42 results on '"Alexandre R. Colas"'
Search Results
2. Multiplatform modeling of atrial fibrillation identifies phospholamban as a central regulator of cardiac rhythm
- Author
-
Anaïs Kervadec, James Kezos, Haibo Ni, Michael Yu, James Marchant, Sean Spiering, Suraj Kannan, Chulan Kwon, Peter Andersen, Rolf Bodmer, Eleonora Grandi, Karen Ocorr, and Alexandre R. Colas
- Subjects
cardiac disease modeling ,atrial fibrillation ,human ipsc-derived atrial-like cardiomyocytes ,computational modeling ,high-throughput electrophysiology ,single-cell resolution ,drosophila ,Medicine ,Pathology ,RB1-214 - Published
- 2023
- Full Text
- View/download PDF
3. Trajectory reconstruction identifies dysregulation of perinatal maturation programs in pluripotent stem cell-derived cardiomyocytes
- Author
-
Suraj Kannan, Matthew Miyamoto, Renjun Zhu, Michaela Lynott, Jason Guo, Elaine Zhelan Chen, Alexandre R. Colas, Brian Leei Lin, and Chulan Kwon
- Subjects
CP: Stem cell research ,Biology (General) ,QH301-705.5 - Abstract
Summary: A limitation in the application of pluripotent stem cell-derived cardiomyocytes (PSC-CMs) is the failure of these cells to achieve full functional maturity. The mechanisms by which directed differentiation differs from endogenous development, leading to consequent PSC-CM maturation arrest, remain unclear. Here, we generate a single-cell RNA sequencing (scRNA-seq) reference of mouse in vivo CM maturation with extensive sampling of previously difficult-to-isolate perinatal time periods. We subsequently generate isogenic embryonic stem cells to create an in vitro scRNA-seq reference of PSC-CM-directed differentiation. Through trajectory reconstruction, we identify an endogenous perinatal maturation program that is poorly recapitulated in vitro. By comparison with published human datasets, we identify a network of nine transcription factors (TFs) whose targets are consistently dysregulated in PSC-CMs across species. Notably, these TFs are only partially activated in common ex vivo approaches to engineer PSC-CM maturation. Our study can be leveraged toward improving the clinical viability of PSC-CMs.
- Published
- 2023
- Full Text
- View/download PDF
4. PGC1/PPAR drive cardiomyocyte maturation at single cell level via YAP1 and SF3B2
- Author
-
Sean A. Murphy, Matthew Miyamoto, Anaïs Kervadec, Suraj Kannan, Emmanouil Tampakakis, Sandeep Kambhampati, Brian Leei Lin, Sam Paek, Peter Andersen, Dong-Ik Lee, Renjun Zhu, Steven S. An, David A. Kass, Hideki Uosaki, Alexandre R. Colas, and Chulan Kwon
- Subjects
Science - Abstract
Cardiomyocyte maturation and the acquisition of phenotypes is poorly understood at the single cell level. Here, the authors analyse the transcriptome of single cells from neonatal to adult heart and reveal that peroxisome proliferator-activated receptor coactivator-1 mediates the phenotypic shift.
- Published
- 2021
- Full Text
- View/download PDF
5. Silencing of CCR4-NOT complex subunits affects heart structure and function
- Author
-
Lisa Elmén, Claudia B. Volpato, Anaïs Kervadec, Santiago Pineda, Sreehari Kalvakuri, Nakissa N. Alayari, Luisa Foco, Peter P. Pramstaller, Karen Ocorr, Alessandra Rossini, Anthony Cammarato, Alexandre R. Colas, Andrew A. Hicks, and Rolf Bodmer
- Subjects
cnot1 ,gwas ,arrhythmia ,long-qt syndrome ,drosophila heart ,hipsc ,cardiomyocytes ,Medicine ,Pathology ,RB1-214 - Abstract
The identification of genetic variants that predispose individuals to cardiovascular disease and a better understanding of their targets would be highly advantageous. Genome-wide association studies have identified variants that associate with QT-interval length (a measure of myocardial repolarization). Three of the strongest associating variants (single-nucleotide polymorphisms) are located in the putative promotor region of CNOT1, a gene encoding the central CNOT1 subunit of CCR4-NOT: a multifunctional, conserved complex regulating gene expression and mRNA stability and turnover. We isolated the minimum fragment of the CNOT1 promoter containing all three variants from individuals homozygous for the QT risk alleles and demonstrated that the haplotype associating with longer QT interval caused reduced reporter expression in a cardiac cell line, suggesting that reduced CNOT1 expression might contribute to abnormal QT intervals. Systematic siRNA-mediated knockdown of CCR4-NOT components in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) revealed that silencing CNOT1 and other CCR4-NOT genes reduced their proliferative capacity. Silencing CNOT7 also shortened action potential duration. Furthermore, the cardiac-specific knockdown of Drosophila orthologs of CCR4-NOT genes in vivo (CNOT1/Not1 and CNOT7/8/Pop2) was either lethal or resulted in dilated cardiomyopathy, reduced contractility or a propensity for arrhythmia. Silencing CNOT2/Not2, CNOT4/Not4 and CNOT6/6L/twin also affected cardiac chamber size and contractility. Developmental studies suggested that CNOT1/Not1 and CNOT7/8/Pop2 are required during cardiac remodeling from larval to adult stages. To summarize, we have demonstrated how disease-associated genes identified by GWAS can be investigated by combining human cardiomyocyte cell-based and whole-organism in vivo heart models. Our results also suggest a potential link of CNOT1 and CNOT7/8 to QT alterations and further establish a crucial role of the CCR4-NOT complex in heart development and function. This article has an associated First Person interview with the first author of the paper.
- Published
- 2020
- Full Text
- View/download PDF
6. Dietary Emulsifier Sodium Stearoyl Lactylate Alters Gut Microbiota in vitro and Inhibits Bacterial Butyrate Producers
- Author
-
Lisa Elmén, Jaime E. Zlamal, David A. Scott, Ryan B. Lee, Daniel J. Chen, Alexandre R. Colas, Dmitry A. Rodionov, and Scott N. Peterson
- Subjects
microbiome ,food additives ,dysbiosis ,short chain fatty acids ,Western diet ,bacterial genome reconstruction ,Microbiology ,QR1-502 - Abstract
Dietary emulsifiers are widely used in industrially processed foods, although the effects of these food additives on human gut microbiota are not well studied. Here, we investigated the effects of five different emulsifiers [glycerol monoacetate, glycerol monostearate, glycerol monooleate, propylene glycol monostearate, and sodium stearoyl lactylate (SSL)] on fecal microbiota in vitro. We found that 0.025% (w/v) of SSL reduced the relative abundance of the bacterial class Clostridia and others. The relative abundance of the families Clostridiaceae, Lachnospiraceae, and Ruminococcaceae was substantially reduced whereas that of Bacteroidaceae and Enterobacteriaceae was increased. Given the marked impact of SSL on Clostridia, we used genome reconstruction to predict community-wide production of short-chain fatty acids, which were experimentally assessed by GC-MS analysis. SSL significantly reduced concentrations of butyrate, and increased concentrations of propionate compared to control cultures. The presence of SSL increased lipopolysaccharide, LPS and flagellin in cultured communities, thereby enhancing the proinflammatory potential of SSL-selected bacterial communities.
- Published
- 2020
- Full Text
- View/download PDF
7. An Automated Platform for Assessment of Congenital and Drug-Induced Arrhythmia with hiPSC-Derived Cardiomyocytes
- Author
-
Wesley L. McKeithan, Alex Savchenko, Michael S. Yu, Fabio Cerignoli, Arne A. N. Bruyneel, Jeffery H. Price, Alexandre R. Colas, Evan W. Miller, John R. Cashman, and Mark Mercola
- Subjects
induced pluripotent stem cells ,cardiomyocyte ,voltage sensitive probe ,high throughput screening ,drug development ,cardiotoxicity ,Physiology ,QP1-981 - Abstract
The ability to produce unlimited numbers of human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) harboring disease and patient-specific gene variants creates a new paradigm for modeling congenital heart diseases (CHDs) and predicting proarrhythmic liabilities of drug candidates. However, a major roadblock to implementing hiPSC-CM technology in drug discovery is that conventional methods for monitoring action potential (AP) kinetics and arrhythmia phenotypes in vitro have been too costly or technically challenging to execute in high throughput. Herein, we describe the first large-scale, fully automated and statistically robust analysis of AP kinetics and drug-induced proarrhythmia in hiPSC-CMs. The platform combines the optical recording of a small molecule fluorescent voltage sensing probe (VoltageFluor2.1.Cl), an automated high throughput microscope and automated image analysis to rapidly generate physiological measurements of cardiomyocytes (CMs). The technique can be readily adapted on any high content imager to study hiPSC-CM physiology and predict the proarrhythmic effects of drug candidates.
- Published
- 2017
- Full Text
- View/download PDF
8. Nascent polypeptide-Associated Complex and Signal Recognition Particle have cardiac-specific roles in heart development and remodeling.
- Author
-
Analyne M Schroeder, Tanja Nielsen, Michaela Lynott, Georg Vogler, Alexandre R Colas, and Rolf Bodmer
- Subjects
Genetics ,QH426-470 - Abstract
Establishing a catalog of Congenital Heart Disease (CHD) genes and identifying functional networks would improve our understanding of its oligogenic underpinnings. Our studies identified protein biogenesis cofactors Nascent polypeptide-Associated Complex (NAC) and Signal-Recognition-Particle (SRP) as disease candidates and novel regulators of cardiac differentiation and morphogenesis. Knockdown (KD) of the alpha- (Nacα) or beta-subunit (bicaudal, bic) of NAC in the developing Drosophila heart disrupted cardiac developmental remodeling resulting in a fly with no heart. Heart loss was rescued by combined KD of Nacα with the posterior patterning Hox gene Abd-B. Consistent with a central role for this interaction in cardiogenesis, KD of Nacα in cardiac progenitors derived from human iPSCs impaired cardiac differentiation while co-KD with human HOXC12 and HOXD12 rescued this phenotype. Our data suggest that Nacα KD preprograms cardioblasts in the embryo for abortive remodeling later during metamorphosis, as Nacα KD during translation-intensive larval growth or pupal remodeling only causes moderate heart defects. KD of SRP subunits in the developing fly heart produced phenotypes that targeted specific segments and cell types, again suggesting cardiac-specific and spatially regulated activities. Together, we demonstrated directed function for NAC and SRP in heart development, and that regulation of NAC function depends on Hox genes.
- Published
- 2022
- Full Text
- View/download PDF
9. Patient-specific genomics and cross-species functional analysis implicate LRP2 in hypoplastic left heart syndrome
- Author
-
Jeanne L Theis, Georg Vogler, Maria A Missinato, Xing Li, Tanja Nielsen, Xin-Xin I Zeng, Almudena Martinez-Fernandez, Stanley M Walls, Anaïs Kervadec, James N Kezos, Katja Birker, Jared M Evans, Megan M O'Byrne, Zachary C Fogarty, André Terzic, Paul Grossfeld, Karen Ocorr, Timothy J Nelson, Timothy M Olson, Alexandre R Colas, and Rolf Bodmer
- Subjects
lipoproteins ,iPSC ,cardiogenesis ,congenital heart disease ,hypoplastic left heart syndrome ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Congenital heart diseases (CHDs), including hypoplastic left heart syndrome (HLHS), are genetically complex and poorly understood. Here, a multidisciplinary platform was established to functionally evaluate novel CHD gene candidates, based on whole-genome and iPSC RNA sequencing of a HLHS family-trio. Filtering for rare variants and altered expression in proband iPSCs prioritized 10 candidates. siRNA/RNAi-mediated knockdown in healthy human iPSC-derived cardiomyocytes (hiPSC-CM) and in developing Drosophila and zebrafish hearts revealed that LDL receptor-related protein LRP2 is required for cardiomyocyte proliferation and differentiation. Consistent with hypoplastic heart defects, compared to parents the proband’s iPSC-CMs exhibited reduced proliferation. Interestingly, rare, predicted-damaging LRP2 variants were enriched in a HLHS cohort; however, understanding their contribution to HLHS requires further investigation. Collectively, we have established a multi-species high-throughput platform to rapidly evaluate candidate genes and their interactions during heart development, which are crucial first steps toward deciphering oligogenic underpinnings of CHDs, including hypoplastic left hearts.
- Published
- 2020
- Full Text
- View/download PDF
10. Genetic architecture of natural variation of cardiac performance from flies to humans
- Author
-
Lionel Spinelli, Saswati Saha, Jaime A Castro Mondragon, Anaïs Kervadec, Michaela Lynott, Laurent Kremmer, Laurence Roder, Sallouha Krifa, Magali Torres, Christine Brun, Georg Vogler, Rolf Bodmer, Alexandre R Colas, Karen Ocorr, Laurent Perrin, Theories and Approaches of Genomic Complexity (TAGC), Aix Marseille Université (AMU)-Institut National de la Santé et de la Recherche Médicale (INSERM), Centre for Molecular Medicine Norway (NCMM), Faculty of Medicine [Oslo], University of Oslo (UiO)-University of Oslo (UiO)-Rigshospitalet [Copenhagen], Copenhagen University Hospital-Copenhagen University Hospital, and Sanford Burnham Prebys Medical Discovery Institute
- Subjects
General Immunology and Microbiology ,General Neuroscience ,Quantitative Trait Loci ,Genetic Variation ,Heart ,General Medicine ,General Biochemistry, Genetics and Molecular Biology ,[SDV.GEN.GA]Life Sciences [q-bio]/Genetics/Animal genetics ,Drosophila melanogaster ,Phenotype ,Animals ,Humans ,Gene Regulatory Networks ,Genome-Wide Association Study - Abstract
Deciphering the genetic architecture of human cardiac disorders is of fundamental importance but their underlying complexity is a major hurdle. We investigated the natural variation of cardiac performance in the sequenced inbred lines of the Drosophila Genetic Reference Panel (DGRP). Genome-wide associations studies (GWAS) identified genetic networks associated with natural variation of cardiac traits which were used to gain insights as to the molecular and cellular processes affected. Non-coding variants that we identified were used to map potential regulatory non-coding regions, which in turn were employed to predict transcription factors (TFs) binding sites. Cognate TFs, many of which themselves bear polymorphisms associated with variations of cardiac performance, were also validated by heart-specific knockdown. Additionally, we showed that the natural variations associated with variability in cardiac performance affect a set of genes overlapping those associated with average traits but through different variants in the same genes. Furthermore, we showed that phenotypic variability was also associated with natural variation of gene regulatory networks. More importantly, we documented correlations between genes associated with cardiac phenotypes in both flies and humans, which supports a conserved genetic architecture regulating adult cardiac function from arthropods to mammals. Specifically, roles for PAX9 and EGR2 in the regulation of the cardiac rhythm were established in both models, illustrating that the characteristics of natural variations in cardiac function identified in Drosophila can accelerate discovery in humans.
- Published
- 2022
11. Author response: Genetic architecture of natural variation of cardiac performance from flies to humans
- Author
-
Lionel Spinelli, Saswati Saha, Jaime A Castro Mondragon, Anaïs Kervadec, Michaela Lynott, Laurent Kremmer, Laurence Roder, Sallouha Krifa, Magali Torres, Christine Brun, Georg Vogler, Rolf Bodmer, Alexandre R Colas, Karen Ocorr, and Laurent Perrin
- Published
- 2022
12. Multiplatform Modeling of Atrial Fibrillation Identifies Phospholamban as Central Regulator of Cardiac Rhythm
- Author
-
Anaïs Kervadec, James Kezos, Haibo Ni, Michael Yu, Sean Spiering, Suraj Kannan, Peter Andersen, Eleonora Grandi, Karen Ocorr, and Alexandre R. Colas
- Abstract
Atrial fibrillation (AF) is the most common form of sustained cardiac arrhythmia in humans, present in > 33 million people worldwide. Although AF is often developed secondary to cardiovascular diseases, endocrine disorders, or lifestyle factors, recent GWAS studies have identified >200 genetic variants that substantially contribute to AF risk. However, it is currently not known how these genetic predispositions contribute to the initiation and/or maintenance of AF-associated phenotypes. In this context, one major barrier to progress is the lack of experimental systems enabling to rapidly explore the function of large cohort of genes on rhythm parameters in models with human atrial relevance. To address these modeling challenges, we have developed a new multi-model platform enabling 1) high-throughput characterization of the role of AF-associated genes on action potential duration and rhythm parameters at the cellular level, using human iPSC-derived atrial-like cardiomyocytes (ACMs), and at the whole organ level, using the Drosophila heart model, and 2) validation of the physiological relevance of our experimental results using computational models of heterogenous human adult atrial myocytes (HAMs) and tissue. As proof of concept, we screened a cohort of 20 AF-associated genes and identified Phospholamban (PLN) loss of function as a top conserved hit that significantly shortens action potential duration in ACMs, HAMs and fly cardiomyocytes. Remarkably, while PLN knock-down (KD) was not sufficient to induce arrhythmia phenotypes, addition of environmental stressors (i.e fibroblasts, β-adrenergic stimulation) to the model systems, led to the robust generation of irregular beat to beat intervals, delayed after depolarizations, and triggered action potentials, as compared to controls. Finally, to delineate the mechanism underlying PLN KD-dependent arrhythmia, we used a logistic regression approach in HAM populations, and predicted that PLN functionally interacts with both NCX (loss of function) and L-type calcium channels (gain of function) to mediate these arrhythmic phenotypes. Consistent with our predictions, co-KD of PLN and NCX in ACMs and flies, led to increased arrhythmic events, while treatment of ACMs with L-type calcium channel inhibitor, verapamil, reverted these phenotypes. In summary, these results collectively demonstrate that our integrated multi-model system approach was successful in identifying and characterizing conserved roles (i.e regulation of Ca2+ homeostasis) for AF-associated genes and phenotypes, and thus paves the way for the discovery and molecular delineation of new gene regulatory networks controlling atrial rhythm with application to AF.
- Published
- 2022
13. Author Reply to Peer Reviews of Genetic architecture of natural variation of cardiac performance in flies
- Author
-
Laurent Perrin, Karen Ocorr, Alexandre R. Colas, Rolf Bodmer, Georg Vogler, Christine Brun, Magali Torres, Sallouha Krifa, Laurence Roder, Laurent Kremmer, Anaïs Kervadec, Jaime A Castro-Mondragon, Lionel Spinelli, and Saswati Saha
- Published
- 2022
14. Functional analysis across model systems implicates ribosomal proteins in growth and proliferation defects associated with hypoplastic left heart syndrome
- Author
-
Tanja Nielsen, Anaïs Kervadec, Maria A. Missinato, Michaela Lynott, Xin-Xin I. Zeng, Marie Berenguer, Stanley M. Walls, Analyne Schroeder, Katja Birker, Greg Duester, Paul Grossfeld, Timothy J. Nelson, Timothy M. Olson, Karen Ocorr, Rolf Bodmer, Jeanne L. Theis, Georg Vogler, and Alexandre R. Colas
- Abstract
Hypoplastic left heart syndrome (HLHS) is the most lethal congenital heart disease (CHD). The pathogenesis of HLHS is poorly understood and due to the likely oligogenic complexity of the disease, definitive HLHS-causing genes have not yet been identified. Postulating that impaired cardiomyocyte proliferation as a likely important contributing mechanism to HLHS pathogenesis, and we conducted a genome-wide siRNA screen to identify genes affecting proliferation of human iPSC-derived cardiomyocytes (hPSC-CMs). This yielded ribosomal protein (RP) genes as the most prominent class of effectors of CM proliferation. In parallel, whole genome sequencing and rare variant filtering of a cohort of 25 HLHS proband-parent trios with poor clinical outcome revealed enrichment of rare variants of RP genes. In addition, in another familial CHD case of HLHS proband we identified a rare, predicted-damaging promoter variant affectingRPS15Athat was shared between the proband and a distant relative with CHD. Functional testing with an integrated multi-model system approach reinforced the idea that RP genes are major regulators of cardiac growth and proliferation, thus potentially contributing to hypoplastic phenotype observed in HLHS patients. Cardiac knockdown (KD) of RP genes with promoter or coding variants (RPS15A, RPS17, RPL26L1, RPL39, RPS15) reduced proliferation in generic hPSC-CMs and caused malformed hearts, heart-loss or even lethality inDrosophila. In zebrafish, diminishedrps15afunction caused reduced CM numbers, heart looping defects, or weakened contractility, while reducedrps17orrpl39function caused reduced ventricular size or systolic atrial dysfunction, respectively. Importantly, genetic interactions betweenRPS15Aand core cardiac transcription factorsTBX5in CMs,Drosocross, pannierandtinmanin flies, andtbx5andnkx2-7(nkx2-5paralog) in fish, support a specific role for RP genes in heart development. Furthermore,RPS15AKD-induced heart/CM proliferation defects were significantly attenuated byp53KD in both hPSC-CMs and zebrafish, and by Hippo activation (YAP/yorkieoverexpression) in developing fly hearts. Based on these findings, we conclude that RP genes play critical roles in cardiogenesis and constitute an emerging novel class of gene candidates likely involved in HLHS and other CHDs.
- Published
- 2022
15. Mitochondrial MICOS complex genes, implicated in hypoplastic left heart syndrome, maintain cardiac contractility and actomyosin integrity
- Author
-
Katja Birker, Natalie J. Kirkland, Jeanne L. Theis, Zachary C. Fogarty, Maria Azzurra Missinato, Sreehari Kalvakuri, Paul Grossfeld, Adam J. Engler, Karen Ocorr, Timothy J. Nelson, Alexandre R. Colas, Timothy M. Olson, Georg Vogler, and Rolf Bodmer
- Abstract
Hypoplastic left heart syndrome (HLHS) is a severe congenital heart disease (CHD) with a likely oligogenic etiology, but our understanding of the genetic complexities and pathogenic mechanisms leading to HLHS is limited. We therefore performed whole genome sequencing (WGS) on a large cohort of HLHS patients and their families to identify candidate genes that were then tested in Drosophila heart model for functional and structural requirements. Bioinformatic analysis of WGS data from an index family comprised of a HLHS proband born to consanguineous parents and postulated to have a homozygous recessive disease etiology, prioritized 9 candidate genes with rare, predicted damaging homozygous variants. Of the candidate HLHS gene homologs tested, cardiac-specific knockdown (KD) of mitochondrial MICOS complex subunit dCHCHD3/6 resulted in drastically compromised heart contractility, diminished levels of sarcomeric actin and myosin, reduced cardiac ATP levels, and mitochondrial fission-fusion defects. Interestingly, these heart defects were similar to those inflicted by cardiac KD of ATP synthase subunits of the electron transport chain (ETC), consistent with the MICOS complex’s role in maintaining cristae morphology and ETC complex assembly. Analysis of 183 genomes of HLHS patient-parent trios revealed five additional HLHS probands with rare, predicted damaging variants in CHCHD3 or CHCHD6. Hypothesizing an oligogenic basis for HLHS, we tested 60 additional prioritized candidate genes in these cases for genetic interactions with CHCHD3/6 in sensitized fly hearts. Moderate KD of CHCHD3/6 in combination with Cdk12 (activator of RNA polymerase II), RNF149 (E3 ubiquitin ligase), or SPTBN1 (scaffolding protein) caused synergistic heart defects, suggesting the potential involvement of a diverse set of pathways in HLHS. Further elucidation of novel candidate genes and genetic interactions of potentially disease-contributing pathways is expected to lead to a better understanding of HLHS and other CHDs.
- Published
- 2022
16. PGC1/PPAR drive cardiomyocyte maturation at single cell level via YAP1 and SF3B2
- Author
-
Renjun Zhu, Dong Ik Lee, Suraj Kannan, Hideki Uosaki, Steven S. An, Sean Murphy, Sam Paek, Emmanouil Tampakakis, Sandeep Kambhampati, David A. Kass, Alexandre R. Colas, Brian L. Lin, Matthew Miyamoto, Anaïs Kervadec, Chulan Kwon, and Peter Andersen
- Subjects
Pluripotent Stem Cells ,0301 basic medicine ,Science ,Induced Pluripotent Stem Cells ,Peroxisome Proliferator-Activated Receptors ,Gene regulatory network ,Regulator ,Stem-cell differentiation ,General Physics and Astronomy ,Peroxisome proliferator-activated receptor ,Cell Cycle Proteins ,Biology ,Article ,General Biochemistry, Genetics and Molecular Biology ,Gene regulatory networks ,Transcriptome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Animals ,Humans ,Myocyte ,Myocytes, Cardiac ,Receptor ,Adaptor Proteins, Signal Transducing ,chemistry.chemical_classification ,YAP1 ,Multidisciplinary ,Cell Differentiation ,YAP-Signaling Proteins ,General Chemistry ,Phenotype ,Cell biology ,030104 developmental biology ,chemistry ,Calcium ,RNA Splicing Factors ,Heart stem cells ,030217 neurology & neurosurgery ,Signal Transduction ,Transcription Factors - Abstract
Cardiomyocytes undergo significant structural and functional changes after birth, and these fundamental processes are essential for the heart to pump blood to the growing body. However, due to the challenges of isolating single postnatal/adult myocytes, how individual newborn cardiomyocytes acquire multiple aspects of the mature phenotype remains poorly understood. Here we implement large-particle sorting and analyze single myocytes from neonatal to adult hearts. Early myocytes exhibit wide-ranging transcriptomic and size heterogeneity that is maintained until adulthood with a continuous transcriptomic shift. Gene regulatory network analysis followed by mosaic gene deletion reveals that peroxisome proliferator-activated receptor coactivator-1 signaling, which is active in vivo but inactive in pluripotent stem cell-derived cardiomyocytes, mediates the shift. This signaling simultaneously regulates key aspects of cardiomyocyte maturation through previously unrecognized proteins, including YAP1 and SF3B2. Our study provides a single-cell roadmap of heterogeneous transitions coupled to cellular features and identifies a multifaceted regulator controlling cardiomyocyte maturation., Cardiomyocyte maturation and the acquisition of phenotypes is poorly understood at the single cell level. Here, the authors analyse the transcriptome of single cells from neonatal to adult heart and reveal that peroxisome proliferator-activated receptor coactivator-1 mediates the phenotypic shift.
- Published
- 2021
17. Nascent Polypeptide Associated Complex–alphaand Signal Recognition Particle are required for cardiac development and remodeling
- Author
-
Analyne M. Schroeder, Georg Vogler, Alexandre R. Colas, and Rolf Bodmer
- Abstract
Congenital Heart Disease (CHD) is driven by a strong genetic predisposition, yet only a small subset of patients (∼20%) are diagnosed with a precise genetic cause. Therefore, expanding the pool of genes associated with CHD and establishing the functional relationships between genes can assemble a more comprehensive genetic network to better understand cardiac development and pathogenesis. In our studies, we identified protein biogenesis cofactors Nascent polypeptide Associated Complex (NAC) and Signal Recognition Particle (SRP) that bind disparate subsets of emerging nascent polypeptides at the ribosome exit site to direct polypeptide fates, as novel regulators of cell differentiation and cardiac morphogenesis. Knockdown (KD) of the alpha-(Nacα)or beta- subunit (bicaudal, bic)of NAC in the developingDrosophilaheart led to disruption of cardiac remodeling during pupal stages resulting in an adult fly with no heart. Heart loss was rescued by combined KD ofNacαwith theHoxgeneAbd-B.Consistent with a central role for this interaction in the regulation of cardiogenesis, KD ofNacαin Cardiac Progenitors derived from human iPSCs impaired cardiac differentiation while co-KD with mammalianHoxgenesHOXC12 and HOXD12rescued this phenotype. The effect ofNacαKD on the fly heart was temporally regulated, in that KD in embryo or in pupae caused only a partial loss of the heart, whereas KD during both stages led to heart loss similar to continuous KD throughout life. This suggests thatNacαKD already in the embryo may reprogram cells leading to aberrant cardiac remodeling during pupal stages. Lastly, KD of several SRP subunits individually in the fly heart produced a range of cardiac phenotypes that targeted specific segments and cell types, indicating spatially regulated activities of SRP components in the heart. Together, these data suggest that despite NAC and SRP ubiquitous presence, they displayed spatially and temporally fine-tuned activities for proper cardiac morphogenesis.Nacα’sinteraction with cardiac-specificHoxgene functions builds upon the novel role of this pathway and expands our understanding of the complex genetic networks involved in cardiac development and pathogenesis.
- Published
- 2022
18. Atrial Fibrillation Genomics: Discovery and Translation
- Author
-
Karen Ocorr, Rolf Bodmer, Evan D. Muse, David H. Yoo, Alexandre R. Colas, and Christopher J. Larson
- Subjects
business.industry ,Drug discovery ,Atrial fibrillation ,Genomics ,Genome-wide association study ,Computational biology ,Disease ,medicine.disease ,Precision medicine ,Digital health ,medicine ,Cardiology and Cardiovascular Medicine ,business ,Genetic association - Abstract
Our understanding of the fundamental cellular and molecular factors leading to atrial fibrillation (AF) remains stagnant despite significant advancement in ablation and device technologies. Diagnosis and prevention strategies fall behind that of treatment, but expanding knowledge in AF genetics holds the potential to drive progress. We aim to review how an understanding of the genetic contributions to AF can guide an approach to individualized risk stratification and novel avenues in drug discovery. Rare familial forms of AF identified monogenic contributions to the development of AF. Genome-wide association studies (GWAS) further identified single-nucleotide polymorphisms (SNPs) suggesting polygenic and multiplex nature of this common disease. Polygenic risk scores accounting for the multitude of associated SNPs that each confer mildly elevated risk have been developed to translate genetic information into clinical practice, though shortcomings remain. Additionally, novel laboratory methods have been empowered by recent genetic findings to enhance drug discovery efforts. AF is increasingly recognized as a disease with a significant genetic component. With expanding sequencing technologies and accessibility, polygenic risk scores can help identify high risk individuals. Advancement in digital health tools, artificial intelligence and machine learning based on standard electrocardiograms, and genomic driven drug discovery may be integrated to deliver a sophisticated level of precision medicine in this modern era of emphasis on prevention. Randomized, prospective studies to demonstrate clinical benefits of these available tools are needed to validate this approach.
- Published
- 2021
19. Conserved Transcription Factors Control Chromatin Accessibility and Gene Expression to Maintain Cell Fate Stability and Restrict Reprogramming of Differentiated Cells
- Author
-
Peter Andersen, Li Qian, Maria A. Missinato, Sean Murphy, Prashila Amatya, Christopher Lee, Alessandra Sacco, Michaela Lynott, Suraj Kannan, Alexandre R. Colas, Peter D. Adams, Pier Lorenzo Puri, Chun-Teng Huang, Anaïs Kervadec, Chulan Kwon, Hiroshi Tanaka, Michael S. Yu, Yu-Ling Chang, and Mafalda Loreti
- Subjects
Cell type ,Cellular differentiation ,Biology ,Cell fate determination ,Induced pluripotent stem cell ,Reprogramming ,Embryonic stem cell ,Transcription factor ,Chromatin ,Cell biology - Abstract
The comprehensive characterization of mechanisms safeguarding cell fate identity in differentiated cells is crucial for 1) our understanding of how differentiation is maintained in healthy tissues or misregulated in disease states and 2) to improve our ability to use direct reprogramming for regenerative purposes. To uncover novel fate-stabilizing regulators, we employed a genome-wide TF siRNA screen followed by a high-complexity combinatorial evaluation of top performing hits, in a cardiac reprogramming assay in mouse embryonic fibroblasts, and subsequently validated our findings in cardiac, neuronal and iPSCs reprogramming assays in primary human fibroblasts and adult endothelial cells. This approach identified a conserved set of 4 TFs (ATF7IP, JUNB, SP7, and ZNF207 [AJSZ]) that robustly opposes cell fate reprogramming, as demonstrated by up to 6-fold increases in efficiency upon AJSZ knockdown in both lineage- and cell type-independent manners. Mechanistically, ChIP-seq and single-cell ATAC-seq analyses, revealed that AJSZ bind to both open and closed chromatin in a genome-wide and regionalized fashion, thereby limiting reprogramming TFs access to target DNA and ability to remodel the chromatin. In parallel, integration of ChIP-seq and RNA-seq data followed by systematic functional gene testing, identified that AJSZ also promote cell fate stability by proximally down-regulating a conserved set of genes involved in the regulation of cell fate specification (MEF2C), proteome remodeling (TPP1, PPIC), ATP homeostasis (EFHD1), and inflammation signaling (IL7R), thereby limiting cells ability to undergo large-scale phenotypic changes. Finally, simultaneous knock-down of AJSZ in combination with cardiac reprogramming TFs overexpression improved heart function by 250% as compared to no treatment and 50% as compared to MGT, 1 month after myocardial infarction. In sum, this study uncovers a novel evolutionarily conserved mechanism mediating cell fate stability in differentiated cells and also identifies AJSZ as promising therapeutic targets for regenerative purposes in adult organs.Significance StatementDifferentiated cells can be converted from one cell type into another by overexpressing lineage-determining transcription factors. Direct lineage reprogramming represents a promising strategy for regenerative medicine, but current clinical applications remain limited by the low yield of the reprogramming process. Here, we present the identification and detailed mechanistic study of a novel mechanisms opposing reprogramming process and promoting cell fate stability. Using a highthroughput screening technique, we identified four transcription factors that act as blockades to cell type change. By tracking chromatin and gene expression changes, we reconstructed a conserved pathway mediating cell fate stability in differentiated cells.
- Published
- 2021
20. Atrial Fibrillation Genomics: Discovery and Translation
- Author
-
David H, Yoo, Rolf, Bodmer, Karen, Ocorr, Christopher J, Larson, Alexandre R, Colas, and Evan D, Muse
- Subjects
Artificial Intelligence ,Atrial Fibrillation ,Humans ,Genetic Predisposition to Disease ,Genomics ,Prospective Studies ,Genome-Wide Association Study - Abstract
Our understanding of the fundamental cellular and molecular factors leading to atrial fibrillation (AF) remains stagnant despite significant advancement in ablation and device technologies. Diagnosis and prevention strategies fall behind that of treatment, but expanding knowledge in AF genetics holds the potential to drive progress. We aim to review how an understanding of the genetic contributions to AF can guide an approach to individualized risk stratification and novel avenues in drug discovery.Rare familial forms of AF identified monogenic contributions to the development of AF. Genome-wide association studies (GWAS) further identified single-nucleotide polymorphisms (SNPs) suggesting polygenic and multiplex nature of this common disease. Polygenic risk scores accounting for the multitude of associated SNPs that each confer mildly elevated risk have been developed to translate genetic information into clinical practice, though shortcomings remain. Additionally, novel laboratory methods have been empowered by recent genetic findings to enhance drug discovery efforts. AF is increasingly recognized as a disease with a significant genetic component. With expanding sequencing technologies and accessibility, polygenic risk scores can help identify high risk individuals. Advancement in digital health tools, artificial intelligence and machine learning based on standard electrocardiograms, and genomic driven drug discovery may be integrated to deliver a sophisticated level of precision medicine in this modern era of emphasis on prevention. Randomized, prospective studies to demonstrate clinical benefits of these available tools are needed to validate this approach.
- Published
- 2021
21. Genetic architecture of natural variation of cardiac performance: from flies to Humans
- Author
-
Alexandre R. Colas, Rolf Bodmer, Laurent Kremmer, Georg Vogler, Laurence Röder, Anaïs Kervadec, Karen Ocorr, Laurent Perrin, Krifa Sallouha, Lionel Spinelli, Magali Torres, Jaime A. Castro-Mondragon, Saswati Saha, and Christine Brun
- Subjects
Cardiac function curve ,0303 health sciences ,Computational biology ,Biology ,Genome ,Phenotype ,Genetic architecture ,03 medical and health sciences ,0302 clinical medicine ,Natural population growth ,030220 oncology & carcinogenesis ,Epistasis ,Gene ,Function (biology) ,030304 developmental biology - Abstract
Background Deciphering the genetic architecture of cardiac disorders is of fundamental importance but their underlying complexity is a major hurdle. Drosophila has gained importance as a useful model to study heart development and function and allows the analysis of organismal traits in a physiologically relevant and accessible system. Our aim was to (i) identify in flies the loci associated to natural variations of cardiac performances among a natural population, (ii) decipher how these variants interact with each other and with the environment to impact cardiac traits, (iii) gain insights about the molecular and cellular processes affected, (iv) determine whether the genetic architecture of cardiac disorders is conserved with humans. Methods and Results We investigated the genetic architecture of natural variations of cardiac performance in the sequenced inbred lines of the Drosophila Genetic Reference Panel (DGRP). Genome Wide Associations (GWA) for single markers and epistatic interactions identified genetic networks associated with natural variations of cardiac traits that were extensively validated in vivo. Non-coding variants were used to map potential regulatory non-coding regions which in turn were employed to predict Transcription Factors (TFs) binding sites. Cognate TFs, many of which themselves bear polymorphisms associated with variations of cardiac performance, were validated by heart specific knockdown. We also analyzed natural variations of cardiac traits variance that revealed unique features of their micro-environmental plasticity. More importantly, correlations between genes associated with cardiac phenotypes both in flies and in humans support the conserved genetic architecture of cardiac functioning from arthropods to mammals. The characteristics of natural variations in cardiac function established in Drosophila may thus guide the analysis of cardiac disorders in humans. Using human iPSC-derived cardiomyocytes, we indeed characterized a conserved function for PAX9 and EGR2 in the regulation of the cardiac rhythm Conclusion In-depth analysis of the genetic architecture of natural variations of cardiac performance in flies combined with functional validations in vivo and in human iPSC-CM represents a major achievement in understanding the mechanisms underlying the genetic architecture of these complex traits and a valuable resource for the identification of genes and mechanisms involved in cardiac disorders in humans.
- Published
- 2021
22. Temporal mechanisms of myogenic specification in human induced pluripotent stem cells
- Author
-
Shankar Subramaniam, Shyni Varghese, P. Nayak, Alexandre R. Colas, and Mark Mercola
- Subjects
Mesoderm ,Induced Pluripotent Stem Cells ,Biology ,Muscle Development ,Transcriptome ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Gene expression ,medicine ,Humans ,Myocyte ,Health and Medicine ,Muscle, Skeletal ,Research Articles ,030304 developmental biology ,0303 health sciences ,Gene knockdown ,Multidisciplinary ,Myogenesis ,Systems Biology ,SciAdv r-articles ,Skeletal muscle ,Cell Differentiation ,Cell biology ,medicine.anatomical_structure ,030217 neurology & neurosurgery ,Research Article - Abstract
Mechanisms of myogenic commitment are deciphered through omics analysis of hiPSC-specific lineage specification measurements., Understanding the mechanisms of myogenesis in human induced pluripotent stem cells (hiPSCs) is a prerequisite to achieving patient-specific therapy for diseases of skeletal muscle. hiPSCs of different origin show distinctive kinetics and ability to differentiate into myocytes. To address the unique cellular and temporal context of hiPSC differentiation, we perform a longitudinal comparison of the transcriptomic profiles of three hiPSC lines that display differential myogenic specification, one robust and two blunted. We detail temporal differences in mechanisms that lead to robust myogenic specification. We show gene expression signatures of putative cell subpopulations and extracellular matrix components that may support myogenesis. Furthermore, we show that targeted knockdown of ZIC3 at the outset of differentiation leads to improved myogenic specification in blunted hiPSC lines. Our study suggests that β-catenin transcriptional cofactors mediate cross-talk between multiple cellular processes and exogenous cues to facilitate specification of hiPSCs to mesoderm lineage, leading to robust myogenesis.
- Published
- 2021
23. Patient-specific genomics and cross-species functional analysis implicate LRP2 in hypoplastic left heart syndrome
- Author
-
Megan M. O’Byrne, Anaïs Kervadec, Almudena Martinez-Fernandez, Zeng Xi, Rolf Bodmer, Katja Birker, Timothy J. Nelson, Xing Li, Paul Grossfeld, Andre Terzic, Georg Vogler, Jeanne L. Theis, Stanley M. Walls, Karen Ocorr, Jared M. Evans, Alexandre R. Colas, James N. Kezos, Zachary C. Fogarty, Timothy M. Olson, Tanja Nielsen, and Maria A. Missinato
- Subjects
Male ,0301 basic medicine ,Proband ,Candidate gene ,QH301-705.5 ,Science ,Genomics ,Bioinformatics ,General Biochemistry, Genetics and Molecular Biology ,Hypoplastic left heart syndrome ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Biology (General) ,Zebrafish ,Gene knockdown ,iPSC ,D. melanogaster ,General Immunology and Microbiology ,biology ,Heart development ,General Neuroscience ,cardiogenesis ,Heart ,Genetics and Genomics ,hypoplastic left heart syndrome ,General Medicine ,medicine.disease ,biology.organism_classification ,LRP2 ,congenital heart disease ,Tools and Resources ,lipoproteins ,Low Density Lipoprotein Receptor-Related Protein-2 ,Drosophila melanogaster ,030104 developmental biology ,Medicine ,Female ,030217 neurology & neurosurgery ,Human - Abstract
Congenital heart diseases (CHDs), including hypoplastic left heart syndrome (HLHS), are genetically complex and poorly understood. Here, a multidisciplinary platform was established to functionally evaluate novel CHD gene candidates, based on whole-genome and iPSC RNA sequencing of a HLHS family-trio. Filtering for rare variants and altered expression in proband iPSCs prioritized 10 candidates. siRNA/RNAi-mediated knockdown in healthy human iPSC-derived cardiomyocytes (hiPSC-CM) and in developingDrosophilaand zebrafish hearts revealed that LDL receptor-related proteinLRP2is required for cardiomyocyte proliferation and differentiation. Consistent with hypoplastic heart defects, compared to parents the proband’s iPSC-CMs exhibited reduced proliferation. Interestingly, rare, predicted-damaging LRP2 variants were enriched in a HLHS cohort; however, understanding their contribution to HLHS requires further investigation. Collectively, we have established a multi-species high-throughput platform to rapidly evaluate candidate genes and their interactions during heart development, which are crucial first steps toward deciphering oligogenic underpinnings of CHDs, including hypoplastic left hearts.
- Published
- 2020
24. Author response: Patient-specific genomics and cross-species functional analysis implicate LRP2 in hypoplastic left heart syndrome
- Author
-
Paul Grossfeld, Maria A. Missinato, Megan M. O’Byrne, Almudena Martinez-Fernandez, Katja Birker, Jared M. Evans, James N. Kezos, Anaïs Kervadec, Rolf Bodmer, Georg Vogler, Timothy J. Nelson, Jeanne L. Theis, Zeng Xi, Zachary C. Fogarty, Timothy M. Olson, Karen Ocorr, Tanja Nielsen, Alexandre R. Colas, Andre Terzic, Xing Li, and Stanley M. Walls
- Subjects
medicine.medical_specialty ,business.industry ,Internal medicine ,medicine ,Cardiology ,Genomics ,Patient specific ,medicine.disease ,LRP2 ,business ,Functional analysis (psychology) ,Hypoplastic left heart syndrome - Published
- 2020
25. Dietary Emulsifier Sodium Stearoyl Lactylate Alters Gut Microbiota in vitro and Inhibits Bacterial Butyrate Producers
- Author
-
Scott N. Peterson, Ryan B. Lee, Dmitry A. Rodionov, Jaime E. Zlamal, David Scott, Daniel J. Chen, Lisa Elmén, and Alexandre R. Colas
- Subjects
Microbiology (medical) ,lcsh:QR1-502 ,microbiome ,Butyrate ,Gut flora ,Microbiology ,lcsh:Microbiology ,Clostridia ,bacterial genome reconstruction ,03 medical and health sciences ,chemistry.chemical_compound ,Glycerol monostearate ,Glycerol ,Clostridiaceae ,Food science ,Western diet ,030304 developmental biology ,0303 health sciences ,biology ,030306 microbiology ,Lachnospiraceae ,Sodium stearoyl lactylate ,dysbiosis ,biology.organism_classification ,food additives ,chemistry ,short chain fatty acids - Abstract
Dietary emulsifiers are widely used in industrially processed foods, although the effects of these food additives on human gut microbiota are not well studied. Here, we investigated the effects of five different emulsifiers [glycerol monoacetate, glycerol monostearate, glycerol monooleate, propylene glycol monostearate, and sodium stearoyl lactylate (SSL)] on fecal microbiota in vitro. We found that 0.025% (w/v) of SSL reduced the relative abundance of the bacterial class Clostridia and others. The relative abundance of the families Clostridiaceae, Lachnospiraceae, and Ruminococcaceae was substantially reduced whereas that of Bacteroidaceae and Enterobacteriaceae was increased. Given the marked impact of SSL on Clostridia, we used genome reconstruction to predict community-wide production of short-chain fatty acids, which were experimentally assessed by GC-MS analysis. SSL significantly reduced concentrations of butyrate, and increased concentrations of propionate compared to control cultures. The presence of SSL increased lipopolysaccharide, LPS and flagellin in cultured communities, thereby enhancing the proinflammatory potential of SSL-selected bacterial communities.
- Published
- 2020
26. Lipid availability influences the metabolic maturation of human pluripotent stem cell-derived cardiomyocytes
- Author
-
Hui Zhang, Christian M. Metallo, Sean Spiering, Mark Mercola, Anne N. Murphy, Michael S. Yu, Alexandre R. Colas, Divakaruni Ajit Srinivas, Mehmet G. Badur, and Noah Meurs
- Subjects
0303 health sciences ,Chemistry ,Metabolism ,030204 cardiovascular system & hematology ,In vitro maturation ,Cell biology ,Transcriptome ,03 medical and health sciences ,0302 clinical medicine ,Metabolic flux analysis ,Ketone bodies ,Induced pluripotent stem cell ,Beta oxidation ,Function (biology) ,030304 developmental biology - Abstract
ObjectivesPluripotent stem cell-derived cardiomyocytes are phenotypically immature, which limits their utility in downstream applications. Metabolism is dramatically reprogramed during cardiac maturationin vivoand presents a potential avenue to drivein vitromaturation. We aimed to identify and address metabolic bottlenecks in the generation of human pluripotent stem cell (hPSC)-derived cardiomyocytes.MethodshPSCs were differentiated into cardiomyocytes using an established, chemically-defined differentiation protocol. We applied 13C metabolic flux analysis (MFA) and targeted transcriptomics to characterize cardiomyocyte metabolism in during differentiation in the presence or absence of exogenous lipids.ResultshPSC-derived cardiomyocytes induced some cardiometabolic pathways (i.e. ketone body and branched-chain amino acid oxidation) but failed to effectively activate fatty acid oxidation. MFA studies indicated that lipid availability in cultures became limited during differentiation, suggesting potential issues with nutrient availability. Exogenous supplementation of lipids improved cardiomyocyte morphology, mitochondrial function, and promoted increased fatty acid oxidation in hPSC-derivatives.ConclusionhPSC-derived cardiomyocytes are dependent upon exogenous sources of lipids for metabolic maturation. Proper supplementation removes a potential roadblock in the generation of metabolically mature cardiomyocytes. These studies further highlight the importance of considering and exploiting metabolic phenotypes in thein vitroproduction and utilization of functional hPSC-derivatives.
- Published
- 2020
- Full Text
- View/download PDF
27. Silencing of CCR4-NOT complex subunits affects heart structure and function
- Author
-
Anaïs Kervadec, Karen Ocorr, Sreehari Kalvakuri, Rolf Bodmer, Santiago Pineda, Andrew A. Hicks, Claudia B. Volpato, Anthony Cammarato, Lisa Elmén, Peter P. Pramstaller, Nakissa N. Alayari, Alexandre R. Colas, Luisa Foco, and Alessandra Rossini
- Subjects
0301 basic medicine ,Medicine (miscellaneous) ,lcsh:Medicine ,Action Potentials ,030204 cardiovascular system & hematology ,Long-QT syndrome ,hiPSC ,Animals, Genetically Modified ,0302 clinical medicine ,Immunology and Microbiology (miscellaneous) ,Heart Rate ,Drosophila heart ,Gene expression ,Morphogenesis ,Drosophila Proteins ,GWAS ,Myocytes, Cardiac ,Cardiomyocytes ,Gene knockdown ,Heart development ,Intracellular Signaling Peptides and Proteins ,Gene Expression Regulation, Developmental ,RNA-Binding Proteins ,Cell biology ,Long QT Syndrome ,Drosophila melanogaster ,Arrhythmia ,lcsh:RB1-214 ,Research Article ,Induced Pluripotent Stem Cells ,Neuroscience (miscellaneous) ,Biology ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Ribonucleases ,CCR4-NOT complex ,lcsh:Pathology ,Gene silencing ,Animals ,Humans ,Gene Silencing ,Gene ,Cell Proliferation ,Messenger RNA ,lcsh:R ,Promoter ,Dros ,Repressor Proteins ,030104 developmental biology ,Exoribonucleases ,CNOT1 ,Genome-Wide Association Study ,HeLa Cells ,Transcription Factors - Abstract
The identification of genetic variants that predispose individuals to cardiovascular disease and a better understanding of their targets would be highly advantageous. Genome-wide association studies have identified variants that associate with QT-interval length (a measure of myocardial repolarization). Three of the strongest associating variants (single-nucleotide polymorphisms) are located in the putative promotor region of CNOT1, a gene encoding the central CNOT1 subunit of CCR4-NOT: a multifunctional, conserved complex regulating gene expression and mRNA stability and turnover. We isolated the minimum fragment of the CNOT1 promoter containing all three variants from individuals homozygous for the QT risk alleles and demonstrated that the haplotype associating with longer QT interval caused reduced reporter expression in a cardiac cell line, suggesting that reduced CNOT1 expression might contribute to abnormal QT intervals. Systematic siRNA-mediated knockdown of CCR4-NOT components in human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) revealed that silencing CNOT1 and other CCR4-NOT genes reduced their proliferative capacity. Silencing CNOT7 also shortened action potential duration. Furthermore, the cardiac-specific knockdown of Drosophila orthologs of CCR4-NOT genes in vivo (CNOT1/Not1 and CNOT7/8/Pop2) was either lethal or resulted in dilated cardiomyopathy, reduced contractility or a propensity for arrhythmia. Silencing CNOT2/Not2, CNOT4/Not4 and CNOT6/6L/twin also affected cardiac chamber size and contractility. Developmental studies suggested that CNOT1/Not1 and CNOT7/8/Pop2 are required during cardiac remodeling from larval to adult stages. To summarize, we have demonstrated how disease-associated genes identified by GWAS can be investigated by combining human cardiomyocyte cell-based and whole-organism in vivo heart models. Our results also suggest a potential link of CNOT1 and CNOT7/8 to QT alterations and further establish a crucial role of the CCR4-NOT complex in heart development and function. This article has an associated First Person interview with the first author of the paper., Summary: Genome-wide association studies combined with in vitro human cardiac cell assays and a model organism suitable for heart studies in vivo connect CNOT1, CNOT7 and overall the CCR4-NOT complex to human heart disease and morbidity.
- Published
- 2020
28. PGC1/PPAR Drive Cardiomyocyte Maturation through Regulation of Yap1 and SF3B2
- Author
-
Sam Paek, Emmanouil Tampakakis, Renjun Zhu, Anaïs Kervadec, Chulan Kwon, David A. Kass, Sean Murphy, Brian L. Lin, Sandeep Kambhampati, Matthew Miyamoto, Suraj Kannan, Hideki Uosaki, Steven S. An, Peter Andersen, Alexandre R. Colas, and Dong Ik Lee
- Subjects
chemistry.chemical_classification ,YAP1 ,Cardiac muscle ,Regulator ,Gene regulatory network ,Peroxisome proliferator-activated receptor ,Biology ,Cell biology ,Contractility ,medicine.anatomical_structure ,chemistry ,medicine ,Myocyte ,Transcription factor - Abstract
Cardiomyocytes undergo significant levels of structural and functional changes after birth—fundamental processes essential for the heart to produce the volume and contractility to pump blood to the growing body. However, due to the challenges in isolating single postnatal/adult myocytes, how individual newborn cardiomyocytes acquire multiple aspects of mature phenotypes remains poorly understood. Here we implemented large-particle sorting and analyzed single myocytes from neonatal to adult hearts. Early myocytes exhibited a wide-ranging transcriptomic and size heterogeneity, maintained until adulthood with a continuous transcriptomic shift. Gene regulatory network analysis followed by mosaic gene deletion revealed that peroxisome proliferator-activated receptor coactivator-1 signaling—activated in vivo but inactive in pluripotent stem cell-derived cardiomyocytes—mediates the shift. The signaling regulated key aspects of cardiomyocyte maturation simultaneously through previously unrecognized regulators, including Yap1 and SF3B2. Our study provides a single-cell roadmap of heterogeneous transitions coupled to cellular features and unveils a multifaceted regulator controlling cardiomyocyte maturation.Significance StatementHow the individual single myocytes achieve full maturity remains a ‘black box’, largely due to the challenges with the isolation of single mature myocytes. Understanding this process is particularly important as the immaturity and early developmental arrest of pluripotent stem cell-derived myocytes has emerged a major concern in the field. Here we present the first study of high-quality single-cell transcriptomic analysis of cardiac muscle cells from neonatal to adult hearts. We identify a central transcription factor and its novel targets that control key aspects of myocyte maturation, including cellular hypertrophy, contractility, and mitochondrial activity.
- Published
- 2020
- Full Text
- View/download PDF
29. Acellular therapeutic approach for heart failure: in vitro production of extracellular vesicles from human cardiovascular progenitors
- Author
-
Solenne Paiva, Nadia El Harane, Jean-Sébastien Silvestre, Yeranuhi Hovhannisyan, Camille Brunaud, Léa Thiebault, Jeanne Gauthier, Onnik Agbulut, Marc P. Renault, Philippe Menasché, Hany J Neametalla, Marie-Cécile Perier, Nicolas Cagnard, Albert Hagège, Mathilde Lemitre, Bruna Lima Correa, Valérie Bellamy, Angéline Duché, Anaïs Kervadec, Alexandra T Bourdillon, Nisa Renault, Laetitia Pidial, and Alexandre R. Colas
- Subjects
Pluripotent Stem Cells ,0301 basic medicine ,Cell Survival ,Cell ,Myocardial Infarction ,Mice, Nude ,Pharmacology ,Extracellular Vesicles ,03 medical and health sciences ,Basic Science ,Animals ,Humans ,Myocyte ,Medicine ,Myocytes, Cardiac ,Progenitor cell ,Induced pluripotent stem cell ,Embryonic Stem Cells ,Cell Proliferation ,Heart Failure ,Tube formation ,business.industry ,Cell growth ,Embryonic stem cell ,Endothelial stem cell ,MicroRNAs ,Treatment Outcome ,030104 developmental biology ,medicine.anatomical_structure ,Cardiology and Cardiovascular Medicine ,business - Abstract
Aims We have shown that extracellular vesicles (EVs) secreted by embryonic stem cell-derived cardiovascular progenitor cells (Pg) recapitulate the therapeutic effects of their parent cells in a mouse model of chronic heart failure (CHF). Our objectives are to investigate whether EV released by more readily available cell sources are therapeutic, whether their effectiveness is influenced by the differentiation state of the secreting cell, and through which mechanisms they act. Methods and results The total EV secreted by human induced pluripotent stem cell-derived cardiovascular progenitors (iPSC-Pg) and human induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) were isolated by ultracentrifugation and characterized by Nanoparticle Tracking Analysis, western blot, and cryo-electron microscopy. In vitro bioactivity assays were used to evaluate their cellular effects. Cell and EV microRNA (miRNA) content were assessed by miRNA array. Myocardial infarction was induced in 199 nude mice. Three weeks later, mice with left ventricular ejection fraction (LVEF) ≤ 45% received transcutaneous echo-guided injections of iPSC-CM (1.4 × 106, n = 19), iPSC-Pg (1.4 × 106, n = 17), total EV secreted by 1.4 × 106 iPSC-Pg (n = 19), or phosphate-buffered saline (control, n = 17) into the peri-infarct myocardium. Seven weeks later, hearts were evaluated by echocardiography, histology, and gene expression profiling, blinded to treatment group. In vitro, EV were internalized by target cells, increased cell survival, cell proliferation, and endothelial cell migration in a dose-dependent manner and stimulated tube formation. Extracellular vesicles were rich in miRNAs and most of the 16 highly abundant, evolutionarily conserved miRNAs are associated with tissue-repair pathways. In vivo, EV outperformed cell injections, significantly improving cardiac function through decreased left ventricular volumes (left ventricular end systolic volume: -11%, P
- Published
- 2018
30. Patient-specific functional genomics and disease modeling suggest a role for LRP2 in hypoplastic left heart syndrome
- Author
-
Xing Li, Jared M. Evans, Karen Ocorr, Stanley M. Walls, Georg Vogler, James N. Kezos, Rolf Bodmer, Almudena Martinez-Fernandez, Katja Birker, Megan M. O’Byrne, Andre Terzic, Paul Grossfeld, Jeanne L. Theis, Timothy J. Nelson, Tanja Nielsen, Maria A. Missinato, Zeng Xi, Alexandre R. Colas, Zachary C. Fogarty, Timothy M. Olson, and Anaïs Kervadec
- Subjects
Genetics ,Gene knockdown ,Candidate gene ,Heart disease ,medicine ,Biology ,Induced pluripotent stem cell ,medicine.disease ,Gene ,Functional genomics ,Phenotype ,Hypoplastic left heart syndrome - Abstract
Congenital heart diseases (CHD), such as hypoplastic left heart syndrome (HLHS), are considered to have complex genetic underpinnings that are poorly understood. Here, an integrated multi-disciplinary approach was applied to identify novel genes and underlying mechanisms associated with HLHS. A family-based strategy was employed that coupled whole genome sequencing (WGS) with RNA sequencing of patient-derived induced pluripotent stem cells (iPSCs) from a sporadic HLHS proband-parent trio to identify, prioritize and functionally evaluate candidate genes in model systems. Consistent with the hypoplastic phenotype, the proband’s iPSCs had reduced proliferation capacity. Filtering WGS for rare de novo, recessive, and loss-of-function variants revealed 10 candidate genes with recessive variants and altered expression compared to the parents’ iPSCs. siRNA/RNAi-mediated knockdown in generic human iPSC-derived cardiac progenitors and in thein vivo Drosophilaheart model revealed that LDL receptor related proteinLRP2and apolipoproteinAPOBare required for robust hiPSC-derived cardiomyocyte proliferation and normal hear structure and function, possibly involving an oligogenic mechanism via growth-promoting WNT and SHH signaling.LRP2was further validated as a CHD gene in a zebrafish heart model and rare variant burden testing in an HLHS cohort. Collectively, this cross-functional genetic approach to complex congenital heart disease revealed LRP2 dysfunction as a likely novel genetic driver of HLHS, and hereby established a scalable approach to decipher the oligogenic underpinnings of maladaptive left heart development.One sentence summaryWhole genome sequencing and a multi-model system candidate gene validation - human iPSC-derived cardiomyocytes andDrosophilaand zebrafish hearts - identified lipoproteinLRP2as a new potential driver in congenital heart disease and suggests a deficit in proliferation as a hallmark of hypoplastic left heart syndrome.
- Published
- 2019
31. Genetic architecture of natural variations of cardiac performances in flies: Conserved features with humans
- Author
-
Lionel Spinelli, Laurence Röder, Laurent Kremmer, Karen Ocorr, J.A. Castro Mondragon, S. Krifa, Anaïs Kervadec, Christine Brun, Rolf Bodmer, S. Saha, Laurent Perrin, and Alexandre R. Colas
- Subjects
Phenotypic plasticity ,biology ,business.industry ,Genome-wide association study ,Computational biology ,biology.organism_classification ,Genome ,Genetic architecture ,Epistasis ,Medicine ,Cardiology and Cardiovascular Medicine ,business ,Drosophila ,Gene ,Function (biology) - Abstract
Introduction Deciphering the genetic architecture of cardiac disorders is of fundamental importance but their underlying complexity is a major hurdle. Drosophila has gained importance as a useful model to study heart development and function and allows the analysis of organismal traits in a physiologically relevant and accessible system. Objectives Our aim was to (i) identify in flies the variants associated to natural variations of cardiac performances among a natural population, (ii) decipher how these variants interact with each other and with the environment to impact cardiac traits, (iii) gain insights about the molecular and cellular processes affected, (iv) determine whether the genetic architecture of cardiac disorders is conserved with humans. Methods We used the Drosophila Genetic Reference Panel, a community resource of sequenced inbred lines. Genome Wide Associations (GWA) for single markers and epistatic interactions were analyzed on cardiac traits related to rhythm and contractility. Results Genetic networks were unraveled and extensively validated in vivo. Non-coding variants were used to map potential regulatory non-coding regions and to predict Transcription Factors (TFs) binding sites. Cognate TFs were validated by heart specific knockdown. Natural variations of cardiac traits variance revealed unique features of the phenotypic plasticity of cardiac performance. Importantly, correlations between genes associated to GWAS for cardiac traits both in flies and humans supported conservation of the genetic architecture of cardiac disorders from arthropods to mammals. In addition, several conserved genes and pathways were validated in human iPSC-derived cardiomyocytes. Conclusion We provide an in-depth analysis of the genetic architecture of natural variations of cardiac performance in flies. Our data may guide the analysis of cardiac disorders in humans.
- Published
- 2021
32. Small-molecule probe reveals a kinase cascade that links stress signaling to TCF/LEF and Wnt responsiveness
- Author
-
Isaac Perea-Gil, Joseph C. Wu, Joseph J. Lancman, Karl J. Okolotowicz, Travis L. Biechele, Paul J. Bushway, P. Duc Si Dong, Randall T. Moon, Justine Quach, Arne A. N. Bruyneel, Wesley L. McKeithan, Jiongjia Cheng, Mark Mercola, Jaechol Lee, Nirmal Vadgama, Mary Dwyer, Ioannis Karakikes, John R. Cashman, Masanao Tsuda, Dennis Schade, and Alexandre R. Colas
- Subjects
Male ,Xenopus ,Clinical Biochemistry ,Morphogenesis ,Genotoxic Stress ,Protein Serine-Threonine Kinases ,01 natural sciences ,Biochemistry ,Article ,Small Molecule Libraries ,Neoplasms ,Drug Discovery ,Animals ,Humans ,Wnt Signaling Pathway ,Molecular Biology ,Gene ,Mitosis ,Cells, Cultured ,Zebrafish ,beta Catenin ,Pharmacology ,biology ,010405 organic chemistry ,Mechanism (biology) ,Wnt signaling pathway ,Small molecule ,0104 chemical sciences ,Cell biology ,Tubulin ,Molecular Probes ,biology.protein ,Molecular Medicine ,Tumor Suppressor Protein p53 ,TCF Transcription Factors - Abstract
Summary Wnt signaling plays a central role in tissue maintenance and cancer. Wnt activates downstream genes through β-catenin, which interacts with TCF/LEF transcription factors. A major question is how this signaling is coordinated relative to tissue organization and renewal. We used a recently described class of small molecules that binds tubulin to reveal a molecular cascade linking stress signaling through ATM, HIPK2, and p53 to the regulation of TCF/LEF transcriptional activity. These data suggest a mechanism by which mitotic and genotoxic stress can indirectly modulate Wnt responsiveness to exert coherent control over cell shape and renewal. These findings have implications for understanding tissue morphogenesis and small-molecule anticancer therapeutics.
- Published
- 2021
33. Early molecular events during retinoic acid induced differentiation of neuromesodermal progenitors
- Author
-
Alexandre R. Colas, Gregg Duester, and Thomas J. Cunningham
- Subjects
0301 basic medicine ,Embryonic stem cells ,QH301-705.5 ,Science ,Retinoic acid ,Nkx1-2 ,Raldh2 knockout embryos ,Id1 ,Biology ,Gbx2 ,General Biochemistry, Genetics and Molecular Biology ,Transcriptome ,03 medical and health sciences ,chemistry.chemical_compound ,FGF8 ,SOX2 ,Paraxial mesoderm ,Biology (General) ,Genetics ,Zfp503 ,Zfp703 ,Neuroectoderm ,Neuromesodermal progenitors ,Wnt signaling pathway ,Retinoic acid response elements ,3. Good health ,Cell biology ,Retinoic acid target genes ,030104 developmental biology ,chemistry ,Epiblast ,embryonic structures ,General Agricultural and Biological Sciences ,Research Article - Abstract
Bipotent neuromesodermal progenitors (NMPs) residing in the caudal epiblast drive coordinated body axis extension by generating both posterior neuroectoderm and presomitic mesoderm. Retinoic acid (RA) is required for body axis extension, however the early molecular response to RA signaling is poorly defined, as is its relationship to NMP biology. As endogenous RA is first seen near the time when NMPs appear, we used WNT/FGF agonists to differentiate embryonic stem cells to NMPs which were then treated with a short 2-h pulse of 25 nM RA or 1 µM RA followed by RNA-seq transcriptome analysis. Differential expression analysis of this dataset indicated that treatment with 25 nM RA, but not 1 µM RA, provided physiologically relevant findings. The 25 nM RA dataset yielded a cohort of previously known caudal RA target genes including Fgf8 (repressed) and Sox2 (activated), plus novel early RA signaling targets with nearby conserved RA response elements. Importantly, validation of top-ranked genes in vivo using RA-deficient Raldh2−/− embryos identified novel examples of RA activation (Nkx1-2, Zfp503, Zfp703, Gbx2, Fgf15, Nt5e) or RA repression (Id1) of genes expressed in the NMP niche or progeny. These findings provide evidence for early instructive and permissive roles of RA in controlling differentiation of NMPs to neural and mesodermal lineages., Summary: The findings here demonstrate that the signaling molecule retinoic acid (RA) plays an early role in determining how embryonic progenitor cells decide to differentiate into either mesodermal or neural tissues.
- Published
- 2016
34. Generation of First Heart Field-like Cardiac Progenitors and Ventricular-like Cardiomyocytes from Human Pluripotent Stem Cells
- Author
-
Michael S. Yu, Sean Spiering, and Alexandre R. Colas
- Subjects
0301 basic medicine ,Pluripotent Stem Cells ,Cardiac progenitors ,General Immunology and Microbiology ,General Chemical Engineering ,General Neuroscience ,Heart Ventricles ,Cell ,030105 genetics & heredity ,Biology ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,Activin a ,03 medical and health sciences ,medicine.anatomical_structure ,medicine ,Humans ,Myocytes, Cardiac ,Progenitor cell ,Induced pluripotent stem cell ,Developmental biology ,Developmental Biology - Abstract
The generation of large amounts of functional human pluripotent stem cells-derived cardiac progenitors and cardiomyocytes of defined heart field origin is a pre-requisite for cell-based cardiac therapies and disease modeling. We have recently shown that Id genes are both necessary and sufficient to specify first heart field progenitors during vertebrate development. This differentiation protocol leverages these findings and uses Id1 overexpression in combination with Activin A as potent specifying cues to produce first heart field-like (FHF-L) progenitors. Importantly, resulting progenitors efficiently differentiate (~70–90%) into ventricular-like cardiomyocytes. Here we describe a detailed method to 1) generate Id1-overexpressing hPSCs and 2) differentiate scalable quantities of cryopreservable FHF-L progenitors and ventricular-like cardiomyocytes.
- Published
- 2018
35. An Automated Platform for Assessment of Congenital and Drug-Induced Arrhythmia with hiPSC-Derived Cardiomyocytes
- Author
-
John R. Cashman, Jeffery H Price, Mark Mercola, Michael S. Yu, Alexandre R. Colas, Wesley L. McKeithan, Arne A. N. Bruyneel, Alex Savchenko, Evan W. Miller, and Fabio Cerignoli
- Subjects
0301 basic medicine ,induced pluripotent stem cells ,Physiology ,Computer science ,High-throughput screening ,cardiotoxicity ,cardiomyocyte ,Computational biology ,Pharmacology ,Drug induced arrhythmia ,lcsh:Physiology ,03 medical and health sciences ,Physiology (medical) ,Methods ,medicine ,Induced pluripotent stem cell ,Proarrhythmia ,lcsh:QP1-981 ,Drug discovery ,medicine.disease ,drug development ,CiPA ,030104 developmental biology ,voltage sensitive probe ,Drug development ,Voltage sensing ,Robust analysis ,high throughput screening - Abstract
The ability to produce unlimited numbers of human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CMs) harboring disease and patient-specific gene variants creates a new paradigm for modeling congenital heart diseases (CHDs) and predicting proarrhythmic liabilities of drug candidates. However, a major roadblock to implementing hiPSC-CM technology in drug discovery is that conventional methods for monitoring action potential (AP) kinetics and arrhythmia phenotypes in vitro have been too costly or technically challenging to execute in high throughput. Herein, we describe the first large-scale, fully automated and statistically robust analysis of AP kinetics and drug-induced proarrhythmia in hiPSC-CMs. The platform combines the optical recording of a small molecule fluorescent voltage sensing probe (VoltageFluor2.1.Cl), an automated high throughput microscope and automated image analysis to rapidly generate physiological measurements of cardiomyocytes (CMs). The technique can be readily adapted on any high content imager to study hiPSC-CM physiology and predict the proarrhythmic effects of drug candidates.
- Published
- 2017
36. Id genes are essential for early heart formation
- Author
-
Pier Lorenzo Puri, Sonia Albini, Gregg Duester, Sean Spiering, Pilar Ruiz-Lozano, Paul J. Bushway, Alessandra Sacco, Mark Mercola, Miguel Mano, Jean-François Riou, Wesley L. McKeithan, Mauro Giacca, Michael S. Yu, Chun-Teng Huang, Alexandre R. Colas, Matthew T. Tierney, Florent Carrette, Thomas J. Cunningham, Muriel Umbhauer, Sanford Burnham Prebys Medical Discovery Institute, Department of Bioengineering, University of California [San Diego] (UC San Diego), University of California (UC)-University of California (UC), Stanford University, International Centre for Genetic Engineering and Biotechnology (ICGEB) (Trieste), University of Coimbra [Portugal] (UC), Signalisation et morphogenèse = Signalling and morphogenesis (LBD-E12), Laboratoire de Biologie du Développement (LBD), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut de Biologie Paris Seine (IBPS), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre National de la Recherche Scientifique (CNRS)-Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut de Biologie Paris Seine (IBPS), Université Pierre et Marie Curie - Paris 6 (UPMC)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Santé et de la Recherche Médicale (INSERM)-Centre National de la Recherche Scientifique (CNRS)-Centre National de la Recherche Scientifique (CNRS), Sanford Burnham Medical Research Institute, La Jolla, and University of California-University of California
- Subjects
0301 basic medicine ,Embryo, Nonmammalian ,Organogenesis ,Bioinformatics ,Regenerative medicine ,Mesoderm ,Mice ,Xenopus laevis ,cardiac progenitors ,CRISPR/Cas9-mediated quadruple knockout ,Basic Helix-Loop-Helix Transcription Factors ,CRISPR ,Heart formation ,11 Medical and Health Sciences ,Genetics & Heredity ,Gene Editing ,platform for cardiac disease modeling and drug discovery ,Drug discovery ,Gene Expression Regulation, Developmental ,Cell Differentiation ,Heart ,cardiac mesoderm specification ,17 Psychology and Cognitive Sciences ,3. Good health ,Cell biology ,embryonic structures ,Seeds ,MESP1 ,Life Sciences & Biomedicine ,Research Paper ,Heart Defects, Congenital ,animal structures ,PROTEINS ,CARDIOVASCULAR PROGENITOR CELLS ,heartless ,Biology ,Cell Line ,WNT ,03 medical and health sciences ,MOUSE GASTRULATION ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,Genetics ,XENOPUS-LAEVIS ,Animals ,Humans ,Cell Lineage ,Progenitor cell ,Id proteins ,Embryonic Stem Cells ,Science & Technology ,Embryogenesis ,Cell Biology ,MAMMALIAN HEART ,06 Biological Sciences ,Embryo, Mammalian ,Embryonic stem cell ,MYOCARDIAL-CELLS ,[SDV.BDD.EO]Life Sciences [q-bio]/Development Biology/Embryology and Organogenesis ,030104 developmental biology ,Mesoderm formation ,Mutation ,Inhibitor of Differentiation Proteins ,EMBRYONIC STEM-CELLS ,Developmental Biology - Abstract
International audience; Deciphering the fundamental mechanisms controlling cardiac specification is critical for our understanding of how heart formation is initiated during embryonic development and for applying stem cell biology to regenerative medicine and disease modeling. Using systematic and unbiased functional screening approaches, we discovered that the Id family of helix–loop–helix proteins is both necessary and sufficient to direct cardiac mesoderm formation in frog embryos and human embryonic stem cells. Mechanistically, Id proteins specify cardiac cell fate by repressing two inhibitors of cardiogenic mesoderm formation—Tcf3 and Foxa2—and activating inducers Evx1, Grrp1, and Mesp1. Most importantly, CRISPR/Cas9-mediated ablation of the entire Id (Id1–4) family in mouse embryos leads to failure of anterior cardiac progenitor specification and the development of heartless embryos. Thus, Id proteins play a central and evolutionarily conserved role during heart formation and provide a novel means to efficiently produce cardiovascular progenitors for regenerative medicine and drug discovery applications.
- Published
- 2017
37. Retinoic Acid Activity in Undifferentiated Neural Progenitors Is Sufficient to Fulfill Its Role in Restricting Fgf8 Expression for Somitogenesis
- Author
-
Gregg Duester, Mark Lewandoski, Lisa L. Sandell, Mark Mercola, Paul A. Trainor, Thomas J. Cunningham, Thomas Brade, and Alexandre R. Colas
- Subjects
medicine.medical_specialty ,Mesoderm ,animal structures ,Fibroblast Growth Factor 8 ,Neurogenesis ,TBX6 ,lcsh:Medicine ,Tretinoin ,Biology ,Embryo Culture Techniques ,Mice ,Somitogenesis ,Internal medicine ,medicine ,Paraxial mesoderm ,Animals ,lcsh:Science ,Neural Plate ,Multidisciplinary ,lcsh:R ,Gene Expression Regulation, Developmental ,Aldehyde Oxidoreductases ,ddc ,Cell biology ,Somite ,Endocrinology ,medicine.anatomical_structure ,Neurulation ,Somites ,Epiblast ,embryonic structures ,lcsh:Q ,Neural plate ,Research Article - Abstract
Bipotent axial stem cells residing in the caudal epiblast during late gastrulation generate neuroectodermal and presomitic mesodermal progeny that coordinate somitogenesis with neural tube formation, but the mechanism that controls these two fates is not fully understood. Retinoic acid (RA) restricts the anterior extent of caudal fibroblast growth factor 8 (Fgf8) expression in both mesoderm and neural plate to control somitogenesis and neurogenesis, however it remains unclear where RA acts to control the spatial expression of caudal Fgf8. Here, we found that mouse Raldh2-/- embryos, lacking RA synthesis and displaying a consistent small somite defect, exhibited abnormal expression of key markers of axial stem cell progeny, with decreased Sox2+ and Sox1+ neuroectodermal progeny and increased Tbx6+ presomitic mesodermal progeny. The Raldh2-/- small somite defect was rescued by treatment with an FGF receptor antagonist. Rdh10 mutants, with a less severe RA synthesis defect, were found to exhibit a small somite defect and anterior expansion of caudal Fgf8 expression only for somites 1-6, with normal somite size and Fgf8 expression thereafter. Rdh10 mutants were found to lack RA activity during the early phase when somites are small, but at the 6-somite stage RA activity was detected in neural plate although not in presomitic mesoderm. Expression of a dominant-negative RA receptor in mesoderm eliminated RA activity in presomitic mesoderm but did not affect somitogenesis. Thus, RA activity in the neural plate is sufficient to prevent anterior expansion of caudal Fgf8 expression associated with a small somite defect. Our studies provide evidence that RA restriction of Fgf8 expression in undifferentiated neural progenitors stimulates neurogenesis while also restricting the anterior extent of the mesodermal Fgf8 mRNA gradient that controls somite size, providing new insight into the mechanism that coordinates somitogenesis with neurogenesis.
- Published
- 2015
38. Whole-genome microRNA screening identifies let-7 and mir-18 as regulators of germ layer formation during early embryogenesis
- Author
-
Gregg Duester, Alexandre R. Colas, Thomas J. Cunningham, Paul J. Bushway, Shankar Subramaniam, Mark Mercola, Wesley L. McKeithan, and Lana X. Garmire
- Subjects
Mesoderm ,animal structures ,DNA Mutational Analysis ,Embryonic Development ,Ectoderm ,Germ layer ,Histogenesis ,Biology ,FGF and mesoderm formation ,Mice ,Xenopus laevis ,Genetics ,medicine ,Animals ,Cells, Cultured ,Embryonic Stem Cells ,Genome ,Gene Expression Regulation, Developmental ,MicroRNAs ,medicine.anatomical_structure ,Epiblast ,Gene Knockdown Techniques ,embryonic structures ,Endoderm ,NODAL ,Germ Layers ,Developmental Biology ,Research Paper - Abstract
Tight control over the segregation of endoderm, mesoderm, and ectoderm is essential for normal embryonic development of all species, yet how neighboring embryonic blastomeres can contribute to different germ layers has never been fully explained. We postulated that microRNAs, which fine-tune many biological processes, might modulate the response of embryonic blastomeres to growth factors and other signals that govern germ layer fate. A systematic screen of a whole-genome microRNA library revealed that the let-7 and miR-18 families increase mesoderm at the expense of endoderm in mouse embryonic stem cells. Both families are expressed in ectoderm and mesoderm, but not endoderm, as these tissues become distinct during mouse and frog embryogenesis. Blocking let-7 function in vivo dramatically affected cell fate, diverting presumptive mesoderm and ectoderm into endoderm. siRNA knockdown of computationally predicted targets followed by mutational analyses revealed that let-7 and miR-18 down-regulate Acvr1b and Smad2, respectively, to attenuate Nodal responsiveness and bias blastomeres to ectoderm and mesoderm fates. These findings suggest a crucial role for the let-7 and miR-18 families in germ layer specification and reveal a remarkable conservation of function from amphibians to mammals.
- Published
- 2012
39. Alternative splicing regulates mouse embryonic stem cell pluripotency and differentiation
- Author
-
Laura Pereira, Tyson A. Clark, Bruce R. Conklin, Christine Wahlquist, Matthew J. Spindler, Alexander C. Zambon, Alexander R. Pico, Eva Samal, Alexandre R. Colas, Mark Mercola, Melissa S. Cline, Christopher R. Schlieve, John E. Blume, Karen Vranizan, Bradley J. Merrill, Alan Williams, and Nathan Salomonis
- Subjects
Pluripotent Stem Cells ,Transcription, Genetic ,Cellular differentiation ,Biology ,Kruppel-Like Factor 4 ,Mice ,RNA Isoforms ,Animals ,Humans ,Selection, Genetic ,Induced pluripotent stem cell ,Promoter Regions, Genetic ,Embryonic Stem Cells ,Genetics ,Multidisciplinary ,Gene Expression Profiling ,Alternative splicing ,Wnt signaling pathway ,Cell Differentiation ,Exons ,Biological Sciences ,Embryonic stem cell ,Cell biology ,Gene expression profiling ,Wnt Proteins ,Alternative Splicing ,MicroRNAs ,Gene Expression Regulation ,KLF4 ,Female ,Signal Transduction - Abstract
Two major goals of regenerative medicine are to reproducibly transform adult somatic cells into a pluripotent state and to control their differentiation into specific cell fates. Progress toward these goals would be greatly helped by obtaining a complete picture of the RNA isoforms produced by these cells due to alternative splicing (AS) and alternative promoter selection (APS). To investigate the roles of AS and APS, reciprocal exon–exon junctions were interrogated on a genome-wide scale in differentiating mouse embryonic stem (ES) cells with a prototype Affymetrix microarray. Using a recently released open-source software package named AltAnalyze, we identified 144 genes for 170 putative isoform variants, the majority (67%) of which were predicted to alter protein sequence and domain composition. Verified alternative exons were largely associated with pathways of Wnt signaling and cell-cycle control, and most were conserved between mouse and human. To examine the functional impact of AS, we characterized isoforms for two genes. As predicted by AltAnalyze, we found that alternative isoforms of the gene Serca2 were targeted by distinct microRNAs (miRNA-200b, miRNA-214), suggesting a critical role for AS in cardiac development. Analysis of the Wnt transcription factor Tcf3, using selective knockdown of an ES cell-enriched and characterized isoform, revealed several distinct targets for transcriptional repression (Stmn2, Ccnd2, Atf3, Klf4, Nodal, and Jun) as well as distinct differentiation outcomes in ES cells. The findings herein illustrate a critical role for AS in the specification of ES cells with differentiation, and highlight the utility of global functional analyses of AS.
- Published
- 2010
40. Mitochondrial MICOS complex genes, implicated in hypoplastic left heart syndrome, maintain cardiac contractility and actomyosin integrity
- Author
-
Katja Birker, Shuchao Ge, Natalie J Kirkland, Jeanne L Theis, James Marchant, Zachary C Fogarty, Maria A Missinato, Sreehari Kalvakuri, Paul Grossfeld, Adam J Engler, Karen Ocorr, Timothy J Nelson, Alexandre R Colas, Timothy M Olson, Georg Vogler, and Rolf Bodmer
- Subjects
MICOS ,CHCHD3/6 ,HLHS ,Drosophila ,genetics ,CHD ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Hypoplastic left heart syndrome (HLHS) is a severe congenital heart disease (CHD) with a likely oligogenic etiology, but our understanding of the genetic complexities and pathogenic mechanisms leading to HLHS is limited. We performed whole genome sequencing (WGS) on 183 HLHS patient-parent trios to identify candidate genes, which were functionally tested in the Drosophila heart model. Bioinformatic analysis of WGS data from an index family of a HLHS proband born to consanguineous parents prioritized 9 candidate genes with rare, predicted damaging homozygous variants. Of them, cardiac-specific knockdown (KD) of mitochondrial MICOS complex subunit dCHCHD3/6 resulted in drastically compromised heart contractility, diminished levels of sarcomeric actin and myosin, reduced cardiac ATP levels, and mitochondrial fission-fusion defects. These defects were similar to those inflicted by cardiac KD of ATP synthase subunits of the electron transport chain (ETC), consistent with the MICOS complex’s role in maintaining cristae morphology and ETC assembly. Five additional HLHS probands harbored rare, predicted damaging variants in CHCHD3 or CHCHD6. Hypothesizing an oligogenic basis for HLHS, we tested 60 additional prioritized candidate genes from these patients for genetic interactions with CHCHD3/6 in sensitized fly hearts. Moderate KD of CHCHD3/6 in combination with Cdk12 (activator of RNA polymerase II), RNF149 (goliath, E3 ubiquitin ligase), or SPTBN1 (β-Spectrin, scaffolding protein) caused synergistic heart defects, suggesting the likely involvement of diverse pathways in HLHS. Further elucidation of novel candidate genes and genetic interactions of potentially disease-contributing pathways is expected to lead to a better understanding of HLHS and other CHDs.
- Published
- 2023
- Full Text
- View/download PDF
41. Genetic architecture of natural variation of cardiac performance from flies to humans
- Author
-
Saswati Saha, Lionel Spinelli, Jaime A Castro Mondragon, Anaïs Kervadec, Michaela Lynott, Laurent Kremmer, Laurence Roder, Sallouha Krifa, Magali Torres, Christine Brun, Georg Vogler, Rolf Bodmer, Alexandre R Colas, Karen Ocorr, and Laurent Perrin
- Subjects
heart function ,GWAS ,gene regulatory networks ,Drosophila ,human ,conserved genetic architecture ,Medicine ,Science ,Biology (General) ,QH301-705.5 - Abstract
Deciphering the genetic architecture of human cardiac disorders is of fundamental importance but their underlying complexity is a major hurdle. We investigated the natural variation of cardiac performance in the sequenced inbred lines of the Drosophila Genetic Reference Panel (DGRP). Genome-wide associations studies (GWAS) identified genetic networks associated with natural variation of cardiac traits which were used to gain insights as to the molecular and cellular processes affected. Non-coding variants that we identified were used to map potential regulatory non-coding regions, which in turn were employed to predict transcription factors (TFs) binding sites. Cognate TFs, many of which themselves bear polymorphisms associated with variations of cardiac performance, were also validated by heart-specific knockdown. Additionally, we showed that the natural variations associated with variability in cardiac performance affect a set of genes overlapping those associated with average traits but through different variants in the same genes. Furthermore, we showed that phenotypic variability was also associated with natural variation of gene regulatory networks. More importantly, we documented correlations between genes associated with cardiac phenotypes in both flies and humans, which supports a conserved genetic architecture regulating adult cardiac function from arthropods to mammals. Specifically, roles for PAX9 and EGR2 in the regulation of the cardiac rhythm were established in both models, illustrating that the characteristics of natural variations in cardiac function identified in Drosophila can accelerate discovery in humans.
- Published
- 2022
- Full Text
- View/download PDF
42. AlleleProfileR: A versatile tool to identify and profile sequence variants in edited genomes.
- Author
-
Arne A N Bruyneel, Alexandre R Colas, Ioannis Karakikes, and Mark Mercola
- Subjects
Medicine ,Science - Abstract
Gene editing strategies, such as zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9), are revolutionizing biology. However, quantitative and sensitive detection of targeted mutations are required to evaluate and quantify the genome editing outcomes. Here we present AlleleProfileR, a new analysis tool, written in a combination of R and C++, with the ability to batch process the sequence analysis of large and complex genome editing experiments, including the recently developed base editing technologies.
- Published
- 2019
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.