Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
Add more filters










Publication year range
1.
Nature ; 627(8005): 854-864, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38480880

ABSTRACT

The heart, which is the first organ to develop, is highly dependent on its form to function1,2. However, how diverse cardiac cell types spatially coordinate to create the complex morphological structures that are crucial for heart function remains unclear. Here we integrated single-cell RNA-sequencing with high-resolution multiplexed error-robust fluorescence in situ hybridization to resolve the identity of the cardiac cell types that develop the human heart. This approach also provided a spatial mapping of individual cells that enables illumination of their organization into cellular communities that form distinct cardiac structures. We discovered that many of these cardiac cell types further specified into subpopulations exclusive to specific communities, which support their specialization according to the cellular ecosystem and anatomical region. In particular, ventricular cardiomyocyte subpopulations displayed an unexpected complex laminar organization across the ventricular wall and formed, with other cell subpopulations, several cellular communities. Interrogating cell-cell interactions within these communities using in vivo conditional genetic mouse models and in vitro human pluripotent stem cell systems revealed multicellular signalling pathways that orchestrate the spatial organization of cardiac cell subpopulations during ventricular wall morphogenesis. These detailed findings into the cellular social interactions and specialization of cardiac cell types constructing and remodelling the human heart offer new insights into structural heart diseases and the engineering of complex multicellular tissues for human heart repair.


Subject(s)
Body Patterning , Heart , Myocardium , Animals , Humans , Mice , Heart/anatomy & histology , Heart/embryology , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Ventricles/anatomy & histology , Heart Ventricles/cytology , Heart Ventricles/embryology , In Situ Hybridization, Fluorescence , Models, Animal , Myocardium/cytology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Single-Cell Gene Expression Analysis
2.
Nature ; 622(7981): 37-39, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37759109

Subject(s)
Heart , Heart Rate
3.
Front Physiol ; 14: 1126111, 2023.
Article in English | MEDLINE | ID: mdl-36960155

ABSTRACT

Mechanosignaling describes processes by which biomechanical stimuli are transduced into cellular responses. External biophysical forces can be transmitted via structural protein networks that span from the cellular membrane to the cytoskeleton and the nucleus, where they can regulate gene expression through a series of biomechanical and/or biochemical mechanosensitive mechanisms, including chromatin remodeling, translocation of transcriptional regulators, and epigenetic factors. Striated muscle cells, including cardiac and skeletal muscle myocytes, utilize these nuclear mechanosignaling mechanisms to respond to changes in their intracellular and extracellular mechanical environment and mediate gene expression and cell remodeling. In this brief review, we highlight and discuss recent experimental work focused on the pathway of biomechanical stimulus propagation at the nucleus-cytoskeleton interface of striated muscles, and the mechanisms by which these pathways regulate gene regulation, muscle structure, and function. Furthermore, we discuss nuclear protein mutations that affect mechanosignaling function in human and animal models of cardiomyopathy. Furthermore, current open questions and future challenges in investigating striated muscle nuclear mechanosignaling are further discussed.

4.
Circ Res ; 131(12): 1001-1003, 2022 12 02.
Article in English | MEDLINE | ID: mdl-36454855

Subject(s)
RNA Splicing , Heart
5.
Bioinform Adv ; 2(1): vbac051, 2022.
Article in English | MEDLINE | ID: mdl-35967929

ABSTRACT

Motivation: Unsupervised clustering of single-cell transcriptomics is a powerful method for identifying cell populations. Static visualization techniques for single-cell clustering only display results for a single resolution parameter. Analysts will often evaluate more than one resolution parameter but then only report one. Results: We developed Cell Layers, an interactive Sankey tool for the quantitative investigation of gene expression, co-expression, biological processes and cluster integrity across clustering resolutions. Cell Layers enhances the interpretability of single-cell clustering by linking molecular data and cluster evaluation metrics, providing novel insight into cell populations. Availability and implementation: https://github.com/apblair/CellLayers.

6.
G3 (Bethesda) ; 12(3)2022 03 04.
Article in English | MEDLINE | ID: mdl-35100349

ABSTRACT

How and to what degree gene duplication events create regulatory innovation, redundancy, or neofunctionalization remain important questions in animal evolution and comparative genetics. Ankfn1 genes are single copy in most invertebrates, partially duplicated in jawed vertebrates, and only the derived copy retained in most mammals. Null mutations in the single mouse homolog have vestibular and neurological abnormalities. Null mutation of the single Drosophila homolog is typically lethal with severe sensorimotor deficits in rare survivors. The functions and potential redundancy of paralogs in species with two copies are not known. Here, we define a vestibular role for Ankfn1 homologs in zebrafish based on the simultaneous disruption of each locus. Zebrafish with both paralogs disrupted showed vestibular defects and early lethality from swim bladder inflation failure. One intact copy at either locus was sufficient to prevent major phenotypes. Our results show that vertebrate Ankfn1 genes are required for vestibular-related functions, with at least partial redundancy between ancestral and derived paralogs.


Subject(s)
Evolution, Molecular , Zebrafish , Animals , Gene Duplication , Mice , Organogenesis , Penetrance , Phylogeny , Zebrafish/genetics
7.
Nat Cardiovasc Res ; 1(9): 830-843, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36817700

ABSTRACT

The heart, a vital organ which is first to develop, has adapted its size, structure and function in order to accommodate the circulatory demands for a broad range of animals. Although heart development is controlled by a relatively conserved network of transcriptional/chromatin regulators, how the human heart has evolved species-specific features to maintain adequate cardiac output and function remains to be defined. Here, we show through comparative epigenomic analysis the identification of enhancers and promoters that have gained activity in humans during cardiogenesis. These cis-regulatory elements (CREs) are associated with genes involved in heart development and function, and may account for species-specific differences between human and mouse hearts. Supporting these findings, genetic variants that are associated with human cardiac phenotypic/disease traits, particularly those differing between human and mouse, are enriched in human-gained CREs. During early stages of human cardiogenesis, these CREs are also gained within genomic loci of transcriptional regulators, potentially expanding their role in human heart development. In particular, we discovered that gained enhancers in the locus of the early human developmental regulator ZIC3 are selectively accessible within a subpopulation of mesoderm cells which exhibits cardiogenic potential, thus possibly extending the function of ZIC3 beyond its conserved left-right asymmetry role. Genetic deletion of these enhancers identified a human gained enhancer that was required for not only ZIC3 and early cardiac gene expression at the mesoderm stage but also cardiomyocyte differentiation. Overall, our results illuminate how human gained CREs may contribute to human-specific cardiac attributes, and provide insight into how transcriptional regulators may gain cardiac developmental roles through the evolutionary acquisition of enhancers.

8.
Cell Syst ; 12(11): 1094-1107.e6, 2021 11 17.
Article in English | MEDLINE | ID: mdl-34411509

ABSTRACT

Patients with neurodevelopmental disorders, including autism, have an elevated incidence of congenital heart disease, but the extent to which these conditions share molecular mechanisms remains unknown. Here, we use network genetics to identify a convergent molecular network underlying autism and congenital heart disease. This network is impacted by damaging genetic variants from both disorders in multiple independent cohorts of patients, pinpointing 101 genes with shared genetic risk. Network analysis also implicates risk genes for each disorder separately, including 27 previously unidentified genes for autism and 46 for congenital heart disease. For 7 genes with shared risk, we create engineered disruptions in Xenopus tropicalis, confirming both heart and brain developmental abnormalities. The network includes a family of ion channels, such as the sodium transporter SCN2A, linking these functions to early heart and brain development. This study provides a road map for identifying risk genes and pathways involved in co-morbid conditions.


Subject(s)
Autism Spectrum Disorder , Autistic Disorder , Heart Defects, Congenital , Autism Spectrum Disorder/genetics , Autistic Disorder/genetics , Heart Defects, Congenital/genetics , Humans
10.
Sci Adv ; 7(20)2021 05.
Article in English | MEDLINE | ID: mdl-33990324

ABSTRACT

Misregulated gene expression in human hearts can result in cardiovascular diseases that are leading causes of mortality worldwide. However, the limited information on the genomic location of candidate cis-regulatory elements (cCREs) such as enhancers and promoters in distinct cardiac cell types has restricted the understanding of these diseases. Here, we defined >287,000 cCREs in the four chambers of the human heart at single-cell resolution, which revealed cCREs and candidate transcription factors associated with cardiac cell types in a region-dependent manner and during heart failure. We further found cardiovascular disease-associated genetic variants enriched within these cCREs including 38 candidate causal atrial fibrillation variants localized to cardiomyocyte cCREs. Additional functional studies revealed that two of these variants affect a cCRE controlling KCNH2/HERG expression and action potential repolarization. Overall, this atlas of human cardiac cCREs provides the foundation for illuminating cell type-specific gene regulation in human hearts during health and disease.


Subject(s)
Heart , Regulatory Sequences, Nucleic Acid , Humans , Promoter Regions, Genetic , Regulatory Sequences, Nucleic Acid/genetics , Transcription Factors/metabolism
11.
Dev Cell ; 56(2): 159-160, 2021 01 25.
Article in English | MEDLINE | ID: mdl-33497621

ABSTRACT

Organ maturation entails the reshaping of simple tissues into more complex structures critical for function. In a recent issue of Nature, Priya et al. show how tension heterogeneity between developing cardiomyocytes can coordinate the cell behaviors that remodel the architecture of the cardiac chamber wall.


Subject(s)
Myocardium , Physical Distancing , Morphogenesis , Myocytes, Cardiac , Organogenesis
12.
Development ; 147(12)2020 06 17.
Article in English | MEDLINE | ID: mdl-32439760

ABSTRACT

Physical forces are important participants in the cellular dynamics that shape developing organs. During heart formation, for example, contractility and blood flow generate biomechanical cues that influence patterns of cell behavior. Here, we address the interplay between function and form during the assembly of the cardiac outflow tract (OFT), a crucial connection between the heart and vasculature that develops while circulation is under way. In zebrafish, we find that the OFT expands via accrual of both endocardial and myocardial cells. However, when cardiac function is disrupted, OFT endocardial growth ceases, accompanied by reduced proliferation and reduced addition of cells from adjacent vessels. The flow-responsive TGFß receptor Acvrl1 is required for addition of endocardial cells, but not for their proliferation, indicating distinct modes of function-dependent regulation for each of these essential cell behaviors. Together, our results indicate that cardiac function modulates OFT morphogenesis by triggering endocardial cell accumulation that induces OFT lumen expansion and shapes OFT dimensions. Moreover, these morphogenetic mechanisms provide new perspectives regarding the potential causes of cardiac birth defects.


Subject(s)
Endocardium/metabolism , Heart/physiology , Zebrafish/metabolism , Activin Receptors/antagonists & inhibitors , Activin Receptors/genetics , Activin Receptors/metabolism , Animals , Animals, Genetically Modified/growth & development , Animals, Genetically Modified/metabolism , Cell Proliferation , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/metabolism , Endocardium/cytology , Heart/anatomy & histology , Heart/growth & development , Morpholinos/metabolism , Troponin T/antagonists & inhibitors , Troponin T/genetics , Troponin T/metabolism , Zebrafish/growth & development , Zebrafish Proteins/antagonists & inhibitors , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
13.
Nat Commun ; 11(1): 1122, 2020 02 28.
Article in English | MEDLINE | ID: mdl-32111823

ABSTRACT

Heart failure is a major public health problem affecting over 23 million people worldwide. In this study, we present the results of a large scale meta-analysis of heart failure GWAS and replication in a comparable sized cohort to identify one known and two novel loci associated with heart failure. Heart failure sub-phenotyping shows that a new locus in chromosome 1 is associated with left ventricular adverse remodeling and clinical heart failure, in response to different initial cardiac muscle insults. Functional characterization and fine-mapping of that locus reveal a putative causal variant in a cardiac muscle specific regulatory region activated during cardiomyocyte differentiation that binds to the ACTN2 gene, a crucial structural protein inside the cardiac sarcolemma (Hi-C interaction p-value = 0.00002). Genome-editing in human embryonic stem cell-derived cardiomyocytes confirms the influence of the identified regulatory region in the expression of ACTN2. Our findings extend our understanding of biological mechanisms underlying heart failure.


Subject(s)
Actinin/genetics , Genetic Predisposition to Disease/genetics , Heart Failure/genetics , ABO Blood-Group System/genetics , Atrial Fibrillation/genetics , Chromosomes, Human, Pair 1 , Enhancer Elements, Genetic , Gene Expression Regulation , Genome-Wide Association Study , Heart Failure/pathology , Human Embryonic Stem Cells/cytology , Humans , Musculoskeletal Diseases/genetics , Myocytes, Cardiac/cytology , Polymorphism, Single Nucleotide , Quantitative Trait Loci
14.
Dev Cell ; 50(6): 729-743.e5, 2019 09 23.
Article in English | MEDLINE | ID: mdl-31402282

ABSTRACT

Pacemaker cardiomyocytes that create the sinoatrial node are essential for the initiation and maintenance of proper heart rhythm. However, illuminating developmental cues that direct their differentiation has remained particularly challenging due to the unclear cellular origins of these specialized cardiomyocytes. By discovering the origins of pacemaker cardiomyocytes, we reveal an evolutionarily conserved Wnt signaling mechanism that coordinates gene regulatory changes directing mesoderm cell fate decisions, which lead to the differentiation of pacemaker cardiomyocytes. We show that in zebrafish, pacemaker cardiomyocytes derive from a subset of Nkx2.5+ mesoderm that responds to canonical Wnt5b signaling to initiate the cardiac pacemaker program, including activation of pacemaker cell differentiation transcription factors Isl1 and Tbx18 and silencing of Nkx2.5. Moreover, applying these developmental findings to human pluripotent stem cells (hPSCs) notably results in the creation of hPSC-pacemaker cardiomyocytes, which successfully pace three-dimensional bioprinted hPSC-cardiomyocytes, thus providing potential strategies for biological cardiac pacemaker therapy.


Subject(s)
Homeobox Protein Nkx-2.5/metabolism , Mesoderm/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction , Wnt Proteins/metabolism , Animals , Base Sequence , Bioprinting , Cell Differentiation , Gene Expression Regulation, Developmental , Humans , Loss of Function Mutation/genetics , Models, Cardiovascular , Myocytes, Cardiac/cytology , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Stem Cells/metabolism , Zebrafish
15.
Nat Genet ; 51(9): 1380-1388, 2019 09.
Article in English | MEDLINE | ID: mdl-31427791

ABSTRACT

Chromatin architecture has been implicated in cell type-specific gene regulatory programs, yet how chromatin remodels during development remains to be fully elucidated. Here, by interrogating chromatin reorganization during human pluripotent stem cell (hPSC) differentiation, we discover a role for the primate-specific endogenous retrotransposon human endogenous retrovirus subfamily H (HERV-H) in creating topologically associating domains (TADs) in hPSCs. Deleting these HERV-H elements eliminates their corresponding TAD boundaries and reduces the transcription of upstream genes, while de novo insertion of HERV-H elements can introduce new TAD boundaries. The ability of HERV-H to create TAD boundaries depends on high transcription, as transcriptional repression of HERV-H elements prevents the formation of boundaries. This ability is not limited to hPSCs, as these actively transcribed HERV-H elements and their corresponding TAD boundaries also appear in pluripotent stem cells from other hominids but not in more distantly related species lacking HERV-H elements. Overall, our results provide direct evidence for retrotransposons in actively shaping cell type- and species-specific chromatin architecture.


Subject(s)
Chromatin/genetics , Endogenous Retroviruses/genetics , Gene Expression Regulation , Pluripotent Stem Cells/cytology , Response Elements , Retroelements/genetics , Transcription, Genetic , Animals , Cell Differentiation , Humans , Pluripotent Stem Cells/physiology , Primates , Transcription Factors/genetics , Transcription Factors/metabolism
16.
Elife ; 82019 06 25.
Article in English | MEDLINE | ID: mdl-31237233

ABSTRACT

Lower vertebrate and neonatal mammalian hearts exhibit the remarkable capacity to regenerate through the reprogramming of pre-existing cardiomyocytes. However, how cardiac injury initiates signaling pathways controlling this regenerative reprogramming remains to be defined. Here, we utilize in vivo biophysical and genetic fate mapping zebrafish studies to reveal that altered hemodynamic forces due to cardiac injury activate a sequential endocardial-myocardial signaling cascade to direct cardiomyocyte reprogramming and heart regeneration. Specifically, these altered forces are sensed by the endocardium through the mechanosensitive channel Trpv4 to control Klf2a transcription factor expression. Consequently, Klf2a then activates endocardial Notch signaling which results in the non-cell autonomous initiation of myocardial Erbb2 and BMP signaling to promote cardiomyocyte reprogramming and heart regeneration. Overall, these findings not only reveal how the heart senses and adaptively responds to environmental changes due to cardiac injury, but also provide insight into how flow-mediated mechanisms may regulate cardiomyocyte reprogramming and heart regeneration.


Subject(s)
Endocardium/physiology , Heart Injuries/pathology , Hemodynamics , Mechanotransduction, Cellular , Myocytes, Cardiac/physiology , Regeneration , Animals , Kruppel-Like Transcription Factors/metabolism , Receptors, Notch/metabolism , TRPV Cation Channels/metabolism , Zebrafish , Zebrafish Proteins/metabolism
17.
Nat Biomed Eng ; 3(2): 147-157, 2019 02.
Article in English | MEDLINE | ID: mdl-30923642

ABSTRACT

Dilated cardiomyopathy (DCM) is a leading cause of morbidity and mortality worldwide; yet how genetic variation and environmental factors impact DCM heritability remains unclear. Here, we report that compound genetic interactions between DNA sequence variants contribute to the complex heritability of DCM. By using genetic data from a large family with a history of DCM, we discovered that heterozygous sequence variants in the TROPOMYOSIN 1 (TPM1) and VINCULIN (VCL) genes cose-gregate in individuals affected by DCM. In vitro studies of patient-derived and isogenic human-pluripotent-stem-cell-derived cardio-myocytes that were genome-edited via CRISPR to create an allelic series of TPM1 and VCL variants revealed that cardiomyocytes with both TPM1 and VCL variants display reduced contractility and sarcomeres that are less organized. Analyses of mice genetically engineered to harbour these human TPM1 and VCL variants show that stress on the heart may also influence the variable penetrance and expressivity of DCM-associated genetic variants in vivo. We conclude that compound genetic variants can interact combinatorially to induce DCM, particularly when influenced by other disease-provoking stressors.


Subject(s)
Cardiomyopathy, Dilated/genetics , Genetic Predisposition to Disease , Genetic Variation , Animals , Cardiomyopathy, Dilated/physiopathology , Extracellular Matrix/metabolism , Female , Gene Expression Regulation , Humans , Inheritance Patterns/genetics , Male , Mice , Models, Biological , Muscle Contraction/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Pedigree , Pluripotent Stem Cells/metabolism , Up-Regulation/genetics
18.
Stem Cell Reports ; 11(3): 828-841, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30122443

ABSTRACT

To facilitate understanding of human cardiomyocyte (CM) subtype specification, and the study of ventricular CM biology in particular, we developed a broadly applicable strategy for enrichment of ventricular cardiomyocytes (VCMs) derived from human embryonic stem cells (hESCs). A bacterial artificial chromosome transgenic H9 hESC line in which GFP expression was driven by the human ventricular-specific myosin light chain 2 (MYL2) promoter was generated, and screened to identify cell-surface markers specific for MYL2-GFP-expressing VCMs. A CD77+/CD200- cell-surface signature facilitated isolation of >97% cardiac troponin I-positive cells from H9 hESC differentiation cultures, with 65% expressing MYL2-GFP. This study provides a tool for VCM enrichment when using some, but not all, human pluripotent stem cell lines. Tools generated in this study can be utilized toward understanding CM subtype specification, and enriching for VCMs for therapeutic applications.


Subject(s)
Heart Ventricles/cytology , Human Embryonic Stem Cells/cytology , Myocytes, Cardiac/cytology , Antigens, CD/analysis , Cardiac Myosins/analysis , Cell Differentiation , Cell Line , Cells, Cultured , Humans , Myosin Light Chains/analysis , Trihexosylceramides/analysis
19.
Biol Open ; 6(11): 1756-1770, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28982700

ABSTRACT

The cardiovascular system facilitates body-wide distribution of oxygen, a vital process for the development and survival of virtually all vertebrates. However, the zebrafish, a vertebrate model organism, appears to form organs and survive mid-larval periods without a functional cardiovascular system. Despite such dispensability, it is the first organ to develop. Such enigma prompted us to hypothesize other cardiovascular functions that are important for developmental and/or physiological processes. Hence, systematic cellular ablations and functional perturbations were performed on the zebrafish cardiovascular system to gain comprehensive and body-wide understanding of such functions and to elucidate the underlying mechanisms. This approach identifies a set of organ-specific genes, each implicated for important functions. The study also unveils distinct cardiovascular mechanisms, each differentially regulating their expressions in organ-specific and oxygen-independent manners. Such mechanisms are mediated by organ-vessel interactions, circulation-dependent signals, and circulation-independent beating-heart-derived signals. A comprehensive and body-wide functional landscape of the cardiovascular system reported herein may provide clues as to why it is the first organ to develop. Furthermore, these data could serve as a resource for the study of organ development and function.

20.
Blood ; 130(5): 666-676, 2017 08 03.
Article in English | MEDLINE | ID: mdl-28576875

ABSTRACT

Deficiency of factor X (F10) in humans is a rare bleeding disorder with a heterogeneous phenotype and limited therapeutic options. Targeted disruption of F10 and other common pathway factors in mice results in embryonic/neonatal lethality with rapid resorption of homozygous mutants, hampering additional studies. Several of these mutants also display yolk sac vascular defects, suggesting a role for thrombin signaling in vessel development. The zebrafish is a vertebrate model that demonstrates conservation of the mammalian hemostatic and vascular systems. We have leveraged these advantages for in-depth study of the role of the coagulation cascade in the developmental regulation of hemostasis and vasculogenesis. In this article, we show that ablation of zebrafish f10 by using genome editing with transcription activator-like effector nucleases results in a major embryonic hemostatic defect. However, widespread hemorrhage and subsequent lethality does not occur until later stages, with absence of any detectable defect in vascular development. We also use f10-/- zebrafish to confirm 5 novel human F10 variants as causative mutations in affected patients, providing a rapid and reliable in vivo model for testing the severity of F10 variants. These findings as well as the prolonged survival of f10-/- mutants will enable us to expand our understanding of the molecular mechanisms of hemostasis, including a platform for screening variants of uncertain significance in patients with F10 deficiency and other coagulation disorders. Further study as to how fish tolerate what is an early lethal mutation in mammals could facilitate improvement of diagnostics and therapeutics for affected patients with bleeding disorders.


Subject(s)
Blood Coagulation/genetics , Factor X , Gene Editing , Mutation , Zebrafish Proteins , Zebrafish , Animals , Factor X/genetics , Factor X/metabolism , Factor X Deficiency/embryology , Factor X Deficiency/genetics , Humans , Mice , Zebrafish/embryology , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...