp and scl loss-of-function

models. Endocardial expression is lost in cloche mutants, significantly reduced in etv2

mutants and only modestly effected upon scl loss-of-function. Bmp signalling was

also examined: overactivation of Bmp signalling increased endocardial expression,

whilst Bmp inhibition decreased expression. Finally, epistasis experiments showed that overactivation of Bmp signalling was incapable of restoring endocardial

expression in etv2 mutants. By contrast, overexpression of either npas4l or etv2 was

sufficient to rescue endocardial expression upon Bmp inhibition. Together, these

results describe the differentiation of the endocardium, distinct from vasculature, and

place npas4l and etv2 downstream of Bmp signalling in regulating its differentiation.


Samuel J CAPON, Veronica URIBE SOKOLOV (Melbourne, Australia), Nicole DOMINADO, Ophelia EHRLICH, Kelly A SMITH
19:00 - 20:45 #30656 - 163. Scn10a-short; an electrophysiological enhancing partner of NaV1.5.
163. Scn10a-short; an electrophysiological enhancing partner of NaV1.5.

Jianan Wang, Joyce C.K. Man, Karel van Duijvenboden, Fernanda M. Bosada, Bas J. Boukens, Arie O. Verkerk, Geert J. Boink, Vincent M. Christoffels and Phil Barnett

AmsterdamUMC, AMC Amsterdam, Department of Medical Biology, Amsterdam, The Netherlands.

 

Expression of NaV1.8 (Scn10a), in cardiomyocytes, has been a topic associated with a certain level of controversy; is it there or not? With ongoing improvements in RNAseq output quality and sequence depth, genes and other genetic elements transcribed at low levels can now be detected with greater confidence. This lead to our observation that a short transcript comprising the last 7 exons of Scn10a, was being expressed in various cardiomyocyte components of the conduction system, cells in which we have never observed expression of Scn10a, or as we came to realize, full-length Scn10a. Aside from offering an explanation as to the expression controversy, we also show that this short version of NaV1.8 (Scn10a-short), also retains the ability to mediate an increased sodium current in the presence of the well described cardiac sodium channel NaV1.5 (SCN5A). Further, we have previously described a genetic variant (rs6801957), in an enhancer linked to the SCN5A/SCN10A gene locus and known to be associated with slow conduction, predisposing to arrhythmia. We now show that modification of this enhancer leads to reduced cardiac Scn10a-short expression, reduced sodium current, atrial conduction slowing and arrhythmia. Expression of the presumed target of the enhancer, Scn5a, remained unchanged.


Jianan WANG, Joyce MAN, Fernanda BOSADA, Karel VAN DUIJVENBODEN, Bas BOUKENS, Arie VERKERK, Geert BOINK, Vincent CHRISTOFFELS, Phil BARNETT (Amsterdam, The Netherlands)
19:00 - 20:45 #30659 - 165. Genetic mapping reveals that congenital heart defects in Down Syndrome are caused by increased dosage of DYRK1A.
165. Genetic mapping reveals that congenital heart defects in Down Syndrome are caused by increased dosage of DYRK1A.

Down syndrome (DS), trisomy 21, is a gene dosage disorder which results in multiple phenotypes including congenital heart defects (CHD). The genes on human chromosome 21 required in three copies to cause CHD and the altered cellular mechanisms that cause this pathology are unknown. We show that human DS fetal hearts and embryonic hearts from mouse models of DS have common transcriptional and proteomic changes, with reduced expression of oxidative phosphorylation and cell proliferation genes correlating with CHD. Using systematic genetic mapping, we identify Dyrk1a as a causative gene required in three copies to cause CHD and show that increased DYRK1A results in decreased mitochondrial respiration and reduced cell proliferation in embryonic cardiomyocytes. Thus, increased dosage of DYRK1A impairs mitochondrial function and causes CHD in DS.


Eva LANA-ELOLA (London), Rifdat AOIDI, Miriam LLORIAN, Helen FLYNN, Sheona WATSON-SCALES, Marie HAUGSTEN HANSEN, Darryl HAYWARD, Yann HERAULT, Ambrosius SNIJDERS, Elizabeth FISHER, Victor TYBULEWICZ
19:00 - 20:45 #30687 - 167. Folate supplementation protects cardiac outflow tract development from synergistic teratogenic effects of prenatal alcohol exposure and notch pathway mutations.
167. Folate supplementation protects cardiac outflow tract development from synergistic teratogenic effects of prenatal alcohol exposure and notch pathway mutations.

Background:

Acute prenatal alcohol exposure (PAE) in combination with Notch mutations in the second heart field causes highly penetrant congenital heart defects (CHD), particularly conotruncal defects. The mechanism of this interaction and potential prophylactic interventions against it remain unexplored.

 

Hypothesis:

We hypothesize that alcohol inhibits Notch signaling via the gamma-secretase complex and that prophylactic folic acid supplementation would prevent this dysregulation.  

 

Methods:

We studied the effects of acute PAE in vivo in our murine model (two injections of 3g/kg 30% ethanol on E6.5) on cardiac phenotype, Notch signaling, SHF viability, and Notch intracellular domain (NICD) and presenilin-1 localization. Experiments were repeated using C2C12 cells exposed for 24 hours to growth media supplemented with 100mM ethanol with and without folic acid or its metabolite s-adenosyl methionine (SAM).

 

Results:

Acute PAE in vivo and in vitro resulted in reduction and intracellular redistribution of presenilin-1. NICD was retained in the cell membrane, reducing downstream Notch signaling. Both folic acid and SAM supplementation prevented these molecular changes. Folic acid supplementation preserved SHF proliferation, increasing proliferation 64% compared to those on a standard diet (p<0.001), and reduced CHD incidence 78% (p<0.001). 

 

Conclusion:

Acute PAE reduces Notch signaling through disruption of the gamma-secretase complex, inhibiting nuclear translocation of NICD. This results in reduced SHF proliferation and aberrant outflow tract development. Folic acid supplementation prevented the deleterious effects of alcohol. Further study is required to decipher the molecular mechanism coupling PAE to disruption of gamma secretase complex and the impact of folic acid supplementation on this mechanism. 


Drayton HARVEY (Los Angeles, USA), Hidekazu TSUKAMOTO, Ram Kumar SUBRAMANYAN
19:00 - 20:45 #30691 - 169. Histone modifying enzyme protein arginine methyltransferase-1 is a novel epigenetic regulator of Wnt5a expression in cardiac neural crest cells during outflow tract development.
169. Histone modifying enzyme protein arginine methyltransferase-1 is a novel epigenetic regulator of Wnt5a expression in cardiac neural crest cells during outflow tract development.

We previously showed that neural crest cell (NCC)-derived Wnt5a is crucially required for planar cell polarity (PCP) signaling in second heart field (SHF) progenitors and appropriate cardiac outflow tract (OFT) alignment. While previous studies indicate that histone modifying enzyme protein arginine methyltransferase-1 (Prmt-1) regulates Wnt5a expression in cranial NCCs during palatogenesis, there is limited understanding of the regulatory mechanisms of Wnt5a during heart development. We examined the impact of cardiac NCC Prmt-1 on Wnt5a expression and outflow tract maturation in mice. We found that Prmt-1 is globally expressed in migratory and post-migratory NCCs throughout outflow tract morphogenesis. Conditional knockout of Prmt-1 in NCCs (Wnt1-Cre) resulted in fully penetrant cleft palate and double outlet right ventricle (DORV) phenotype at E14.5. Cardiac NCC migration into the OFT was preserved in Prmt-1 mutants. However, SHF progenitor cells demonstrated perturbed PCP signaling and failed to migrate into the OFT, phenocopying SHF defects previously described in NCC-Wnt5a mutants. Wnt5a transcript expression was downregulated in Prmt-1 mutant NCCs by RNAseq, RT-PCR, and in situ hybridization. 75% of Wnt1-Cre, Prmt-1F/+, Wnt5a F/+ double heterozygous mice developed DORV at E14.5, establishing the genetic synergy of Prmt-1 and Wnt5a pathways in vivo. Concomitant overexpression of Wnt5a-v5 in NCCs was sufficient to rescue DORV phenotypes in 71% (n=5/7) of Wnt1-Cre, Prmt-1F/F mice. Taken together, we show that Prmt-1 regulates Wnt5a expression in cardiac NCCs and is required for SHF PCP signaling and OFT alignment. This work defines a novel role for Prmt-1 in cardiac NCC and outflow tract biology.


Omar TOUBAT, Jongkyu CHOI, Prashan DE ZOYSA, Riya VERMA, Drayton HARVEY (Los Angeles, USA), Henry SUCOV, Ram Kumar SUBRAMANYAN
19:00 - 20:45 #30471 - 171. CHD7 modulates expression of a cardiogenic Gene Regulatory Network across the first and second heart fields.
171. CHD7 modulates expression of a cardiogenic Gene Regulatory Network across the first and second heart fields.

CHD7 is the chromatin remodeler haploinsufficient in CHARGE syndrome. Mouse models of Chd7 haploinsufficiency have cardiovascular defects similar to 22q11.2DS, and TBX1 haploinsufficiency. We assessed the role of CHD7 in the cardiopharyngeal mesoderm (CPM) by conditional mutagenesis and identified genes and pathways dysregulated in these mutants at E9.5. In cKO embryos cardiomyocyte differentiation genes and anterior SHF (aSHF) markers were decreased, but pSHF markers were increased, while expression of Tbx1 was not changed. We performed genome-wide profiling of CHD7 binding in cardiac progenitor (CP) cells and identified differentially regulated genes with CHD7 binding sites. Combination of our transcriptomics with the CHD7 binding profile shows that CHD7 acts at enhancers regulating first and second heart field gene networks, as well as a well characterized Fgf10 enhancer controlling differential expression in left ventricle versus second heart field. Thus, CHD7 is an important modulator of the mesodermal cardiogenic gene network.


Nancy STATHOPOULOU (London, United Kingdom), Ping WANG, Deyou ZHENG, Peter SCAMBLER
19:00 - 20:45 #30703 - 173. Immunosuppressant Dexamethasone prevents dilated cardiomyopathy in a mouse model of LMNA-related cardiomyopathies.
173. Immunosuppressant Dexamethasone prevents dilated cardiomyopathy in a mouse model of LMNA-related cardiomyopathies.

Dilated cardiomyopathies caused by mutations in LMNA, encoding nuclear Lamin A/C, are highly malignant and prevalent. How LMNA mutations cause cardiomyopathies remains unknown. We characterized cellular, molecular, and pathological evolution of a mouse model of LMNA-related cardiomyopathy and provide evidence for a model in which nuclear envelope rupture unleashes nuclear-localized proinflammatory signaling, as a candidate molecular mechanism underlying disease pathogenesis. We observed that cardiomyocyte-specific, tamoxifen-inducible deletion of Lmna in adult mice (LmnaCMKO) caused extensive fibrosis, increased macrophages, reduced ejection fraction, and chamber dilation by 3 weeks after Lmna gene deletion. LmnaCMKO­ cardiomyocytes exhibited localized rupture of the nuclear envelope 2 weeks prior to the development of fibrosis and reduction of ejection fraction. Nuclear envelope rupture in LmnaCMKO was immediately followed by an extensive upregulation of pro-inflammatory gene expression programs. We found that HMGB1, a potent proinflammatory protein normally sequestered in the nucleus, was released from the ruptured nuclei in LmnaCMKO cardiomyocytes. Cardiomyocyte-targeted transcriptome analysis indicated that LmnaCMKO cardiomyocytes did not upregulate the proinflammatory transcriptional program, raising the possibility that HMGB1 was further released to the extracellular space to active innate immune response in non-cardiomyocytes. Remarkably, treatment of LmnaCMKO mice with immunosuppressant Dexamethasone prevented dilated cardiomyopathy, indicating that myocardial inflammation is the major contributor to the development of Lmna-related cardiomyopathy. Future work will examine the hypothesis that extracellular HMGB1 triggers pathogenic sterile inflammation leading to dilated cardiomyopathies in LmnaCMKO mice. In conclusion, we identified the nuclear rupture-induced proinflammatory signaling as a candidate mechanism underlying LMNA-related cardiomyopathies.


Hanumakumar BOGIREDDI, Alexis STUTZMAN, Sachie IKEGAMI, Briana THOMAS, Ivan MOSKOWITZ, Kohta IKEGAMI (Cincinnati, USA)
19:00 - 20:45 #29487 - 175. Absence of Hif1α in the secondary heart field and sympathetic ganglia causes abnormalities in embryonic heart innervation and coronary vascularization.
175. Absence of Hif1α in the secondary heart field and sympathetic ganglia causes abnormalities in embryonic heart innervation and coronary vascularization.

Embryonic hypoxia is important mechanism driving heart development. Hif1α is a subunit of hypoxia-inducible factor 1 (HIF-1), regulator of transcriptional responses to decreased O2 availability. Islet1 expressing cells are found in secondary heart field, right ventricle, proximal parts of coronary arteries and among others also in the sympathetic chain.

Hif1α loxP/loxP female mice were crossed with Islet1-Cre, Hif1α loxP/+, Cx40:GFP homozygous males to generate Hif1α CKO embryos, heterozygous for the Cx40-GFP reporter to visualize coronary artery endothelium. Whole mount imunohistochemistry (TH-tyrosine hydroxylase) was used to visualize sympathetic innervation. Further analysis was performed by immunohistochemistry and microCT. 

Hif1α CKO ED17.5 hearts exhibited misconnected main coronary arteries: septal artery misconnections, variations in proximal parts of coronary arterial vasculature, and variations in branching. Venous part of the embryonic vasculature is also analyzed.

Superficial nervous plexus of the Hif1α CKO embryonic heart was less developed mainly around the base of the heart compared to controls. The alignment and patterning of the nervous and vascular plexus on ventricular surface will be further analyzed.  

 In Hif1α CKO hypoxia induced signalization altered sympathetic heart innervation. Coronary vasculature is also affected by the loss of hypoxia signalization in SHF-derived myocardium. Together Hif1α CKO exhibit disrupted patterning of coronary vasculature as well as heart innervation and its navigation along the vasculature.

Hypoxic signaling is an important developmental mechanism in cardiac nerves and coronary vasculature development. Understanding mechanisms underlaying the development of cardiac nerves and vessels can clarify the mechanisms behind sudden cardiac death in young and adult patients.  


Hana KOLESOVÁ (Prague, Czech Republic), Adrian ADZIMA, Veronika OLEJNÍČKOVÁ, Martin BARTOŠ, Gabriela PAVLÍNKOVÁ, David SEDMERA
19:00 - 20:45 #30479 - 177. Unravelling the chromatin landscape and enhancer logic mediating spatiotemporal patterning of early cardiovascular progenitors.
177. Unravelling the chromatin landscape and enhancer logic mediating spatiotemporal patterning of early cardiovascular progenitors.

The mammalian heart arises from various populations of Mesp1-expressing cardiovascular progenitors (CPs) that are specified during the early stages of gastrulation. Mesp1 is a transcription factor (TF) that acts as a master regulator of CP specification and differentiation. However, how Mesp1 regulates the chromatin landscape of nascent mesodermal cells to define the temporal and spatial patterning of the distinct populations of CPs remains unknown. Here, by combining ChIP-seq, RNA-seq and ATAC-seq during mouse pluripotent stem cell differentiation, we defined the dynamic remodelling of the chromatin landscape mediated by Mesp1. We identified different enhancers that are temporally regulated to erase the pluripotent state and specify the pools of CPs that mediate heart development. We identified Zic2 and Zic3 as essential cofactors that act with Mesp1 to regulate its TF activity at key mesodermal enhancers, thereby regulating the chromatin remodelling and gene expression associated with the specification of the different populations of CPs in vivo. Our study identifies the dynamics of the chromatin landscape and enhancer remodelling associated with temporal patterning of early mesodermal cells into the distinct populations of CPs that mediate heart development. 


Xionghui LIN, Benjamin SWEDLUND, Elodie BAUDELET (Bruxelles, Belgium)
19:00 - 20:45 #30496 - 179. LRP1 is required for epicardial development and coronary vessel formation.
179. LRP1 is required for epicardial development and coronary vessel formation.

Ligand-Receptor inference analysis identified LDL receptor related protein 1 (LRP1), an endocytic receptor involved in regulating vascular remodelling, differentiation and cell migration, as a major signalling receptor in the developing epicardium. Currently, very little is known about the role of LRP1 in epicardial formation and function. Using an inducible epicardial-specific Cre (WT1-CreERT2), LRP1 was deleted from E9.5 (LRP1 iKO). LRP1 iKO embryos showed gross morphological defects including oedema, haemorrhage, and aberrant arrangement of the myocardial wall. Histological phenotyping of LRP1 iKO showed epicardial and coronary vessel defects at E14.5, with impaired invasion of epicardium-derived cells and abnormal vascular structures within surface nodules. By E17.5, the coronary vasculature was incorrectly patterned in LRP1 iKO and epicardial-derived vascular smooth muscle cells were largely absent. Epicardial epithelial-to-mesenchymal transition (EMT) and migration were further investigated in E11.5 explants from constitutive epicardial LRP1 KO hearts (Tg(Gata5-Cre)-driven; LRP1 cKO). Compared with controls, LRP1 cKO epicardial cells displayed reduced migration, incomplete EMT and cytoskeletal defects. These data suggest that LRP1 is required for epicardial EMT, a necessary process for differentiation to mural cell derivatives. EMT is critically influenced by signalling pathways including TGF-B, PDGF-B, FGF, Wnt and Notch1, all of which can be diversely controlled by LRP1, either through co-receptor activation or receptor clearance, through relaying to signalling pathways, or by modification and turnover of extracellular matrix and heparan sulphate proteoglycans. Ongoing studies seek to define the precise mechanisms through which LRP1 impacts epicardial signalling, to further our understanding of EMT in heart development and regeneration.


Jacinta KALISCH-SMITH (Oxford, United Kingdom), Andia REDPATH, Karina DUBE, Nicola SMART
19:00 - 20:45 #30532 - 181. Tbx1-dependent and independent pathways promote Six gene expression downstream of retinoic acid signaling to pattern the cardiomyocyte progenitor field.
181. Tbx1-dependent and independent pathways promote Six gene expression downstream of retinoic acid signaling to pattern the cardiomyocyte progenitor field.

Tight regulation of retinoic acid (RA) levels is critical for normal heart development in all vertebrates. In zebrafish, early RA signaling restricts the size of the first heart field (FHF) and is required to promote differentiation of second heart field (SHF) at the arterial and venous poles of the developing heart. Here, we sought to elucidate RA-dependent regulatory networks patterning cardiac progenitors within the anterior lateral plate mesoderm (ALPM). A screen for RA-responsive genes within the ALPM of 8 somite (s) stage zebrafish embryos showed that RA loss results in an expansion of six1b and six2a, whose orthologs regulate SHF-derived outflow tract (OFT) development in mice. Quantifying differentiated cardiomyocytes (CMs) in six1b; six2a mutants and treatment with the RA-inhibitor, DEAB, indicated that RA-deficient six1b; six2a double mutants had a reduction in the number of differentiated Nkx2.5+ CMs at 48 hours post-fertilization (hpf) compared to RA-deficient six2a single mutant embryos. Previous work has indicated a Tbx1-Six1-Fgf signaling network promotes OFT development in mice. To determine if a similar signaling network functions downstream of early RA signaling, we performed in situ hybridization on Tbx1 knockdown embryos that were treated with DEAB, as tbx1 is also expanded by the 8s stage in RA deficient embryos. We found that the early expansions of six1b and fgf8a were dependent on Tbx1, while the expansion of six2a expression was independent of Tbx1. Together, our data indicate that Tbx1-dependent and independent pathways restrict Six dosage downstream of RA within the ALPM to pattern the CM progenitor field.


Tiffany DUONG (Cincinnati, USA), Andrew HOLOWIECKI, Joshua WAXMAN
19:00 - 20:45 #30556 - 183. Developmental Origin of Mammalian Cardiac Pacemaker Cells: A Clonal Fate Mapping Study.
183. Developmental Origin of Mammalian Cardiac Pacemaker Cells: A Clonal Fate Mapping Study.

BackgroundProgenitors of cardiac pacemaker cells (PCs) in the sinoatrial node (SAN) have not been genetically isolated in the mammalian embryo, making the mammalian SAN one of the last major cardiac structures whose cellular origins remain obscure. We have previously identified an enhancer for Isl1 that is active specifically in PCs during development and maturation of the SAN (Isl1-SAN-Enhancer or ISE). Here, we explored whether ISE could be used as a genetic marker to probe lineage dynamics of early SAN progenitors. 

Methods and Results: ISE-hsp68-CreERT2 transgenic mouse lines were generated with pronuclear injection to facilitate genetic labeling of putative SAN progenitors. Fate mapping with this mouse line showed that ISE-CreERT2+ cells contribute to SAN and right atrial myocardium beginning at E8.5, with progressive restriction to SAN fate at later timepoints, raising the possibility that ISE-CreERT2+ cells are initially bipotent at E8.5 and commit to PC or right atrial cardiomyocyte fate between E8.5 and E9.5. To test this possibility, we performed clonal fate mapping using Mosaic Analysis with Dual Markers (MADM), a strategy that relies on mitotic recombination to generate red/green-fluorescent or unlabeled/yellow-fluorescent “twin spots” that can be visually tracked to demonstrate clonal relationships. To do this, we crossed ISE-CreErt2; MADM-11GT/GT with MADM-11TG/TG, induced Cre activity with tamoxifen at E8.5, and analyzed clones at E14.5. 30 twin spots have been observed thus far, and immunostaining is underway to accurately determine locations and cellular identities of each twin spot. 

Conclusions:  (1) ISE-CreERT2 mice permit labeling, isolation, and genetic manipulation of PCs and their progenitors. (2) ISE-CreERT2+ progenitors at E8.5 contribute to the SAN and portions of the right atrium. (3) Clonal fate mapping will define the potency of these progenitors and the timing of their commitment to PC fate.


Giselle GALANG (San Francisco, USA), Gagandeep CHOUHAN, Hongmei RUAN, Rajiv MOHAN, Tanvi SINHA, Patrick DEVINE, Brian BLACK, Vasanth VEDANTHAM
19:00 - 20:45 #30582 - 185. Defining the genetic program pre-specifying early cardiac progenitor cells fate.
185. Defining the genetic program pre-specifying early cardiac progenitor cells fate.

During gastrulation, cardiac progenitors (CPs) are identified by the expression of the transcription factor Mesp1. Previous studies have demonstrated that CPs are already spatiotemporally restricted to specific cardiac lineages by that stage. Thus, the heart is built from distinct CPs that display different potential. Recent transcriptional analyses have uncovered the molecular heterogeneity of CPs. In order to understand how CP heterogeneity affects their fate in the adult heart, our aim is to define and modulate the mouse genetic program or “heart code” driving CP specification. We are focusing, in particular, on homeodomain transcription factors that are heterogeneously expressed in CPs. To this end, our objectives are to identify CP sub-populations by defining the spatiotemporal pattern of homeodomain genes during gastrulation and their final cardiac contributions. The role of the homeodomain genes in regulating the pre-specification of CPs are assessed by modulating their expression and analyzing how it impacts CP subpopulations in vivo and in vitro. Our promising preliminary results have already pinpointed the spatial heterogeneity of homeodomain gene expression during gastrulation and have suggested an important role for Lhx1 in Mesp1+ progenitors to correctly form head structures. Together these results will allow the dynamic characterization of heterogeneity within CPs and the identification of putative factors that could trigger the differentiation toward one particular cardiac cell type. These studies have important implications for better understanding the etiology of congenital cardiac malformations and should be the starting point of further applications for improving cell therapy during cardiac repair.


Caroline CHOQUET (Marseille), Fabienne LESCROART
19:00 - 20:45 #30604 - 187. Ventricular, atrial, and outflow tract heart progenitors arise from spatially and molecularly distinct regions of the primitive streak.
187. Ventricular, atrial, and outflow tract heart progenitors arise from spatially and molecularly distinct regions of the primitive streak.

The heart develops from 2 sources of mesoderm progenitors, the first and second heart field (FHF and SHF). Using a single-cell transcriptomic assay combined with genetic lineage tracing and live imaging, we find the FHF and SHF are subdivided into distinct pools of progenitors in gastrulating mouse embryos at earlier stages than previously thought. Each subpopulation has a distinct origin in the primitive streak. The first progenitors to leave the primitive streak contribute to the left ventricle, shortly after right ventricle progenitor emigrate, followed by the outflow tract and atrial progenitors. Moreover, a subset of atrial progenitors are gradually incorporated in posterior locations of the FHF. Although cells allocated to the outflow tract and atrium leave the primitive streak at a similar stage, they arise from different regions. Outflow tract cells originate from distal locations in the primitive streak while atrial progenitors are positioned more proximally. Moreover, single-cell RNA sequencing demonstrates that the primitive streak cells contributing to the ventricles have a distinct molecular signature from those forming the outflow tract and atrium. We conclude that cardiac progenitors are prepatterned within the primitive streak and this prefigures their allocation to distinct anatomical structures of the heart. Together, our data provide a new molecular and spatial map of mammalian cardiac progenitors that will support future studies of heart development, function, and disease.


Kenzo IVANOVITCH (UK, United Kingdom)
19:00 - 20:45 #30628 - 189. Why the heart of coelacanth fish necessitates a major revision of the ancestral heart of terrestrial vertebrates.
189. Why the heart of coelacanth fish necessitates a major revision of the ancestral heart of terrestrial vertebrates.

The heart of the first land-living vertebrates, tetrapods, is inferred by comparing present-day lungfish and amphibians. These groups belong to lobe-finned fish as do the rare coelacanths. Few coelacanth hearts have been studied and it has been concluded they are very primitive because of a linear chamber topology. Until now, the coelacanth heart has been omitted from inferences of the ancestral tetrapod heart. Using MRI, CT, and 3D-modelling, we assessed the morphology of in-situ hearts of two coelacanths, 13 specimens from all three major groups of amphibians, five specimens from all three major groups of lungfish, nine ray-finned fish, 22 cartilaginous fish, two lampreys, and two hagfish. The coelacanth heart is long mostly due to an elongate outflow tract, but this setting resembles that of fellow ray-finned non-teleost fish. The cardiac chambers have a substantial left-right symmetry as in other bony fish, while the coelacanth sinus venosus and atrium are positioned relatively caudally. Nonetheless, the coelacanth heart is much different from the hearts of cartilaginous fish, lampreys, and hagfish, and it is not primitive. We then assessed 20 gross morphological characters in amphibians, lungfishes, and coelacanths. Nine were shared by all groups. Five were shared by amphibians and lungfishes only, such as a lung vein connecting to the left atrium and an atrial septum. Six were shared by amphibians and coelacanth, such as a valved atrioventricular junction (lungfishes have a ‘plug’) and absent ventricular septum. We conclude the ancestral tetrapod heart likely had a lungfish-like atrium and a coelacanth-like ventricle.


Bjarke JENSEN (Amsterdam, The Netherlands), Henrik LAURIDSEN
19:00 - 20:45 #29485 - 191. The cardiomyocyte mechanical code – mechanical imprinting of talin interactions regulate cardiomyocyte mechanosensing and function.
191. The cardiomyocyte mechanical code – mechanical imprinting of talin interactions regulate cardiomyocyte mechanosensing and function.

Mechanical properties are cues for many biological processes in health or disease(1). In the heart, changes to the extracellular matrix composition and cross-linking results in stiffening of the cellular microenvironment during development. Moreover, remodelling after myocardial infarction, or in cardiomyopathies lead to fibrosis and a stiffer environment. By combining nanopillar arrays, PDMS gels with defined stiffness and FRET molecular tension sensors, we previously identified a fundamental mechanism for cardiomyocyte rigidity sensing, whereby single cardiomyocyte adhesions sense simultaneous (fast oscillating) cardiac and (slow) non-muscle myosin contractions. Together this results in extracellular matrix dependent levels and dynamics of tension on the mechanosensitive adaptor protein talin(2). To investigate the mechanical signalling further, we analyse force dependent and force-independent talin interactions using specifically designed biofunctionalized nanoarrays, biochemistry and various microscopy approaches(3)(and unpublished data). These experiments surprisingly reveal a mechanical imprinting that is resulting in changes to the talin interactome, cytoskeletal organisation and cardiomyocyte mechanics.

 

 

1.         T. Iskratsch, H. Wolfenson, M. P. Sheetz, Appreciating force and shape-the rise of mechanotransduction in cell biology. Nat Rev Mol Cell Biol 15, 825-833 (2014).

2.         P. Pandey et al., Cardiomyocytes Sense Matrix Rigidity through a Combination of Muscle and Non-muscle Myosin Contractions. Dev Cell 44, 326-336 e323 (2018).

3.         W. Hawkes et al., Probing the nanoscale organisation and multivalency of cell surface receptors: DNA origami nanoarrays for cellular studies with single-molecule control. Faraday Discuss 219, 203-219 (2019).


Emilie MARHUENDA, Pragati PANDEY, Ioannis XANTHIS, Caoimhe O'NEILL, Iskratsch THOMAS (London, United Kingdom)
19:00 - 20:45 #30425 - 193. Recapitulating the phenotype of long QT syndrome type 2 in induced pluripotent stem cells-derived cardiomyocytes.
193. Recapitulating the phenotype of long QT syndrome type 2 in induced pluripotent stem cells-derived cardiomyocytes.

Introduction: Long QT syndrome (LQTS) is an inherited primary arrhythmia syndrome with a hallmark of QT prolongation on 12-lead ECG. The mechanism of disease is secondary to a delayed repolarization and pathologically prolonged action potential duration of cardiomyocytes. LQTS type 2 (LQT2) is associated with loss-of-function variants in the KCNH2 gene, affecting the rapid rectifier potassium current (IKr). 

Objective: Studying LQTS in an induced pluripotent stem cell-derived cardiomyocyte (iPSC-CM) model allows for patient-specific phenotype characterization in vitro. We report such a strategy in a LQT2 patient (ECG QTc 492msec) with KCNH2 (c.2464G>A) pathogenic variant, and in a healthy control for comparison (ECG QTc 388msec).

Methods: Human iPSCs were generated from patient and control dermal fibroblasts using a non-integrational Sendai reprogramming method. Beating cardiomyocyte clusters were dissociated and seeded onto Maestro CytoView multi-electrode array plates (Axion BioSystems). Serial corrected field potential duration (FPDc) measurements (n=26), equivalent to QTc values on ECG, were recorded on days 24 to 39 of cardiomyocyte differentiation per sample. Cardiomyocytes were exposed to varying concentrations of Verapamil, Mexiletine, Sotalol, Clarithromycin, and E-4031 through cell culture media exchange. Paired t-tests (non-parametric) were utilized for P<0.05.

Results: Reprogrammed iPSCs displayed pluripotency characteristics. IPSC-CMs had confirmed cardiac lineage. Prolonged FPD values, corrected for beating rate, were observed in the LQT2 patient-derived iPSC-CMs (P<0.0001). Verapamil, Mexiletine, Sotalol, Clarithromycin, and E-4031 altered FPDc in a dose-dependent response.  

Conclusions: Our findings indicated that an iPSC-CM model reliably recapitulates the LQT2 disease phenotype. Drug effects on cardiomyocyte repolarization were demonstratable in the iPSC-CM model.


Ning GE (Galway, Ireland), Min LIU, Janusz KRAWCZYK, Veronica MCINERNEY, Joseph GALVIN, Deirdre WARD, Catherine MCGORRIAN, Timothy O’BRIEN, Sanbing SHEN, Terence W PRENDIVILLE
19:00 - 20:45 #30514 - 197. Gastruloids as an in vitro models of cardiopharyngeal mesoderm specification into skeletal and cardiac muscle lineages.
197. Gastruloids as an in vitro models of cardiopharyngeal mesoderm specification into skeletal and cardiac muscle lineages.

Cardiopharyngeal mesoderm (CPM) contributes to the formation of the heart and head muscles. While the mechanisms governing CPM specification remains relatively unknown, there is a lack of an in vitro platform that would allow the differentiation of both head and heart muscles. Here, we show that the formation of embryonic organoids from mouse embryonic stem cells (mESC), also called gastruloids, allow the formation of the CPM and its specification toward the cardiac and muscle lineages. By using single-cell RNAseq, and high-resolution imaging of single molecule FISH experiments, we show that the CPM is established in gastruloids, with a kinetic of gene expression that is similar to the mouse embryo. By performing lineage tracing in gastruloids, we further demonstrate that the head and heart muscles formed in this in vitro model derive from CPM progenitors that activates the anterior second heart field specific enhancer of Mef2c. These findings unveil the potential of mESCs-derived gastruloids to undergo CPM specification in both head and heart lineages, allowing the investigation of the mechanisms of CPM specification in development and how this could be affected in congenital diseases.


Laurent ARGIRO (Marseille), Celine CHEVALIER, Nitya NANDKISHORE, Caroline CHOQUET, Anaïs BAUDOT, Stephane ZAFFRAN, Fabienne LESCROART
19:00 - 20:45 #30548 - 199. Modeling cardiac fibroblast heterogeneity from human pluripotent stem cell derived epicardial cells.
199. Modeling cardiac fibroblast heterogeneity from human pluripotent stem cell derived epicardial cells.

Cardiac fibroblasts derive from the fetal epicardium and are known to play essential roles in heart development during fetal life and in disease progression in the adult organ.  To model cardiac fibroblast development, we established organoids consisting of hPSC-derived epicardial cells and cardiomyocytes. Within a few days of organoid formation, the two populations segregated in the structures, with the epicardial cells establishing an epithelial layer surrounding the inner population of cardiomyocytes, recapitulating the organization of these two cell types in the fetal heart.   This segregation disappears over the next 10 days of culture as the epicardial cells undergo an epithelial to mesenchymal transition and give rise to fibroblasts and smooth muscle cells that invade the organoid.  To be able to use the organoids to model disease, we treated them with a combination of hormones and activators of fatty acid metabolism to promote the maturation of both the cardiomyocyte and fibroblast populations.  When subjected to pathological stimuli, both the cardiomyocytes and fibroblasts within the mature organoids recapitulated changes observed in the corresponding populations in the failing human heart. Single-cell RNA-seq and RT-qPCR analyses revealed a high degree of fibroblast heterogeneity and identified a unique subpopulation of cells with reparative features in the ‘heart failure’ organoids. Together, these findings demonstrate that it is possible to recapitulate the epicardial-cardiomyocyte interactions in simple organoid structures that lead to the development of cardiac fibroblasts. They also show that these organoids can be used to model aspects of heart failure and enable the identification of specific fibroblast populations that may play specific roles in the progression of heart disease.   

 


Ian FERNANDES (Toronto, Canada), Shunsuke FUNAKOSHI, Homaira HAMIDZADA, Slava EPELMAN, Gordon KELLER
19:00 - 20:45 #30607 - 201. Defining molecular mechanisms of endothelial differentiation during the early stages of embryonic cardiopharyngeal mesoderm.
201. Defining molecular mechanisms of endothelial differentiation during the early stages of embryonic cardiopharyngeal mesoderm.

Introduction. The cardiopharyngeal mesoderm (CM) differentiates into endothelial (EC), skeletal and cardiac muscle cells. Our goal is to identify enhancers required for EC differentiation from CM. We have used a serum-free differentiation protocol in mouse ES cells (mESCs) that induces CM differentiation and performed RNA-seq and ATAC-seq to define transcription and chromatin accessibility profiles.
Results. mESCs differentiation induced the expression of EC-specific markers at day 4 of differentiation (d4), including Pecam1, Cdh5, Eng, Kdr, Gata2, Gata6, Ets1, Flt1. RNAseq performed between d2 and d4 identified 1735 differentially expressed genes, many of which are involved in angiogenesis, indicating the activation of an EC transcription program. ATAC-seq revealed 6348 regions that became accessible during this time window. Based on these data, we identified 10 putative enhancers associated with genes involved in EC differentiation. Sequence analyses of regions opened at d4 identified Gata1, Gata2 and JunB transcription factors. We are following two strategies to validate the putative enhancers: DNA editing and epigenetic decommissioning. We first generated mES cells with deletion of putative enhancers of Pecam1 and Notch1, which we then differentiated towards CM-EC lineages. However, Pecam1 and Notch1 expression was unchanged, suggesting that these regions are unrelated to the transcription process. For the second approach, we recently obtained mES cells that stably express dCas9-LSD1 that will be used to test enhancer decommissioning. We performed an EC-specific modification of differentiation protocol, that induced ECs with high efficiency (91%). Pecam1 and Notch1 enhancers will be tested on this new differentiation conditions.
Conclusions. Our ATAC-seq data efficiently identifyied putative endothelial enhancers. Genetic and epigenetic manipulation of these sequences will establish their requirement for EC differentiation from cardiopharyngeal mesoderm.


Ilaria AURIGEMMA (Salerno, Italy), Andrea CIRINO, Gabriella LANIA, Varsha POONDI KRISHNAN, Claudia ANGELINI, Monica FRANZESE, Antonio BALDINI, Elizabeth Anne ILLINGWORTH
19:00 - 20:45 #30613 - 203. Improved mitochondrial metabolism in human pluripotent stem cell-derived left ventricular-like cardiomyocytes.
203. Improved mitochondrial metabolism in human pluripotent stem cell-derived left ventricular-like cardiomyocytes.

Cardiac disease is the leading cause of death in the western world and a great proportion of cardiac-related deaths are a result of heart failure or acute myocardial infarction. Good cellular models to mimic primary cardiac cells suitable for drug screening and the generation of disease models are therefore needed. In our lab, we developed a novel protocol for human pluripotent stem cell differentiation into left ventricular-like cardiomyocytes (hPSC-LV-CMs). This protocol results in a homogeneous population of cells with more mature features compared to time matched cells produced by the widely used wnt-on/wnt-off protocol (Standard protocol). Mitochondrial metabolism is an important feature of a mature cardiomyocyte; mitochondria are the main producers of energy in the cardiac tissue, they control calcium handling, signalling and apoptosis. When we sought to understand what happens to mitochondrial function and shape we found that our hPSC-LV-CMs have more mitochondria spread along the myofibers, enhanced respiration and higher mitochondrial membrane potential compared to cardiomyocytes generated using the standard protocol. In addition, hPSC-LV-CMs have more elongated mitochondria, a feature that correlates usually with improved function and increased ATP production. When we grew our LV cardiomyocytes in media rich in fatty acids-the main energy fuel of the adult heart- we were able to further improve mitochondrial spare respiratory capacity, suggesting that modulation of nutrients alone can metabolically mature our LV cardiomyocytes. Fine tuning the maturation media regime will further improve our model and make it more suitable for research aimed at ameliorating LV function and disease phenotypes.


Lorenza Iolanda TSANSIZI (London, United Kingdom), Elisa FERRARO, Nicola DARK, Marie-Victoire COSSON, Rifdat AOIDI, Anne WESTON, Lucy COLLINSON, Victor TYBULEWICZ, Andreia Sofia BERNARDO
19:00 - 20:45 #30624 - 205. Deciphering the molecular mechanisms underlying congenital heart defects in Noonan syndrome using hiPSCs.
205. Deciphering the molecular mechanisms underlying congenital heart defects in Noonan syndrome using hiPSCs.

Noonan syndrome (NS) constitutes one of the most common causes of congenital heart defects (CHD). NS infants with mutations in RAF1 present with cardiomyopathy and a variety of CHD. No treatment exists for NS children with CHD or cardiomyopathy, therefore there is an urgent need to understand the mechanisms underlying the disorder to identify new therapies.

Toward that goal, we used hiPSCs as a human cardiogenesis model and made hiPSCs from children with NS carrying mutations in the CR2 domain of RAF1. In addition, we generated isogenic corrected as well as RAF1 KO cells using CRISPR-Cas9dn. To investigate the impact of NS and KO RAF1 on cardiogenesis, we differentiated our isogenic lines towards a cardiac fate. Using flow cytometry for cTNT+ cells at day 15, we first found that NS RAF1 increased cardiac differentiation efficiency (74% vs 58%, n=5, p<0.05), while RAF1 deletion drastically reduced it (15%, n=5, p<0.05), demonstrating that RAF1 regulates cardiac development and NS mutations impair this process. At day 6, cardiac progenitor differentiation measured by flow cytometry for NKX2.5+ cells was diminished in KO cells (14% vs 63%, n=5, p<0.05), while it was potentiated by NS mutations (73%, n=5, p<0.05), suggesting that RAF1 regulates cardiac differentiation early on during development. Using RNA-seq at day 2, we found that NS RAF1 significantly increased expression of mesoderm genes (DLL1, MESP1/2, TBX6), whereas RAF1 KO reduced it. Moreover, GSEA revealed that NS RAF1 was associated with increased expression of EMT (SNAI2, VIM, FBN1) as well as Wnt/β-catenin target genes (TCF7, LEF1, FZD1, DVL2). On the contrary, RAF1 KO led to a strong reduction of the expression of those genes.

Taken together, we show that RAF1 is required for human cardiac differentiation and reveal that NS mutants impair cardiac mesoderm specification by impacting the Wnt/β-catenin pathway. Experiments are in progress to further delineate how RAF1 regulates cardiac mesoderm specification.


Kelly BANKS, Miriam GORDILLO, Todd EVANS, Fabrice JAFFRE (New York, USA)
19:00 - 20:45 #30634 - 207. Deciphering the endothelial cell-cardiomyocyte crosstalk in LMNA cardiomyopathy using 3D models.
207. Deciphering the endothelial cell-cardiomyocyte crosstalk in LMNA cardiomyopathy using 3D models.

Mutations in LMNA, the gene that encodes lamin A/C are the most common cause of familial dilated cardiomyopathy (DCM), often referred as cardiolaminopathy. Despite LMNA being ubiquitously present, the mechanisms that underlie cardiolaminopathy remain elusive. Using induced pluripotent stem cell (iPSCs)-derived endothelial cells (iPSC-ECs), we recently showed that LMNA-induced DCM, due to a frameshift variant caused endothelial dysfunction. Next generation sequencing identified Krüppel-like Factor 2 as the transcription factor responsible for the EC dysfunction, which was reversed by a subset of statins, including lovastatin both in vitro and in vivo. Importantly, iPSC-cardiomyocytes (iPSC-CMs) from LMNA-DCM patients showed improvement in their function when co-cultured with iPSC-ECs and lovastatin, indicating an intricate crosstalk between the ECs and CMs in LMNA cardiomyopathy. Despite impressive progress, little attention has been given to the potential importance of cell-to-cell signaling between ECs and CMs, even though ECs serve a paracrine function to enhance signaling in CMs. To decipher this, we fabricated in vivo-like cardiac 3D engineered heart tissues (EHTs) from LMNA patient’s iPSC-CMs and iPSC-ECs and performed high-resolution assessment of their phenotype. Moreover, single-cell omics on these 3D EHTs revealed an exquisite ligand-receptor (L-R) interaction that we believe to be involved in the EC-CM crosstalk in cardiolaminopathy patients. Indeed, loss of function studies further validated the importance of this L-R pair both in vitro as well as in vivo. Results from this study, which leverages multidisciplinary platforms such as iPSCs, 3D bioengineering models, and single cell-omics, could potentially lead to new therapeutic strategies for DCM patients.


Chun LIU, Sadhana GADDAM, Dilip THOMAS, Pedro MEDINA, Mohamed AMEEN, Karim SALLAM, Nazish SAYED (Stanford, USA)
19:00 - 20:45 #30717 - 209. Cardioids reveal compartment specific characteristics of human fetal heartbeats.
209. Cardioids reveal compartment specific characteristics of human fetal heartbeats.

Understanding how the heartbeat starts and is propagated in the human tissue, has been unexplored, because the earliest we can get a recording is well into heart development (around day 26). However, in mice, the developing heart starts beating earlier during the formation of the cardiac crescent (similar to day 16 in humans). During this early stage of development, the propagation of signal undergoes a complex shift, in the initiation of the beat, before the pacemakers have formed. Here we use human self-organizing cardioids to decipher the ontogeny of early heartbeats by tracking contraction, field potentials, and calcium transients for different compartments of the heart, including the left ventricle, right ventricle, atria, and atrioventricular canal.  We find that each compartment has distinct characteristics which develop differently over time. With this knowledge we will be able to gain deeper understanding of the mechanisms of the initiation and propagation of human heartbeats.


Alison DEYETT (Vienna, Austria), Clara SCHMIDT, Tobias ILMER, Aranxa TORRES CABALLERO, Simon HAENDELER, Lokesh PIMPALE, Sasha MENDJAN
19:00 - 20:45 #30511 - 211. Transcription factor WT1 modulates HSPG-dependent signalling by directly regulating endosulfatase expression in the embryonic epicardium.
211. Transcription factor WT1 modulates HSPG-dependent signalling by directly regulating endosulfatase expression in the embryonic epicardium.

During development, the epicardium contributes to the heart’s cellular components and guides and supports expansion of the vascular network. A multitude of coordinated heparan sulfate proteoglycan (HSPG)-dependent pathways control epicardial cell activity. Sulf1 and Sulf2 - enzymes that modify HSPGs – present another level of control to refine signalling separate to ligand presence. WT1 is required for heart development and denotes the epicardium’s activation state both in the embryonic and injured heart. Wt1 and Sulf1 are enriched in the activated adult epicardium post-injury. Regulation of Sulfs and their specific roles in the epicardium are unknown. Here, we used in situ hybridisation techniques and scRNA-seq to investigate Sulfs over the course of embryonic mouse heart development. In addition, we carried out ATAC-seq to investigate open chromatin in the Sulf genes, and to identify potential WT1 transcription factor occupancy. Our study shows Sulf1 is expressed in epicardial progenitors and the forming epicardium, whilst Sulf2 is expressed in cardiomyocytes during early cardiac development. Sulf1 strongly co-localises with Wt1 expression, with levels further coinciding with loss of Wt1 as the epicardium quiesces or undergoes epiEMT. Wt1 null embryonic hearts, which form an irregular epicardial layer, demonstrated lower levels of Sulf1 and upregulated Sulf2. We found that WT1 binds to the Sulf1 promoter, but no binding was detected within the Sulf2 gene in E13.5 epicardial cells. Thus, while WT1 directly regulates Sulf1 expression, upregulation of Sulf2 instead occurs indirectly. Our findings show WT1 differentially regulates Sulfs to mediate HSPG-dependent signalling in the embryonic epicardium.


Andia Nicole REDPATH (Oxford, United Kingdom), Irina-Elena LUPU, Samuel KRASNER, Joaquim Miguel Nunes VIEIRA, Nicola SMART
19:00 - 20:45 #30621 - 213. Cardiac microRNA let-7 is necessary for heart function and non-autonomously controls metabolic pathways in adipose tissue.
213. Cardiac microRNA let-7 is necessary for heart function and non-autonomously controls metabolic pathways in adipose tissue.

microRNAs (miRNAs) are an evolutionary conserved regulatory RNAs involved in many biological processes, including heart development as well as cardiac disease. miRNAs of the let-7 family are highly prevalent in the heart, and their expression is altered in several cardiovascular diseases, such as dilated cardiomyopathy and arrhythmia. Overexpression of let-7 was promotes hPSC-derived cardiomyocyte maturation and adult-like metabolism. In humans, the let-7 family consists of 12 paralogous miRNAs with largely redundant function, which has been limiting the identification of target genes and understanding let-7’s role in establishing or maintaining heart function.

To explore the role of let-7 we used the Drosophila model that only has a single let-7 miRNA family member. We found that in flies let-7 expression is enriched in the heart, and loss of let-7 causes cardiac dilation, systolic dysfunction, and arrhythmia, indicating that let-7 is a critical for many aspects of heart function. To identify let-7 effector genes we performed proteome analysis on flies where let-7 was depleted in cardiac and somatic muscle. We found a very specific response of gene products involved in lipid and glucose metabolism, such as APOB/Apolpp and SORD/Sodh-1, as well as signaling proteins like TGFB1/Dawdle and BMPR1B/Thickveins. Most responding proteins seemed to be highly enriched in the fat body, the fly’s adipose tissue, which points to non-autonomous consequences of loss of let-7 in heart and muscle on the adipose tissue.

We also cross-validated these findings with single-cell sequencing data from female and male adult fly tissues and were able to confirm that most proteins identified in our proteomics study are indeed transcribed in the fat body, many of them being differentially expressed between female and male Drosophila tissues. We therefore hypothesize that let-7 might also serve as a switch that controls tissue metabolism downstream of sex-specifying pathways


Jerome CARTRY, James KEZOS, Marco TAMAYO, Rolf BODMER, Georg VOGLER (San Diego, USA)
19:00 - 20:45 #30661 - 215. Targeting cardiomyocyte survival for cardioprotection.
215. Targeting cardiomyocyte survival for cardioprotection.

Congenital heart disease is an independent risk factor for arrhythmias and cardiac remodelling which leads to heart dysfunction and failure. Heart failure remains one of the main causes of death worldwide. It is associated with cardiomyocyte death, a mechanism that is not fully understood. The zinc-finger transcription factor GATA4, a known regulator in cardiac development, is essential to circumvent cell death along with the AMPK/Sirtuins/PGC1α energy sensor pathway. In collaboration with a medicinal chemistry lab, LCB-2122 was synthesized to target survival pathways. LCB-2122 enhanced ejection fraction in mice overexpressing angiotensin receptor type 1 (AT1R) and mice treated with the chemotherapeutic agent Doxorubicin (DOX). In AT1R mice, LCB-2122 improved mitochondrial structure and rescued against DOX-induced cell death. Here, I aim to decipher the mechanism of action of LCB-2122. Interestingly, ejection fraction was also enhanced in GATA4 heterozygous mice treated with DOX. We hypothesize that LCB-2122 enhances mitochondrial function and activates energy-producing pathways. In primary cardiomyocytes, LCB2122 abolished the cell death caused by DOX and increased phosphorylation of AMPK and its downstream target acetyl Co-A carboxylase. This effect is proven to be AMPK dependent. In-vitro, the presence of LCB-2122 augmented Sirtuins 1 and 3 activities. MitoSOX-Red shows that LCB-2122 decreases DOX-induced mitochondrial superoxide. Also, LCB-2122 increases GATA4 levels and rescues the DOX-induced GATA4 depletion. We will next use unbiased approaches such as the metabolomics and photoaffinity experiment to guide us to new direct binding partners to LCB-2122. Furthermore, other LCB-2122 derivatives are synthesized and tested to study the structure-function relationship to enhance bioactivity and stability. This project will introduce a new potential drug that protects against cardiotoxicity by activating cardiomyocyte survival pathways.


Abir ALMAZLOUM (OTTAWA, Canada), Starr DOSTIE, Hiba KOMATI, Mona NEMER, Michel PREVOST, Yvan GUINDON
19:00 - 20:45 #29437 - 217. Investigating endothelial cell behavior during vascular regeneration at single-cell resolution in zebrafish.
217. Investigating endothelial cell behavior during vascular regeneration at single-cell resolution in zebrafish.

Endothelial damage is directly associated with various cardiovascular diseases, including stroke and ischemic heart disease, the two leading causes of death globally.  Previous studies showed that vascular regeneration involves a subset of regenerating endothelial cells (ECs) with distinct proliferative ability and molecular signature.  However, the specific molecular mechanisms driving vascular regeneration remain elusive.   

Here, we establish and characterize a zebrafish vascular regeneration model by utilizing nitroreductase-mediated EC ablation at embryonic and larval stages.  By following the regenerating ECs during regeneration using state-of-the-art high-resolution time-lapse imaging, we observed that certain ECs are more proliferative, sprout better, and reconnect with the neighboring ECs.  These data suggest a heterogeneous response to tissue damage by some EC subpopulations, which possess a higher capacity of repopulating the vasculature.  To delineate this heterogeneity, we performed single-cell RNA-sequencing after EC ablation and recovered 6051 cells with 8 major cell types from the larval caudal region.  We further analyzed 1929 ECs and found two sub-clusters displaying a higher proliferative capacity.  One of these proliferative EC sub-clusters expresses genes involved in Notch signaling, leading us to hypothesize that it is involved in vascular repair.  We now aim to unravel the molecular and cellular features of these proliferative EC sub-clusters by performing cell-specific manipulations in vivo.  Overall, our findings can help us gain a better understanding of the gene programs critical for vascular repair and regeneration, which may have therapeutic implications.


Shengnan ZHAO (Bad Nauheim, Germany), Stefan GÜNTHER, Didier STAINIER
19:00 - 20:45 #30482 - 219. Inhibition of cysteine endopeptidase alters the balance between regeneration and fibrotic repair in the neonatal heart.
219. Inhibition of cysteine endopeptidase alters the balance between regeneration and fibrotic repair in the neonatal heart.

After myocardial infarction (MI), the myocardium heals through a process of scarring, in which previously healthy muscle is replaced by fibrous tissue. Remarkably, fish and neonatal mice do not heal by permanent scarring but are able to fully regenerate the heart. Immune cells are essential to this process, though their precise roles are complex and not fully understood.

In order to resolve this complexity we performed scRNA-seq analysis of the murine neonatal heart before and after injury. This analysis allowed us to identify specific genes and pathways involved in the different reparative processes acting in neonatal pro-regenerative (P1) and pro-fibrotic (P7) hearts. Interestingly, differential gene expression and gene set enrichment analysis indicated changes in the P1 granulocytes population such as highly significant enrichment in cysteine endopeptidase inhibitory activity (Stefin A1), dampened inflammation and enriched extracellular matrix (ECM) remodeling. We hypothesized that the inhibition of catalytic enzymes involved in matrix  remodeling in P1 hearts might play a role in altering the structure of the scar tissue. To test the hypothesis, we used a commercially available cysteine endopeptidase inhibitor (E64) to inject P7 mice after MI. The in vivo experiment showed structural changes in the scar formation at 21 days post-injury with a significative reduction in the coefficient of alignment of collagen fibers. These structural changes might lead to the formation of a less compact, more unstable scar which could be more easily resorbed thus creating a more permissive regenerative environment.

In conclusion, here we identify new fibrotic pathways that may be targeted to extend the regenerative capacity of the heart beyond neonatal stages into adult life. 


Daniela PEZZOLLA (Oxford, United Kingdom), Konstantinos KLAOURAKIS, Adam T BRAITHWAITE, Carla DE VILLIERS, Elisabetta GAMEN, Paul R RILEY, Robin P CHOUDHURY
19:00 - 20:45 #30490 - 221. Genomic regulation of heart tube morphogenesis by Tbx5 and Mef2c.
221. Genomic regulation of heart tube morphogenesis by Tbx5 and Mef2c.

Tbx5 and Mef2c encode critical cardiac transcription factors that are necessary for proper development of the looped heart tube. In Tbx5 knockout (KO) mice, the sinoatrial structures and primitive left ventricle are severely hypoplastic at E9.0. Conversely, Mef2c KO leads to the apparent absence of the right ventricle and hypoplastic outflow tract at E9.0. These two complementary, mutant phenotypes provide a suitable platform with which to dissect the genomic regulation of heart tube morphogenesis. To gain a more complete understanding of the genomic regulators underlying heart tube formation, we have performed combined single-nucleus RNA- and ATAC-sequencing on wild type, Tbx5 KO, and Mef2c KO embryos at E7.75 (cardiac crescent) and E9.0 (looped heart tube) using the 10x Genomics Multiome system. Initial analysis of gene expression data revealed a delay in cardiomyocyte differentiation in Mef2c KO mice at E7.75. Additionally, we found an expansion of atrial and atrioventricular cushion markers in the Mef2c KO cardiomyocytes that formed by E9.0. We are currently integrating the ATAC-seq data to determine changes in chromatin accessibility that underlie the improper specification of cardiac progenitors. Next, we will conduct live embryo imaging of wild type and mutant embryos expressing fluorescent markers that demarcate first and second heart field lineages to discern precisely when and where linear heart tube formation goes awry. Together, these studies will provide fundamental insights into how the mammalian heart takes shape during embryogenesis.


Jon MUNCIE (San Francisco, USA), Sarah WINCHESTER, Benoit BRUNEAU
19:00 - 20:45 #30525 - 223. Myocardial infarction ´through the window´: dual dynamics for cardiac fibroblasts activation.
223. Myocardial infarction ´through the window´: dual dynamics for cardiac fibroblasts activation.

Activated cardiac fibroblasts (CFs) are responsible for the healing of the heart tissue after a myocardial infarction (MI). Based on high throughput technologies, several groups have recently demonstrated their heterogeneity and a unique role of each subpopulation of CFs during the ventricular remodelling process. This is relevant towards the discovery of personalized treatments to control the initial post-MI healing scar that will contribute to preserve ventricular function and prevent the onset of heart failure. However, little is known about the moment that CFs are activated, and which genes are potentially involved in this process. Using a mouse model for MI and single cell RNA-Seq, we demonstrate that the activation of Reparative Cardiac Fibroblasts (RCFs), the CFs responsible for the healing scar, happens within the first week after MI. Interestingly, our data reveals that all CFs show high expression of the top markers genes for RCF in a specific moment, but only few of them finally evolve to an RCF transcriptomic identity. Furthermore, we describe two different molecular dynamics that could give rise to this activation and, in consequence, the appearance of definitive RCFs. Using Spatial Transcriptomics, we localized the genes related to each dynamic in different anatomical regions of the infarcted heart, but, remarkably, only one persists seven days after MI. These results highlight the existence of a specific “window of activation” of RCFs at the beginning of the ventricular remodelling process. This potential ´therapeutical window´ could allow us to regulate the size of the healing scar and, in consequence, the poor prognosis for patients that have suffered an ischemic event. 


Adrián RUIZ-VILLALBA (Málaga, Spain), Silvia C HERNANDEZ, Marina AINCIBURU, Amaia VILAS-ZORNOZA, Laura SUDUPE, Sarvide SARAI, Nuria PLANELL, Purificación RIPALDA-CEMBORÁIN, Gloria ABIZANDA, Jose Mª PÉREZ-POMARES, David GOMEZ-CABRERO, Felipe PROSPER
19:00 - 20:45 #30561 - 225. Decoding and engineering multicellular systems and circuits.
225. Decoding and engineering multicellular systems and circuits.

Reconstructing the circuits that control how cells adopt specific fates and engineering these circuits to reprogram cellular functions are major challenges in biology. I will introduce a series of experimental and computational frameworks such as “Waddington-OT”, “Raman2RNA” for reconstructing molecular dynamics over time and in live cells through single-cell multi-omics and imaging. I will introduce how we can use these approaches to decode the cellular and molecular mechanisms governing reprogramming and development. 


Shu JIAN (Boston, USA)
19:00 - 20:45 #30583 - 227. Determining the factors driving pacemaker tissue development and function in zebrafish.
227. Determining the factors driving pacemaker tissue development and function in zebrafish.

In the vertebrate heart, the leading pacemaker role is held by the sinoatrial node (SAN), which is at the origin of each heartbeat. This crucial function originates in the capacity of the pacemaker cells to spontaneously generate action potentials that propagate in an orderly manner across the surrounding cardiac tissue and eventually trigger the synchronized contraction of the heart chambers. The SAN not only displays great variability in size, shape and architecture across vertebrate species, but its complexity increases as development progresses, going from a bundle of spontaneously depolarizing cardiomyocytes to a heterogenous tissue comprising amongst others fibroblasts and cells of neural origin, additionally to specialized cardiomyocytes. How this structure comes together remains unclear, yet its conservation suggests that its assembly is driven by a set of general, elementary principles.

We have carried out single-cell RNA sequencing on cardiomyocytes of zebrafish at two days post-fertilization, the developmental stage at which pacemaker function is first observed. Having identified the cells harboring pacemaker activity, we are currently examining their transcriptomic signature to get more insight into the molecular processes leading to their establishment. Additionally, through analysis of genes differentially expressed in this specific cardiomyocyte sub-population, we hope to get better understanding of the integration of the pacemaker in cardiac function.        


Federico TESSADORI (Utrecht, The Netherlands), Olga VETH, Laurence GARRIC, Jeroen BAKKERS
19:00 - 20:45 #30619 - 229. Trajectory reconstruction identifies dysregulation of perinatal maturation programs in pluripotent stem cell-derived cardiomyocytes.
229. Trajectory reconstruction identifies dysregulation of perinatal maturation programs in pluripotent stem cell-derived cardiomyocytes.

A primary limitation in the clinical application of pluripotent stem cell derived cardiomyocytes (PSC-CMs) is the failure of these cells to achieve full functional maturity. In vivo, cardiomyocytes undergo numerous adaptive changes during perinatal maturation. By contrast, PSC-CMs fail to fully undergo these developmental processes, instead remaining arrested at an embryonic stage of maturation. To date, however, the precise mechanisms by which directed differentiation differs from endogenous development, leading to consequent PSC-CM maturation arrest, are unknown. The advent of single cell RNA-sequencing (scRNA-seq) has offered great opportunities for studying CM maturation at single cell resolution. However, perinatal cardiac scRNA-seq has been limited owing to technical difficulties in the isolation of single CMs. Here, we used our previously developed large particle fluorescence-activated cell sorting approach to generate an scRNA-seq reference of mouse in vivo CM maturation with extensive sampling of perinatal time periods. We subsequently generated isogenic embryonic stem cells and created an in vitro scRNA-seq reference of PSC-CM directed differentiation. Through trajectory reconstruction methods, we identified a perinatal maturation program in endogenous CMs that is poorly recapitulated in vitro. By comparison of our trajectories with previously published human datasets, we identified a network of nine transcription factors (TFs) whose targets are consistently dysregulated in PSC-CMs across species. Notably, we demonstrated that these TFs are only partially activated in common ex vivo approaches to engineer PSC-CM maturation. Our study represents the first direct comparison of CM maturation in vivo and in vitro at the single cell level, and can be leveraged towards improving the clinical viability of PSC-CMs.


Suraj KANNAN, Matthew MIYAMOTO, Brian LIN, Sean MURPHY (Baltimore, MD, USA), Chulan KWON
19:00 - 20:45 #30640 - 231. Cardiovascular Development Data Resource Center (CDDRC).
231. Cardiovascular Development Data Resource Center (CDDRC).

The CDDRC provides an innovative cloud-based platform to facilitate the analysis, visualization and sharing of genomic data from research in cardiac development and regeneration in several species. Leveraging institutional investments in genomics and personalized medicine, the CDDRC will facilitate dynamic and investigator-interactive cloud-based interfaces with multiple -omics datasets from humans and multiple model organisms, resulting in collaborative discoveries of the genetic and epigenetic causes of congenital heart disease (CHD). In addition, our program will have educational components in Ethical, Legal and Social Implications (ELSI) of CHD genetics and genomics, and an outreach program in CHD computational biology for students from underrepresented groups. The CDDRC will bring together researchers with a diverse range of expertise, with the goal of understanding the causes of congenital heart disease in children.

 

The CDDRC is the newest component of the NHLBI Bench-to-Bassinet (B2B) program (https://benchtobassinet.com), along with the Pediatric Cardiac Genomics Consortium (PCGC) and the Pediatric Heart Network (PHN). The goals are 1) to populate the platform with multi-omics data (single cell-omics, RNA-seq, ATAC-seq, ChIP-seq, Hi-C, etc.) from multiple organisms from the previous basic science component of the B2B, the Cardiovascular Development Consortium (CvDC), 2) to recruit new datasets from the cardiovascular community to the platform, and 3) to develop, recruit and incorporate new bioinformatics analytic and visualization tools to the platform, and 4) intersect this cloud-based platform with human CHD genetics datasets and the NHLBI BioData Catalyst (BDC) initiative.

 

At the Weinstein meeting we will be announcing Challenge Prize competitions that will fund new projects in the CDDRC from outside investigators, and a new CDDRC Fellowship program that will provide training opportunities for outside investigators in cloud-based genomics computation.

 


H Joseph YOST (Salt Lake City, USA)
19:00 - 20:45 #30492 - 233. Expanding the Clinical and Genetic Spectrum of SPEG Variants: A Pediatric Case of Dilated-Left Ventricular non-compaction Cardiomyopathy with Compound Heterozygous Variants.
233. Expanding the Clinical and Genetic Spectrum of SPEG Variants: A Pediatric Case of Dilated-Left Ventricular non-compaction Cardiomyopathy with Compound Heterozygous Variants.

Left ventricular non-compaction (LVNC) associated with dilation is characterized by the coexistence of left ventricular dilation and systolic dysfunction. Pediatric cases with dilated-LVNC have worse outcomes than those with isolated dilated cardiomyopathy and adult patients. Herein, we report a pediatric case with early-onset of dilated-LVNC.

Trio-based whole-exome sequencing revealed compound heterozygous variants in SPEG gene, involving one common de novo missense variant and one ultra-rare paternally inherited variant. Both patient's sisters, as well as his father who presented LVNC without cardiac dysfunction harbor the ultra-rare SPEG variant only.

The current study adds further evidence on the cumulative effect of SPEG variants as a mechanism of LVNC cardiomyopathy, the pleiotropic nature of this gene, and the clinical relevance of de novo mutations.


Hager JAOUADI, Hager JAOUADI (Marseille), Fedoua EL-LOUALI, Chloé WANERT, Aline CANO, Caroline OVAERT, Stéphane ZAFFRAN
19:00 - 20:45 #29491 - 235. Do Gata6 and Mib1 cooperate in aortic valve development and disease?
235. Do Gata6 and Mib1 cooperate in aortic valve development and disease?

Bicuspid aortic valve (BAV) is the most common congenital heart defect, affecting up 0.5% to 2% of the population. BAV is characterized by the presence of two leaflets instead of three symmetrical ones in normal aortic valves. BAV patients usually develop premature CAVD leading to valve stenosis that requires valve replacement. Recent studies have shown that Gata6 haploinsufficient mice show RL-type BAV phenotype, the most common in humans. Proliferation, differentiation and migration processes are affected in Gata6+/- mice giving rise to OFT defects which lead to BAV malformation. We previously showed that abrogation of the Notch regulator Mib1 causes BAV.  Gata6 and Mib1 are located in chromosome 18, and only 300Kb apart. We have generated three different mouse lines using CRISPR-Cas9, combining Gata6 nonsense mutation with different Mib1 nonsense and missense mutations to examine the possibility of a common regulation of Gata6 and Mib1 in valve morphogenesis.  Double heterozygous mice with Gata6 and Mib1 in trans configuration show reduction in BAV penetrance to 40%, compared to 70% in Gata6+/- mice. Additionally, double heterozygous mice with Gata6 and Mib1 in cis configuration show a complete suppression of the BAV phenotype. In contrast, combination of Gata6 with Notch1 mutations does not increase or reduce the 70% penetrance of BAV in double heterozygous mice, indicating that the observed BAV reduction is Mib1-dependent. Gata6 and Mib1 belong to the same topologically associating domain (TAD) and we will try to study the regulatory implications that this has and its impact in valve morphogenesis.


Rebeca PIÑEIRO-SABARÍS (Madrid, Spain), Donal MACGROGAN, José Luis DE LA POMPA
19:00 - 20:45 #30491 - 237. Identification of non-synonymous Variations in ROBO1 and GATA5 genes in a Family with Bicuspid Aortic Valve Disease.
237. Identification of non-synonymous Variations in ROBO1 and GATA5 genes in a Family with Bicuspid Aortic Valve Disease.

Bicuspid aortic valve (BAV) is the most common congenital heart defect with a high index of heritability. Patients with BAV have different clinical course and disease progression.

Herein, we report three siblings with clinically heterogeneous BAV. Their clinical presentations include moderate to severe aortic regurgitation, aortic stenosis, and ascending aortic aneurysm. Genetic investigation was carried out using Whole Exome Sequencing for the three patients.

We identified two non-synonymous variants in ROBO1 and GATA5 genes. The ROBO1: p.(Ser327Pro) variant is shared by the three BAV affected siblings. The GATA5: p.(Gln3Arg) variant is shared only by the two brothers who presented BAV and ascending aortic aneurysm. Their sister who is affected by BAV without aneurysm does not harbor the GATA5: p.(Gln3Arg) variant. Both variants were absent in the patients’ fourth brother who is clinically healthy with tricuspid aortic valve.

To our knowledge, this is the first association of ROBO1 and GATA5 variants in a familial BAV with a potential genotype/phenotype correlation. Our findings are suggestive of the implication of ROBO1 gene in BAV and GATA5: p.(Gln3Arg) variant in ascending aortic aneurysm.

Our family-based study further confirms the intrafamilial incomplete penetrance of BAV as well as the complex pattern of inheritance of the disease.


Hager JAOUADI (Marseille), Hilla GERARD, Alexis THÉRON, Gwenaelle COLLOD-BEROUD, Jean-François AVIERINOS, Stéphane ZAFFRAN
19:00 - 20:45 #30539 - 239. Efnb2 orients ventricular endocardial cells against blood flow.
239. Efnb2 orients ventricular endocardial cells against blood flow.

Cardiovascular morphogenesis involves dynamic behaviors of endocardial cells, which respond to blood flow by orchestrating cell orientation, proliferation, migration and cell shapes. How endocardial cells of the zebrafish heart influence blood flow-responsive behaviors such as cell orientation during cardiac chamber development or ingression into the cardiac jelly during valvulogenesis remains unclear. Here, we show that blood flow induces the expression of the transmembrane ligand Efnb2a specifically within the ventricular endocardium of the nascent heart tube and that its activity is necessary and sufficient to reorient these endocardial cells against blood flow. During valvulogenesis, Efnb2a instructs the reorientation of atrioventricular cushion endocardial cells out of the plane of the endocardium prior to their ingression into the cardiac jelly. In the absence of blood flow, forced and clonal overexpression of Efnb2a within endocardial cells is sufficient to trigger their reorientation, thus establishing Efnb2a as a cell-autonomous morphogenetic regulator that mediates endocardial cell orientation in response to blood flow. 


Anusha SATHYANARAYANAN (Potsdam, Germany), Federica FONTANA, Elke OBER, Salim ABDELILAH-SEYFRIED
19:00 - 20:45 #30573 - 241. A SOX17-PDGFB Signaling Axis Regulates Aortic Root Development.
241. A SOX17-PDGFB Signaling Axis Regulates Aortic Root Development.

Developmental etiologies causing complex congenital aortic root abnormalities are unknown. Here we show that deletion of Sox17 in aortic root endothelium in mice causes underdeveloped aortic root leading to a bicuspid aortic valve due to the absence of non-coronary leaflet, as well as mispositioned left coronary ostium. The respective defects are associated with reduced proliferation of non-coronary leaflet mesenchyme and aortic root smooth muscle derived from the second heart field progenitor cells. Mechanistically, SOX17 occupies a Pdgfb transcriptional enhancer to promote its transcription, whereas Sox17 deletion inhibits the endothelial Pdgfb transcription and PDGFB growth signaling to the non-coronary leaflet mesenchyme. Restoration of PDGFB in aortic root endothelium rescues the non-coronary leaflet and left coronary ostium defects in Sox17 nulls. Altogether, these observations support a SOX17-PDGFB axis underlying aortic root development that is critical for aortic valve and coronary ostium patterning, thereby informing a potential shared disease mechanism for concurrent anomalous aortic valve and coronary arteries.  


Pengfei LU, Bingruo WU, Yidong WANG, Yukiko SUGI, Jonathan BUTCHER, Deyou ZHENG, Bin ZHOU (New York, USA)
19:00 - 20:45 #30596 - 243. Pax9 Regulation of Extracellular Matrix Production in the Pharyngeal Endoderm.
243. Pax9 Regulation of Extracellular Matrix Production in the Pharyngeal Endoderm.

The global burden created by congenital heart defects (CHD) is firmly established. Whilst the cause of approximately 80% of CHD is unknown, it is thought likely that there is a tightly regulated genetic network that exists to shape development in the great vessels of the heart. These are derived from the pharyngeal arch arteries (PAAs) which form sequentially in mid-embryogenesis, before remodelling to form the mature vessels. One gene implicated in this network, Paired-box gene 9 (Pax9), plays a key role in developmental processes through functional interaction with Tbx1 in the pharyngeal endoderm for 4th PAA development. The extracellular matrix (ECM) exists as a 3D domain that provides structural and biochemical support to surrounding cells and has been found to facilitate the migration of neural crest cells (NCCs) into the pharyngeal arches and influence their subsequent differentiation into smooth muscle cells to support the developing PAAs. Previous studies have demonstrated that cardiovascular defects are present in mice mutated for ECM genes. RNA-seq data has identified ECM organisation as a significantly differentially expressed signalling pathway between Pax9+/+ and Pax9-/- embryos, highlighting the genes involved as candidates for further analysis. Furthermore, immunostaining of key ECM proteins has shown variable expression between Pax9 control and mutant embryos. Studies are underway to validate the expression of these key ECM genes and uncover the mechanisms by which they interact with Pax9 to increase the understanding of how these genetic interactions influence migratory NCC behaviour and the overall effect this may have on cardiovascular morphogenesis. 


Rebecca STEELE (Newcastle upon Tyne, United Kingdom), Helen PHILLIPS, Simon BAMFORTH
19:00 - 20:45 #30600 - 245. Multiple pkd and piezo gene family members are required for atrioventricular valve formation.
245. Multiple pkd and piezo gene family members are required for atrioventricular valve formation.

Cardiac valves ensure unidirectional blood flow through the heart, and altering their function can result in heart failure.  Flow sensing via wall shear stress and wall stretching through the action of mechanosensors can modulate cardiac valve formation.  However, the identity and precise role of the key mechanosensors and their effectors remains mostly unknown.  Here, we genetically dissect the role of Pkd1a and other mechanosensors in atrioventricular (AV) valve formation in zebrafish and identify a role for several pkd and piezo gene family members in this process.  We show that Pkd1a, together with Pkd2, Pkd1l1, and Piezo2a, promotes AV valve elongation and cardiac morphogenesis.  Mechanistically, Pkd1a, Pkd2, and Pkd1l1 all repress the expression of klf2a, klf2b, and egr1 transcription factor genes implicated in AV valve development.  Furthermore, we find that the calcium-dependent protein kinase Camk2g is required downstream of Pkd function to repress klf2a expression.  Altogether, these data identify and dissect the role of new mechanosensors required for AV valve formation, thereby broadening our understanding of cardiac valvulogenesis.


Thomas JUAN (Bad Nauheim, Germany), Barbara CARDOSO, Soeun LIM, Violette CHARTEAU, Agatha RIBEIRO DA SILVA, Didier Y.r. STAINIER
19:00 - 20:45 #30606 - 247. Generation of valvular interstitial cells from human pluripotent stem cell-derived endocardial cells.
247. Generation of valvular interstitial cells from human pluripotent stem cell-derived endocardial cells.

Heart valve defects are present in ~1/3 of congenital heart disease cases, the most common form of human birth defects. Many affected individuals will require valve replacement. Generation of various valve cell populations in vitro from human pluripotent stem cells (hPSC) would provide unique opportunities to study human valve disease as well as  engineer biological valves. During embryonic development the cell populations comprising heart valves derive from endocardium, a specialized endothelial cell population that lines the chambers of the heart and  induces the formation of the first functional population of cardiomyocytes, known as trabecular cardiomyocytes. To access human endocardial cells, we developed a protocol that promotes the generation of NKX2-5+CD31+ endothelial cells from hPSC-derived cardiovascular progenitors by manipulating BMP10 signaling. These cells express the cohort of genes that identifies the endocardial lineage in vivo and show a high level of transcriptional similarity to primary human fetal endocardium. Furthermore, these hPSC-derived  NKX2-5+CD31+ endocardial-like cells display the ability to induce a trabecular fate following co-culture with target cardiomyocytes, and the capacity to undergo EndoMT to give rise to mesenchymal cells that express markers of valvular interstitial cells (VICs). Further culturing of these VIC-like cells resulted in their segregation into two subpopulations secreting different amounts of collagens and proteoglycans/glycosaminoglycans, recapitulating the heterogeneity observed in the stratified extracellular matrix of normal valves. In summary, the findings presented in this report describe a method for the derivation of valvular interstitial cells from hPSCs.


Alexander MIKRYUKOV (Toronto, Canada), Amine MAZINE, Neda LATIFI, Bei WEI, Donghe YANG, Yifei MIAO, Mingxia GU, Craig SIMMONS, Gordon KELLER
19:00 - 20:45 #30631 - 249. The temporal progression of aortic valve pathogenesis in a mouse model of osteogenesis imperfecta.
249. The temporal progression of aortic valve pathogenesis in a mouse model of osteogenesis imperfecta.

The organization of extracellular matrix (ECM) components, including collagens, proteoglycans, and elastins, into the fibrosa, spongiosa, and ventricularis layers respectively, is essential for maintaining structure and function of heart valves. Mutations in ECM components cause connective tissue disorders, including osteogenesis imperfecta (OI), and debilitating heart valve dysfunction in these patients is common. Currently, treatment is limited to surgical valve repair or replacement, causing insuperable complications for many patients, emphasizing a critical need for developing alternative approaches. This study models connective tissue disorders in mice to investigate valve pathogenesis, with the goal of developing more effective, mechanistic-based therapies.

Mice with a frameshift mutation in the pro-alpha2 chain of collagenI (col1a2) serve as a model of OI Murine (OIM). By histology, aortic valve (AoV) cusps from OIM-/- mice show significant thickening and myxomatous features, including increased proteoglycan deposition and disorganization of the ECM, beginning at 3 months of age that worsen over time. Ultrastructural electron microscopy of AoV cusps reveal a trend towards decreased area and diameter of collagen fiber cross sections in the OIM-/- by 9 months. However, we observe no functional defects under echocardiography until 12-14 months, when 67% of OIM-/- mice exhibit AoV regurgitation and/or left ventricle enlargement, or AoV stenosis. Mass spectrometry of decellularized AoV isolates collected at 3 months of age confirmed decreased ColIa2 in OIM-/- mice, and identified significant changes in the overall ECM niche as a result of the col1a2 mutation that likely contribute to the observed valvular phenotypes.

Together these studies provide mechanistic insights into the temporal pathogenesis of valve dysfunction associated with human connective tissue disorders.


Kaitlyn THATCHER (Milwaukee, USA), Carol MATTERN, Yifei LIU, Michael MCDERMOTT, Jessica FULTON, Christopher K. BREUER, Brian R. HOFFMANN, Joy LINCOLN
19:00 - 20:45 #30646 - 251. Targeted valve disruption during avian cardiac morphogenesis.
251. Targeted valve disruption during avian cardiac morphogenesis.

During cardiac morphogenesis, blood exits the ventricle through the outflow tract and pharyngeal arch arteries (PAAs). Hemodynamics plays a vital role in the maturation of PAAs into the great vessels of the heart. Disruption of established flow patterns during critical windows of development produces a range of cardiac malformations that drastically alter function of the mature heart. Malformation of the great vessels often occur in tandem with outflow tract valve malformations. However, the timing of which defect precedes the other and the role of hemodynamics in disease propagation remains poorly understood. Here, we alter intracardiac flow patterns through early valve disruption. With the chick embryo annimal model we preform highly targeted non-invasive microsurgeries on prevalvular cushions and detail the effects on subsequent PAA morphogenesis. Through the use of two-photon microscopy guided femtosecond laser pulses, we nucleate and control the growth of microbubbles within the developing outflow tract, focally displacing early valve tissue until a permanent ablation that prevents closing of the valve is obtained. We monitor hemodynamic changes through Doppler ultrasound and detail subsequent structural changes using nano-computed tomography. Proximal cushion ablations performed at HH18 (day 3) show instantaneous flow changes that lead to structural changes within 24 hours. We follow these morphological changes in 24-hour increments through HH31 (day 7), detailing arch specific changes in length, curvature and cross-sectional area as well as abnormal aortic sac connections. With these tools, we directly link focal obstructions of the outflow tract to abnormal PAA remodeling that results from local tissue and hemodynamic malformations.


Stephanine LINDSEY (La Jolla, USA)
19:00 - 20:45 #30658 - 253. Accurate quantitative expression analysis and reporting requires PCR efficiency correction.
253. Accurate quantitative expression analysis and reporting requires PCR efficiency correction.

Quantitative PCR (qPCR) aims to measure DNA or RNA concentrations in diagnostic and biological samples based on the quantification cycle (Cq) value observed in the amplification curves. Results of qPCR experiments are mostly reported as Cq, ΔCq, or ΔΔCq values and thus assume that all assays are 100% efficient.

The large variation in reported Cq values among laboratories can be explained readily by differences in qPCR machines, amplification protocols and the setting of the quantification threshold, at the start or at the end of the exponential phase of the amplification curve.

Qualitatively, a reaction that shows specific amplification should be deemed positive, regardless of the observed Cq. Quantitatively, it should be taken into account that the observed Cq is highly dependent on the amplification efficiency. Because this efficiency can vary among targets and samples, meaningful diagnostics or biological interpretation of target quantity and relative gene expression values require that the actual efficiency of the PCR is used in the calculations.

PCR efficiency is frequently derived from standard curves, but this approach is affected by serial dilution errors and hampered by properties of the standard and the diluent. This affects accurate quantification of clinical and biological samples in diagnostic applications and collected in challenging conditions. Determining the mean PCR efficiency from the individual amplification curves per assay avoids these biases in relative quantification.

To take the analysis to the level of absolute quantification and obtain unbiased efficiency-corrected results, we recommend quantification with a single calibrator with known target concentration and efficiency values determined from the amplification curves of the reactions of this calibrator and the unknown samples.

Using this efficiency-corrected approach will help to remove inter-laboratory variability in reported expression levels of genes within the same biological context.


Maurice VAN DEN HOFF (Amsterdam, The Netherlands), Quinn GUNST, Jan RUIJTER
Saturday 14 May
08:45

"Saturday 14 May"

Added to your list of favorites
Deleted from your list of favorites
PS7
08:45 - 10:45

Session VII
Regeneration II - Role of Endothelium and Fibroblasts

Moderators: Chris JOPLING (PI) (Montpellier, France), Eldad TZAHOR (eldad.tzahor@weizmann.ac.il) (Rehovot, Israel)
08:45 - 10:45 #30528 - 032. An injury responsive mmp14b enhancer promotes heart regeneration in zebrafish.
032. An injury responsive mmp14b enhancer promotes heart regeneration in zebrafish.

Mammals have limited ability to regenerate damaged cardiac muscle following injury. Instead, the injured area is replaced by fibrotic scar tissue, leading to irreversible cardiac remodeling and organ failure. Conversely, zebrafish possess an extraordinary capability for heart regeneration. Following injury, zebrafish cardiomyocytes proliferate to generate new myocardium. In addition, other cells in the heart, including endothelial cells, are activated in response to injury and provide regenerative cues to cardiomyocytes. However, heart injury results in the activation of developmental and regeneration-specific gene regulatory networks, the transcriptional basis of endothelial gene activation during regeneration, and the gene products responsible for promoting efficient heart regeneration in zebrafish have not been fully delineated. Here, we performed ATAC-seq on zebrafish regenerating hearts to identify active endothelial enhancers during development and regeneration. Among the many responsive enhancers, we identified a cis-acting element in the intronic region of mmp14b, a known effector of angiogenesis. This element activated expression in endothelial cells in regenerating zebrafish and neonatal mice. mmp14b enhancer activation in response to injury depends on a consensus TEAD-binding motif. Moreover, we found that deletion of mmp14b or the mmp14b regeneration enhancer identified in these studies abrogated efficient cardiac regeneration, demonstrating an essential role for mmp14b in heart regeneration. These findings also reveal shared transcriptional circuitry between fish and mammals for endothelial activation following heart injury, providing a potential therapeutic platform for enhancing cardiac regeneration


Ivana ZLATANOVA (San Francisco, USA), Pauline COLOMBIER, Tanvi SINHA, Barbara CELONA, Shan-Mei XU, Stefan MATERNA, Kenneth POSS, Brian BLACK
08:45 - 10:45 #30440 - 033. The novel regenerative capacity of embryonic endothelium rescues congenital heart defects.
033. The novel regenerative capacity of embryonic endothelium rescues congenital heart defects.

Defective formation or remodeling of the Pharyngeal Arch Arteries (PAAs) result in congenital heart disease phenotypes, such as those seen in 22q11 deletion syndrome. We have previously shown that the PAA endothelium originates in the Second Heart Field (SHF). However, whether SHF-derived endothelial cells (ECs) are required for PAA formation, and the mechanisms regulating SHF progenitor contribution to the PAAs are unclear. To address these questions, we crossed VEGFR2+/-;Mef2c-AHF-Cre and VEGFR2f/f mice, to conditionally ablate VEGFR2 expression in the SHF and inhibit SHF progenitor contribution to the PAA endothelium. Although the loss of VEGFR2 in the SHF abrogated the contribution of the SHF to the PAA endothelium, ECs from a compensatory source migrated into the pharyngeal arches, resulting in the eventual rescue of PAA formation and aortic arch morphogenesis.  The extent of the rescue depended on the proportion of SHF-derived ECs in the pharyngeal arches. In embryos with at least one functional copy of the VEGFR2 in the SHF, the contribution of the SHF-derived ECs to the PAAs was on average 41%. In these cases, the rescue by the alternative EC source was complete. To investigate the source of the compensatory endothelium, we examined the arterial-venous identity and fate of the ECs within the pharyngeal arches. We found that embryos with defective SHF-derived EC contribution had an increased number of venous ECs compared with controls. Altogether, our studies demonstrate that: 1) SHF-derived ECs are required for PAA formation and remodeling, 2) VEGFR2 is essential for these processes, 3) a threshold number of SHF-derived ECs is necessary for proper PAA formation, and 4) the loss of SHF-derived ECs in pharyngeal arches can be compensated by angiogenesis from a venous vascular source. Understanding the mechanisms regulating EC compensation can lead to the prevention or treatments of congenital heart disease-associated conditions such as 22q11 deletion syndrome.


Annjosette RAMIREZ, Sophie ASTROF (Newark, USA)
08:45 - 10:45 #29784 - 034. Sequential Bmp2-Cxcr4 signaling in the cell fate trajectory from endocardium to coronary artery during heart development and regeneration.
034. Sequential Bmp2-Cxcr4 signaling in the cell fate trajectory from endocardium to coronary artery during heart development and regeneration.

Arterial occlusion deprives billions of cardiac myocytes of oxygenated blood, damaging cardiac muscle and inciting scar formation, which ultimately impairs cardiac function. We aim to discover the developmental pathways that activate coronary vessel (CV) differentiation from their progenitors and establish whether reactivating these pathways can stimulate significant coronary genesis during cardiac injury. The endocardium is a progenitor source for CV endothelial cells (EC), but the signaling pathways driving the endocardium-to-coronary cell fate are unknown. Combining temporally regulated lineage tracing and single-cell RNA sequencing (scRNAseq), we demonstrated that the endocardial-to-coronary vessel transition is strictly regulated in time and space and correlates with Bmp2 expression. Time-specific lineage labeling revealed that endocardial cells largely transition to CVs at the onset of septal development. Transitioning cells were detected in scRANseq data at e12 where they downregulated endocardial markers and upregulated BMP2 and Vegfa-dependent genes. This cell fate transition and BMP2 expression in coronary ECs was absent in scRNAseq data at e17.5. Exogenous administration of Bmp2 and VegfA in an in vitro coronary angiogenesis assay induced a large number of endocardial-derived ECs. In vivo, upon coronary artery ligation, neonatal mice treated with adeno-associated virus expressing Bmp2 (AAV-Bmp2) exhibited a dramatic increase in newly formed endocardium-derived CVs at the injury site.  Finally, analysis of Cxcl12 and Cxcr4 knockout hearts showed that this signaling pathway functions after the endocardial-to-coronary transition to mediate artery differentiation. Our data shed light on the molecular mechanism underlying the endocardial to CVs transition and how endocardial-derived ECs contribute to coronary arterial formation, opening a new potential therapeutic target that could promote revascularization of the injured heart.


Gaetano D'AMATO (Stanford, USA), Ragini PHANSALKAR, Jeffrey A. NAFTALY, Pamela E. RIOS CORONADO, Dale O. COWLEY, Kelsey E. QUINN, Bikram SHARMA, Kathleen M. CARON, Alessandra Vigilante VIGILANTE, Kristy RED-HORSE
08:45 - 10:45 #29484 - 035. Origin and function of activated fibroblast states during zebrafish heart regeneration.
035. Origin and function of activated fibroblast states during zebrafish heart regeneration.

The adult zebrafish heart has a high capacity for regeneration after injury. While several pro-regenerative factors have been identified, the cell types orchestrating heart regeneration remain largely elusive. To overcome this limitation, we dissected the diversity of activated cell states in the regenerating zebrafish heart based on single-cell transcriptomics and spatiotemporal analysis. We discovered a dramatic induction of several cell states with fibroblast characteristics upon injury, and we validated the pro-regenerative function of transient collagen 12 expressing fibroblasts. To understand the cascade of events leading to heart regeneration, we determined the origin of these cell states by high-throughput lineage tracing. We found that activated fibroblasts are derived from two separate sources, the epicardium, and the endocardium. Mechanistically, we determined Wnt signalling as a key regulator of the endocardial regenerative response. In summary, our results uncover specialized activated fibroblast cell states as major drivers of heart regeneration, thereby opening up new possibilities to modulate the regenerative capacity of the vertebrate heart.


Sara LELEK-GRESKOVIC (Berlin, Germany), Bo HU, Baastian SPANJAARD, Hadil EL-SAMMAK, Mariana GUEDES SIMOES, Janita MINTCHEVA, Aliee HANANEH, Fabian THEIS, Didier STAINIER, Jan Philipp JUNKER, Daniela PANAKOVA
08:45 - 10:45 #30423 - 036. Targeting the autocrine loop of cardiac myofibroblasts to fight fibrosis.
036. Targeting the autocrine loop of cardiac myofibroblasts to fight fibrosis.

Cardiac fibrosis remains a major challenge in the field of cardiac repair, as there is no effective therapy for the prevention or reversion of cardiac scarring. We asked what are the underlying molecular principles in the development and maintenance of cardiac fibrosis. Recently, we have established an in-silico model for a minimal macrophage (Mf)-myofibroblast (mF) cell-circuit that orchestrates tissue repair and fibrosis. We set out to explore this cell-circuit in the setting of acute myocardial infarction (AMI) that results in sustained fibrosis. By employing a series of unbiased, high-throughput techniques and mathematical modeling we demonstrated that in a regenerative, neonatal model of AMI, both Mf and mF numbers rise and then return to baseline levels. This resembles the theoretical cellular trend for tissue healing. In contrast, in a non-regenerative model of AMI in adult mice, while both Mf and mF rise together for a prolonged period, only Mf numbers return to pre-injury levels. This resembles “cold fibrosis”, a theoretical point in which mF are self-sustained and Mf are depleted. Intriguingly, under cold fibrosis, Mf regain their homeostatic functions, while cardiac fibroblasts acquire a de-novo pro-fibrotic phenotype which persists within the mature scar. In-silico simulation of factors controlling the cell-circuit revealed that the most prominent factor governing the regenerative window is the mF population growth rate, controlled by an autocrine loop. NicheNet analysis predicted a novel cardiac mF autocrine growth factor, Timp1, that drives cardiac fibrosis. Accordingly, In-vivo neutralization of Timp1 by polyclonal antibodies reduced cardiac fibrosis following AMI in adult mice. These findings suggest a central role for the mF autocrine signaling loop in the development of cardiac fibrosis. Such insights may be utilized in future pursuit of effective therapy for cardiac fibrosis.


Shoval MIYARA (Rehovot, Israel, Israel), Miri ADLER, Avi MAYO, Yalin DIVINSKY, Kfir UMANSKY, Jacob ELKAHAL, Avraham SHAKKED, Lingling ZHANG, Alexander GENZELINAKH, Gal AVIEL, David KAIN, Daria LENDENGOLTS, Ruslan MEDZHITOV, Uri ALON, Eldad TZAHOR
08:45 - 10:45 #30449 - 037. Activation of nkx2.5 transcriptional program is required for adult myocardial repair.
037. Activation of nkx2.5 transcriptional program is required for adult myocardial repair.

The cardiac developmental network has been associated with myocardial regenerative potential. However, the embryonic signals triggered following injury have yet to be fully elucidated. Nkx2.5 is a key causative transcription factor associated with human congenital heart disease and one of the earliest markers of cardiac progenitors, thus it serves as a promising candidate. Our previous studies illustrate an early requirement for nkx2.5 in maintenance of ventricular dentity during embryogenesis, however, its molecular function in the adult zebrafish heart has not been explored. Here, we show that cardiac-specific RNA-sequencing studies reveal a disrupted embryonic Nkx2.5 transcriptional profile in the adult Nkx2.5 loss-of-function myocardium. Furthermore, nkx2.5-/- fish exhibit an impaired ability to recover following ventricular apex amputation as illustrated by retention of fibrin and collagen in the injured area. Complex network analyses illuminate that Nkx2.5 is required to mount a proliferative response for cardiomyocyte renewal and to provoke proteolytic pathways necessary for sarcomere disassembly. Moreover, direct Nkx2.5 targets embedded in these distinct gene regulatory modules coordinate appropriate, multi-faceted injury responses. Altogether, our findings support a previously unrecognized, Nkx2.5-dependent regenerative circuit that invokes myocardial proliferation and dedifferentiation to ensure effective regeneration in the teleost heart.


Carmen DE SENA TOMÁS (New York, Portugal), Angelika G. ALEMAN, Caitlin FORD, Akriti VARSHNEY, Di YAO, Jamie K. HARRINGTON, Leonor SAÚDE, Mirana RAMIALISON, Kimara L. TARGOFF
Auditorium
10:45 Break
11:15

"Saturday 14 May"

Added to your list of favorites
Deleted from your list of favorites
PS8
11:15 - 13:00

Session VIII
Cardiac Maturation

Moderators: Gregor ANDELFINGER (Principal Investigator) (Montréal, Canada), Nicole DUBOIS (Associate Professor) (New York, USA)
11:15 - 13:00 #30572 - 038. Investigating the role of ACTN2 enhancer in cardiomyocyte maturation.
038. Investigating the role of ACTN2 enhancer in cardiomyocyte maturation.

Cardiomyocyte (CM) maturation is a dynamic process that occurs from mid-gestation to early postnatal stages and is accompanied by structural, functional, and metabolic changes. Recent studies indicate that ACTN2 might be important for CM maturation and ACTN2 mutations, typically result in cardiomyopathies. However, the link between disrupted CM maturation and heart disease is unknown. Through largescale GWAS analysis, we recently identified a heart failure (HF) associated variant within an evolutionary conserved ACTN2 enhancer region. Engineered hPSC-CM carrying an ACTN2 enhancer deletion, showed mildly reduced ACTN2 expression, and CMs developed myofibrillar disarray, hypertrophy, and suppressed calcium transients and mechanical force. Moreover, mitochondrial respiration and oxidative phosphorylation was reduced and glycolysis was increased. Consistently, single cell transcriptomic analysis suggested disrupted CM maturation. Manipulation of ACTN2 enhancer using a novel CRISPRa line, resulted in more adult-like hPSC-CMs. Additional analyses using ChIP and CRISPRi suggested temporal regulation of ACTN2 expression through enhancer binding both in vitro and in vivo. Interestingly, heterozygous ACTN2 enhancer KO mice, developed hypertrophy, reduced cardiac function, disrupted sarcomeres, and increased glycolysis with upregulation of insulin/PI3K/mTOR signaling pathway suggestive of disrupted in vivo maturation. Our results demonstrate that the conserved and clinically relevant ACTN2 enhancer can regulate CM maturation, and can be leveraged to promote hPSC-CM maturation and study the association between myocyte maturation and HF.


Myo HTET, Marios ARVANITIS, Brian LIN, Harshi GANGRADE, Sean MURPHY, Elaine CHEN, Renjun ZHU, Asimina ZOITOU, Chulan KWON, Emmanouil TAMPAKAKIS (Baltimore, USA)
11:15 - 13:00 #30592 - 039. LRRFIP2 modulates the response to hypoxia during embryonic cardiogenesis.
039. LRRFIP2 modulates the response to hypoxia during embryonic cardiogenesis.

Oxygen is crucial for appropriate embryonic and fetal development, including cardiogenesis. The heart is the first organ formed in the embryo and is required to provide oxygen and nutrients to all cells in the body. Embryonic cardiogenesis is a complex process finely regulated and prone to congenital malformations. It takes place in a hypoxic environment that activates the HIF-1a signaling pathway which mediates cellular and systemic adaptations to low oxygen levels. Since inhibition or overactivation of the HIF-1a signaling pathway in the myocardium lead to severe cardiac malformations and embryonic lethality, it is important that the cellular response to hypoxia be precisely regulated. While many gene regulatory networks involved in embryonic cardiogenesis have been characterized in detail, the modulation of the response of cardiomyocytes (CM) to hypoxia has remained less studied. 

In vivo and Ex vivo approaches were used to investigate the role of LRRFIP2 during cardiogenesis and to determine the cell autonomous contribution to the phenotype. Wild type and Mutant embryos were generated by crossing Lrrfip2+/- mice and then analyzed.  

We identified LRRFIP2 as a novel negative cofactor of HIF-1a. Indeed, we showed that the absence of Lrrfip2 expression in a mouse KI model led to an enhance of many HIF-1a target genes including Aldoc, Bnip3 and Ndufa4l2 in embryonic CM during developmentOveractivation of HIF-1a in Lrrfip2-null mice led to structural and functional mitochondrial dysfunction, a decreased ROS levels and ultimately to a precocious maturation of CM. 

Overall, these defects led to the formation of a smaller heart unable to supply sufficient oxygen to the embryo and ultimately to severe hypoxia and early lethality.  


Laura BEN DRISS (Paris), Christophe HOUBRON, Florian BRITTO, Philippe DAUBAS, Pascal MAIRE
11:15 - 13:00 #30625 - 040. Abnormal nucleotide metabolism in the embryonic heart of diabetic pregnancy causes mitochondrial dysfunction via AMPK.
040. Abnormal nucleotide metabolism in the embryonic heart of diabetic pregnancy causes mitochondrial dysfunction via AMPK.

Glucose is central to the metabolic environment of the embryonic heart, and hyperglycemia is the most common non-genetic cause of heart anomalies.  Using chemically defined in vitro induction of hPSC-derived cardiomyocytes, we previously found that a high level of glucose negatively impacts the maturation of cardiomyocytes through excess nucleotide biosynthesis.  Despite the mounting evidence, little is known about the impact of high glucose on the in vivo formation of embryonic organs.  Here, we present the first comparative in vivo analysis of the metabolic state of over 500 samples of embryonic organs including the heart and three other organs (liver, brain, and placenta) using a diabetic mouse model at mid-to-late gestational stages. Consistent with previous reports, metabolomics analysis revealed that the fetal heart of diabetic pregnancies is more glycolytic than that of normal pregnancies throughout the various stages. scRNA-seq showed that glycolytic enzymes were downregulated in the cardiomyocytes, suggesting a possible feedback downregulation due to metabolic changes. Interestingly, di- and tri-phosphate nucleotides are markedly elevated in the embryonic heart at later stages of diabetic pregnancy, resulting in a significant reduction in the AMP-to-ATP ratio.  Accordingly, AMPKɑ was found less phosphorylated in the fetal heart of diabetic pregnancy.  AMPK signaling is an evolutionarily conserved pathway that maintains cellular catabolism and mitochondrial homeostasis.  While there was no change in mitochondrial content, mitochondrial genes (Ppargc1a, Ulk1, Pink1, etc.) were downregulated in the fetal hearts of diabetic pregnancy.  Furthermore, the oxygen consumption reserve was significantly lower in the fetal heart of diabetic pregnancy. These data suggest that a disproportionate nucleotide pool due to excess glucose leads to mitochondrial dysfunction via AMPK signaling in the late embryonic heart development of diabetic pregnancy.


Haruko NAKANO (Los Angeles, USA), Cesar PEREZ-RAMIREZ, Naofumi KAWAHIRA, Jennifer THOMPSON, Heather CHRISTOFK, Atsushi NAKANO
11:15 - 13:00 #30615 - 041. Vangl2-dependent changes in nuclear mechanosensing regulate YAP/TAZ signaling in heart muscle development.
041. Vangl2-dependent changes in nuclear mechanosensing regulate YAP/TAZ signaling in heart muscle development.

The nucleus acts as a main mechanosensing organelle and its deformations in response to mechanical stimuli have been associated with the modulation of gene expression. During heart formation, tissue tension forces drive remodeling of the linear heart tube (LHT) to the looped and chambered heart. How the tissue tension forces generated during this process affect the nuclear mechanosensing and what signaling pathways are involved in its regulation remain unresolved. Here, we described the major changes in nuclear morphology during the LHT remodeling in both, zebrafish and mouse models. We found morphogenetic Planar Cell Polarity (PCP) signaling directs nuclear mechanosensing of the developing cardiomyocytes through the function of its core component, Vangl2. We discovered the Vangl2-dependent nuclear mechanosensing is linked to transcriptional changes in gene programs regulating muscle differentiation. Functionally, the perturbations in Vangl2-dependent signaling led to impaired mechanical coupling, muscle contractility, and reduced cardiac output in zebrafish as well as impaired cardiomyocyte differentiation in human iPS-CM model. At the molecular level, we linked the activity of Vangl2 to YAP/TAZ nuclear translocation. Altogether, we discovered Vang2/YAP/TAZ signaling axis contributes to muscle differentiation and maturation.


Anne MERKS, Sara LELEK, Kevin MÉNDEZ-ACEVEDO, Laura C. ZELARAYÁN, Daniela PANAKOVA (Berlin, Germany)
11:15 - 13:00 #30560 - 042. Slit signaling is required for mammalian cardiomyocyte cytokinesis.
042. Slit signaling is required for mammalian cardiomyocyte cytokinesis.

Cardiomyocyte (CM) polyploidy is prevalent across mammals.  Shortly after birth, rodent CMs fail cytokinesis and become multinucleated.  This transition coincides with CM cell cycle exit and is associated with the poor regenerative potential of the heart.  However, the regulatory mechanisms of CM polyploidization remain largely unknown.  Using primary postnatal CM cultures as a model, our previous work uncovered a key role for the cardiac extracellular matrix and identified specific matrisome proteins in modulating CM polyploidy.  One of the candidates, SLIT2, is primarily a secreted protein that is important for axon guidance, angiogenesis, and embryonic heart development; how it regulates cell division is currently unknown.  Intriguingly, in addition to expression in cardiac fibroblasts, we found that Slit2 and its receptor Robo1 are expressed in a subset of mouse CMs, particularly the mononuclear population at postnatal day 7, suggestive of a cell-autonomous function.  Functionally, Slit2 knockdown significantly promoted primary CM polyploidization, which could be rescued by recombinant SLIT2.  Furthermore, injection of recombinant SLIT2 was able to promote CM cytokinesis in postnatal mice.  Unexpectedly, we observed that knockdown of Robo1, 2, and 4 (major receptors of SLIT) do not affect primary CM polyploidization, suggesting that Robo-independent signaling is required for CM cytokinesis.  To elucidate the underlying mechanisms, we analyzed the transcriptome of Slit2-knockdown primary postnatal CMs.  We found that Cep55, a centrosome- and midbody-associated protein, is downregulated upon the loss of Slit2 expression.  Consistent with this observation, knockdown of Cep55 significantly inhibited cytokinesis in primary CMs.  Altogether, we found that SLIT2 is required for postnatal CM cytokinesis, potentially through the regulation of Cep55.  These findings further support a key role for extracellular matrix proteins in CM ploidy regulation.


Chi-Chung WU (Mannheim, Germany), Susann BRUCHE, Sylvia JERATSCH, Johannes GRAUMANN, Stefan GÜNTHER, Mathilda MOMMERSTEEG, Didier STAINIER
Auditorium
13:00 Lunch
14:00

"Saturday 14 May"

Added to your list of favorites
Deleted from your list of favorites
BM
14:00 - 14:15

Business meeting
10 minute presentations:

14:00 - 14:15 Fondation Lefoulon Delalande. Margaret BUCKINGHAM (Honorary Professor) (Speaker, PARIS, France)
14:00 - 14:15 European Society of Cardiology Working Group on Development Anatomy and Pathology. Maurice VAN DEN HOFF (Associate Professor) (Speaker, Amsterdam, The Netherlands)
14:00 - 14:15 Cardiovascular Development Data Resource Center. H Joseph YOST (Vice Chair for Basic Sciences) (Speaker, Salt Lake City, USA)
Auditorium
15:15

"Saturday 14 May"

Added to your list of favorites
Deleted from your list of favorites
KS2
15:15 - 16:00

Keynote speaker II

Chairperson: Stéphane ZAFFRAN (Principal Investigator) (Chairperson, Marseille, France)
Speaker: Alfonso MARTINEZ ARIAS (ICREA Research Professor) (Speaker, Barcelona, Spain)
Auditorium
16:00 Break
16:30

"Saturday 14 May"

Added to your list of favorites
Deleted from your list of favorites
PS9
16:30 - 18:15

Session IX
Valves and Vessels

Moderators: Anabela BENSIMON-BRITO (Group leader) (Marseille, France), Joy LINCOLN (Principal Investigator/Faculty) (Milwaukee, USA)
16:30 - 18:15 #30446 - 043. Cell-ECM interaction regulate symmetrical allocation of endothelial progenitors of the pharyngeal arch arteries.
043. Cell-ECM interaction regulate symmetrical allocation of endothelial progenitors of the pharyngeal arch arteries.

 The endothelium of the 3rd, 4th, and 6th pharyngeal arch arteries (PAAs) arises from Isl1-derived progenitors, which remodel into bilaterally-symmetrical PAAs by embryonic day E10.5.  PAAs 3-6 then undergo asymmetric remodeling giving rise to the aortic arch and its major branches. Aberration during these steps of development lead to sever congenital heart defects, emphasizing the need for understand the mechanisms of PAA formation and remodeling.

It is often observed that in mutant embryos with aberrant PAA formation, PAA development is disrupted asymmetrically, with one side affected more than the other. Furthermore, either the left or right side can be affected stochastically. Mutations of integrin-ɑ5 in the Isl1 lineage belong to such a category of mutations. To investigate mechanisms, we analyzed the pharyngeal mesenchyme of control and integrin-ɑ5flox/-;Isl1Cre mutant embryos at E9.5, before the vascular plexus remodels into patent PAAs. In control embryos, the vascular plexus was symmetrically distributed between the left and right sides. Additionally, the numbers of Isl1-derived cells on each side of the pharyngeal mesenchyme were statistically equal. In integrin-ɑ5flox/-;Isl1Cre embryos, the vascular plexus formed asymmetrically, and the allocation of Isl1-derived cells was no longer symmetrical between the left and right pharyngeal mesenchyme. Disruption in the ECM structure has been shown to allow cells to aberrantly migrate contralaterally. Preliminary data show disruption of Laminin-gamma1 expression in integrin ɑ5flox/-;Isl1Cre mutants, suggesting that integrin-ɑ5 signaling in the Isl1-lienage regulates ECM integrity needed for the symmetrical allocation of Isl1-derived endothelial progenitor of the pharyngeal arches.


Michael WARKALA (Newark, New Jersey, USA), Sophie ASTROF
16:30 - 18:15 #30559 - 044. Fluid shear and hydrostatic stress potentiate YAP mechanotransduction to control distinct growth vs maturation programs in remodeling endocardial cushions.
044. Fluid shear and hydrostatic stress potentiate YAP mechanotransduction to control distinct growth vs maturation programs in remodeling endocardial cushions.

It is widely known that hemodynamic perturbations dramatically alter valvuloseptal morphogenesis, but specific mechanobiological mechanisms controlling clinically critical valve remodeling are lacking. We first profiled the in vivo expression of YAP, a key mechanotransduction mediator, during murine fetal valvular remodeling, and associated it with local mechanical forces. We determined YAP activation increased with stage, concomitant with increasing hemodynamic loading, but became restricted to the outflow (arterial) surface that experiences oscillatory shear stress (OSS). To directly test the remodeling mechanisms of specific loading modes, we directly exposed ex vivo chick endocardial cushions to hydrostatic stress (compressive vs tensile) or shear stress (steady laminar vs oscillatory) in organ culture. We determined that oscillatory shear stress (OSS) and compressive stress induce cushioned proliferation and biomechanical softening, supporting a pro-growth phenotype. In contrast, unidirectional shear stress (USS) and tensile hydrostatic stress promoted cushion compaction and stiffening, supporting a pro-maturation behavior. YAP nuclear localization was enhanced in OSS and compressive stress conditions but was suppressed in USS and tensile stress conditions, confirming the in vivo conditions. Further, YAP inhibition in either pro-growth stress conditions reduced proliferation, increased stiffness, and highly compacted morphology in cushions. To further test the role of YAP mechanotransduction in vivo, we applied partial left atrial ligations in embryonic chicks to de-load the ventricle. This resulted in both smaller mitral valves and reduced YAP activity. Together, our evidence supports that local shear and hydrostatic stress orchestrate local growth vs maturation decisions in endocardial cushions by controlling YAP mediated mechanotransduction.


Shuofei SUN (Ithaca, USA), Mingkun WANG, Belle LIN, Jonathan BUTCHER
16:30 - 18:15 #30575 - 045. Single cell multi-omics reveal novel cardiac subpopulation critical to valve morphogenesis.
045. Single cell multi-omics reveal novel cardiac subpopulation critical to valve morphogenesis.

Valvular heart disease is a major source of morbidity and mortality with an anticipated increase in prevalence secondary to an aging population and increase in survivorship for patients with congenital disease. Further developmental studies are required to advance novel therapeutic development. In this study, we have interrogated the later stages of valvulogenesis to understand the molecular mechanisms of valve formation and how these mechanisms are disrupted in the context of disease. Leveraging a combination of single-cell RNA/Chromatin Accessibility sequencing in the developing mouse heart, we identified a novel, rare cell population in the developing valve with a unique transcriptional profile comprised of highly specific developmental signaling pathway genes. These cells are first detectable after valve primordia formation at embryonic day (E) 12.5 and are spatially localized at the leading edge of the developing leaflets. Ablation of this rare subpopulation during development results in highly dysplastic valves, characterized by hyperplastic, redundant, immature leaflets associated with valvular stenosis and regurgitation. These dysplastic features are consistent with the features of several congenital valvulopathies including Ebstein’s Anomaly, and pulmonary or aortic valve stenosis. Single-cell RNA sequencing analysis of a human fetal heart with hypoplastic left heart syndrome and critical aortic stenosis demonstrated a depletion of this cell population in the diseased aortic valve, suggesting these cells may be required for normal human valvular development as well. This study establishes the existence of a novel, rare subpopulation of cardiac cells that are critical to valve development and may contribute to the pathogenesis of congenital valvulopathies.


Alexander MERRIMAN (San Francisco, USA), Mauro COSTA, Chun YE, Deepak SRIVASTAVA
16:30 - 18:15 #30522 - 046. Blood flow-dependent and -independent roles of Wnt9 during zebrafish heart morphogenesis.
046. Blood flow-dependent and -independent roles of Wnt9 during zebrafish heart morphogenesis.

During cardiac development, a blood-flow independent early phase, during which bilateral populations of cardiac progenitor cells assemble the nascent heart tube, is followed by a blood flow-dependent late phase, when cardiac chambers are shaped and cardiac valve leaflets are formed. We previously showed that blood flow-dependent Erk5-Klf2a signaling induces the expression of wnt9a and that this is essential for atrioventricular valve formation in zebrafish. While biomechanical signaling clearly impacts late heart morphogenesis, we still lack a good understanding of how morphogenesis of the early heart is regulated independently of blood flow.  Here, we present new data on an earlier role of Wnt9a and of its paralogue Wnt9b in the assembly of the nascent heart tube prior to the onset of blood flow. In wnt9a/b double mutants, the migratory potential of cardiomyocytes progenitors is affected, as they fail to converge from positions within the anterior lateral plate mesoderm towards the embryonic midline. This prevents heart cone formation and causes a collapsed heart at the midline. These mutants also exhibit premature differentiation of cardiomyocyte progenitor cells. This is in line with the known role of canonical Wnt signaling in preventing cardiac differentiation at stages of early cardiac development.  Using a pharmacological inhibition approach, we find that the physiological effect of Wnt9 in controlling cardiomyocyte progenitor migration towards the embryonic midline is mediated by non-canonical Wnt signaling. Hence, Wnt9 has both blood flow-dependent and -independent functions during heart development.


Alessio PAOLINI (Potsdam, Germany), Tim LANGE, Dinara SHARIPOVA, Salim ABDELILAH-SEYFRIED
16:30 - 18:15 #30636 - 047. A unidirectional shear stress operated Notch-CXCR4 molecular switch controls semilunar valve maturation.
047. A unidirectional shear stress operated Notch-CXCR4 molecular switch controls semilunar valve maturation.

While mechanisms of early heart valve formation are well investigated, the processes of growth and leaflet remodeling are clinically significant, yet poorly understood; genetic ablation studies have revealed limited cell lineage-specific roles in these programs. We here explore the role of local hemodynamic loading as a driver of conditional signaling in the remodeling of fetal semilunar valves.

Immunostaining of murine aortic valve leaflets revealed ventricularis-specific endocardial Notch1 and mesenchymal CXCR4, coincident with unidirectional fluid shear stress (USS). We next generated valve endocardial-specific dnMAML knockout mice and found markedly hyperplastic semilunar valve cushions with reduced mesenchymal CXCR4. In complement, we generated valve specific CXCR4 deletion mice, which demonstrated similar hyperplastic valves and perinatal lethality despite preserved coronary morphogenesis. Both mouse models exhibited elevated Wnt-BMP signaling, increased proliferation, increased nuclear Sox9, increased glycosaminoglycan deposition, and decreased aSMA, which are indicative of impaired leaflet phenotypic maturation and matrix stratification.

We then employed in vitro flow experiments with primary chick aortic valve endocardial cells to demonstrate that ventricularis-occurring USS directly activates Notch1 and upregulates CXCR4-ligand Sdf1 expression, while arterial-sided oscillatory shear stress (OSS) blocks Notch1, thereby activating Wnt-BMP. Treatment with Notch inhibitor DAPT blocked Sdf1 expression in USS treated endocardium. Further, semilunar cushion organ culture experiments showed that CXCR4 inhibition reduced aSMA expression and reduced tissue stiffness.

Together, these results identify a hemodynamic pattern-specific activation of semilunar valve growth (via OSS-Wnt/BMP) or tissue maturation (via USS-Notch1/Sdf1/Cxcr4) programming to robustly coordinate leaflet sculpting during dynamic and variable growth of the outlets.


Charles DAI, Belle LIN, Duc PHAM, Jonathan BUTCHER (Ithaca, USA)
Auditorium
18:15

"Saturday 14 May"

Added to your list of favorites
Deleted from your list of favorites
CC
18:15 - 18:30

Closing comments

Auditorium
19:00

"Saturday 14 May"

Added to your list of favorites
Deleted from your list of favorites
BA
19:00 - 23:00

Gala Dinner and Prize Giving (Palais du Pharo)