Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Nature ; 608(7921): 181-191, 2022 08.
Article in English | MEDLINE | ID: mdl-35732239

ABSTRACT

The heart, the first organ to develop in the embryo, undergoes complex morphogenesis that when defective results in congenital heart disease (CHD). With current therapies, more than 90% of patients with CHD survive into adulthood, but many suffer premature death from heart failure and non-cardiac causes1. Here, to gain insight into this disease progression, we performed single-nucleus RNA sequencing on 157,273 nuclei from control hearts and hearts from patients with CHD, including those with hypoplastic left heart syndrome (HLHS) and tetralogy of Fallot, two common forms of cyanotic CHD lesions, as well as dilated and hypertrophic cardiomyopathies. We observed CHD-specific cell states in cardiomyocytes, which showed evidence of insulin resistance and increased expression of genes associated with FOXO signalling and CRIM1. Cardiac fibroblasts in HLHS were enriched in a low-Hippo and high-YAP cell state characteristic of activated cardiac fibroblasts. Imaging mass cytometry uncovered a spatially resolved perivascular microenvironment consistent with an immunodeficient state in CHD. Peripheral immune cell profiling suggested deficient monocytic immunity in CHD, in agreement with the predilection in CHD to infection and cancer2. Our comprehensive phenotyping of CHD provides a roadmap towards future personalized treatments for CHD.


Subject(s)
Heart Defects, Congenital , Phenotype , Bone Morphogenetic Protein Receptors/metabolism , Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/metabolism , Cardiomyopathy, Dilated/pathology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/immunology , Cardiomyopathy, Hypertrophic/metabolism , Cardiomyopathy, Hypertrophic/pathology , Disease Progression , Fibroblasts/metabolism , Fibroblasts/pathology , Forkhead Transcription Factors/metabolism , Heart Defects, Congenital/genetics , Heart Defects, Congenital/immunology , Heart Defects, Congenital/metabolism , Heart Defects, Congenital/pathology , Humans , Hypoplastic Left Heart Syndrome/genetics , Hypoplastic Left Heart Syndrome/immunology , Hypoplastic Left Heart Syndrome/metabolism , Hypoplastic Left Heart Syndrome/pathology , Image Cytometry , Insulin Resistance , Monocytes/immunology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , RNA-Seq , Signal Transduction/genetics , Single-Cell Analysis , Tetralogy of Fallot/genetics , Tetralogy of Fallot/immunology , Tetralogy of Fallot/metabolism , Tetralogy of Fallot/pathology , YAP-Signaling Proteins/metabolism
2.
JCI Insight ; 7(11)2022 06 08.
Article in English | MEDLINE | ID: mdl-35471998

ABSTRACT

Atrial fibrillation (AF), the most common sustained cardiac arrhythmia and a major risk factor for stroke, often arises through ectopic electrical impulses derived from the pulmonary veins (PVs). Sequence variants in enhancers controlling expression of the transcription factor PITX2, which is expressed in the cardiomyocytes (CMs) of the PV and left atrium (LA), have been implicated in AF predisposition. Single nuclei multiomic profiling of RNA and analysis of chromatin accessibility combined with spectral clustering uncovered distinct PV- and LA-enriched CM cell states. Pitx2-mutant PV and LA CMs exhibited gene expression changes consistent with cardiac dysfunction through cell type-distinct, PITX2-directed, cis-regulatory grammars controlling target gene expression. The perturbed network targets in each CM were enriched in distinct human AF predisposition genes, suggesting combinatorial risk for AF genesis. Our data further reveal that PV and LA Pitx2-mutant CMs signal to endothelial and endocardial cells through BMP10 signaling with pathogenic potential. This work provides a multiomic framework for interrogating the basis of AF predisposition in the PVs of humans.


Subject(s)
Atrial Fibrillation , Homeodomain Proteins , Transcription Factors , Atrial Fibrillation/genetics , Atrial Fibrillation/metabolism , Bone Morphogenetic Proteins/genetics , Bone Morphogenetic Proteins/metabolism , Gene Regulatory Networks , Heart Atria/metabolism , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Homeobox Protein PITX2
3.
Dev Biol ; 478: 163-172, 2021 10.
Article in English | MEDLINE | ID: mdl-34245725

ABSTRACT

The cardiac conduction system is a network of heterogeneous cell population that initiates and propagates electric excitations in the myocardium. Purkinje fibers, a network of specialized myocardial cells, comprise the distal end of the conduction system in the ventricles. The developmental origins of Purkinje fibers and their roles during cardiac physiology and arrhythmia have been reported. However, it is not clear if they play a role during ischemic injury and heart regeneration. Here we introduce a novel tamoxifen-inducible Cre allele that specifically labels a broad range of components in the cardiac conduction system while excludes other cardiac cell types and vital organs. Using this new allele, we investigated the cellular and molecular response of Purkinje fibers to myocardial injury. In a neonatal mouse myocardial infarction model, we observed significant increase in Purkinje cell number in regenerating myocardium. RNA-Seq analysis using laser-captured Purkinje fibers showed a unique transcriptomic response to myocardial infarction. Our finds suggest a novel role of cardiac Purkinje fibers in heart injury.


Subject(s)
Heart Conduction System/physiology , Integrases/genetics , Myocardial Infarction/physiopathology , Purkinje Fibers/physiology , Alleles , Animals , Animals, Newborn , Cell Lineage , Heart Conduction System/physiopathology , Heart Ventricles/pathology , Mice , Mice, Transgenic , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/physiology , Purkinje Fibers/physiopathology , RNA-Seq , Regeneration , Tamoxifen/pharmacology , Transcriptome , Ventricular Function
4.
Circ Res ; 127(1): 34-50, 2020 06 19.
Article in English | MEDLINE | ID: mdl-32717170

ABSTRACT

Genome-wide association studies have uncovered over a 100 genetic loci associated with atrial fibrillation (AF), the most common arrhythmia. Many of the top AF-associated loci harbor key cardiac transcription factors, including PITX2, TBX5, PRRX1, and ZFHX3. Moreover, the vast majority of the AF-associated variants lie within noncoding regions of the genome where causal variants affect gene expression by altering the activity of transcription factors and the epigenetic state of chromatin. In this review, we discuss a transcriptional regulatory network model for AF defined by effector genes in Genome-wide association studies loci. We describe the current state of the field regarding the identification and function of AF-relevant gene regulatory networks, including variant regulatory elements, dose-sensitive transcription factor functionality, target genes, and epigenetic states. We illustrate how altered transcriptional networks may impact cardiomyocyte function and ionic currents that impact AF risk. Last, we identify the need for improved tools to identify and functionally test transcriptional components to define the links between genetic variation, epigenetic gene regulation, and atrial function.


Subject(s)
Atrial Fibrillation/genetics , Epigenesis, Genetic , Gene Regulatory Networks , Animals , Atrial Fibrillation/metabolism , Genetic Loci , Humans , Transcriptome
5.
Article in English | MEDLINE | ID: mdl-31615785

ABSTRACT

Within the realm of zoological study, the question of how an organism reaches a specific size has been largely unexplored. Recently, studies performed to understand the regulation of organ size have revealed that both cellular signals and external cues contribute toward the determination of total cell mass within each organ. The establishment of final organ size requires the precise coordination of cell growth, proliferation, and survival throughout development and postnatal life. In the mammalian heart, the regulation of size is biphasic. During development, cardiomyocyte proliferation predominantly determines cardiac growth, whereas in the adult heart, total cell mass is governed by signals that regulate cardiac hypertrophy. Here, we review the current state of knowledge regarding the extrinsic factors and intrinsic mechanisms that control heart size during development. We also discuss the metabolic switch that occurs in the heart after birth and precedes homeostatic control of postnatal heart size.


Subject(s)
Cardiomegaly/metabolism , Heart/growth & development , Heart/physiology , Hypertrophy/pathology , Zoology/methods , Animals , Cell Cycle , Cell Proliferation , Cell Survival , Humans , Myocardium/metabolism , Myocytes, Cardiac/cytology , Organ Size , Organogenesis , Signal Transduction , Somatomedins/metabolism
6.
Proc Natl Acad Sci U S A ; 116(45): 22692-22698, 2019 11 05.
Article in English | MEDLINE | ID: mdl-31636200

ABSTRACT

Genome-wide association studies found that increased risk for atrial fibrillation (AF), the most common human heart arrhythmia, is associated with noncoding sequence variants located in proximity to PITX2 Cardiomyocyte-specific epigenomic and comparative genomics uncovered 2 AF-associated enhancers neighboring PITX2 with varying conservation in mice. Chromosome conformation capture experiments in mice revealed that the Pitx2c promoter directly contacted the AF-associated enhancer regions. CRISPR/Cas9-mediated deletion of a 20-kb topologically engaged enhancer led to reduced Pitx2c transcription and AF predisposition. Allele-specific chromatin immunoprecipitation sequencing on hybrid heterozygous enhancer knockout mice revealed that long-range interaction of an AF-associated region with the Pitx2c promoter was required for maintenance of the Pitx2c promoter chromatin state. Long-range looping was mediated by CCCTC-binding factor (CTCF), since genetic disruption of the intronic CTCF-binding site caused reduced Pitx2c expression, AF predisposition, and diminished active chromatin marks on Pitx2 AF risk variants located at 4q25 reside in genomic regions possessing long-range transcriptional regulatory functions directed at PITX2.


Subject(s)
Atrial Fibrillation/genetics , Enhancer Elements, Genetic , Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Promoter Regions, Genetic , Transcription Factors/genetics , Animals , CRISPR-Cas Systems , Chromosome Mapping , Epigenesis, Genetic , Genome-Wide Association Study , Mice , Mice, Knockout , Homeobox Protein PITX2
7.
Development ; 146(12)2019 06 14.
Article in English | MEDLINE | ID: mdl-31201182

ABSTRACT

The Pitx2 gene encodes a homeobox transcription factor that is required for mammalian development. Disruption of PITX2 expression in humans causes congenital heart diseases and is associated with atrial fibrillation; however, the cellular and molecular processes dictated by Pitx2 during cardiac ontogeny remain unclear. To characterize the role of Pitx2 during murine heart development we sequenced over 75,000 single cardiac cell transcriptomes between two key developmental timepoints in control and Pitx2 null embryos. We found that cardiac cell composition was dramatically altered in mutants at both E10.5 and E13.5. Interestingly, the differentiation dynamics of both anterior and posterior second heart field-derived progenitor cells were disrupted in Pitx2 mutants. We also uncovered evidence for defects in left-right asymmetry within atrial cardiomyocyte populations. Furthermore, we were able to detail defects in cardiac outflow tract and valve development associated with Pitx2 Our findings offer insight into Pitx2 function and provide a compilation of gene expression signatures for further detailing the complexities of heart development that will serve as the foundation for future studies of cardiac morphogenesis, congenital heart disease and arrhythmogenesis.


Subject(s)
Gene Expression Regulation, Developmental , Heart Valves/embryology , Heart/embryology , Homeodomain Proteins/physiology , Myocytes, Cardiac/metabolism , Transcription Factors/physiology , Alleles , Animals , Heart Atria , Heart Defects, Congenital/genetics , Homeodomain Proteins/genetics , Mice , Mutation , Myocardium/metabolism , Nuclear Proteins/metabolism , Organogenesis , Sequence Analysis, RNA , Transcription Factors/genetics , Transcriptome , Homeobox Protein PITX2
8.
Circ Res ; 124(11): 1647-1657, 2019 05 24.
Article in English | MEDLINE | ID: mdl-31120819

ABSTRACT

After myocardial injury, cardiomyocyte loss cannot be corrected by using currently available clinical treatments. In recent years, considerable effort has been made to develop cell-based cardiac repair therapies aimed at correcting for this loss. An exciting crop of recent studies reveals that inducing endogenous repair and proliferation of cardiomyocytes may be a viable option for regenerating injured myocardium. Here, we review current heart failure treatments, the state of cardiomyocyte renewal in mammals, and the molecular signals that stimulate cardiomyocyte proliferation. These signals include growth factors, intrinsic signaling pathways, microRNAs, and cell cycle regulators. Animal model cardiac regeneration studies reveal that modulation of exogenous and cell-intrinsic signaling pathways can induce reentry of adult cardiomyocytes into the cell cycle. Using direct myocardial injection, epicardial patch delivery, or systemic administration of growth molecules, these studies show that inducing endogenous cardiomyocytes to self-renew is an exciting and promising therapeutic strategy to treat cardiac injury in humans.


Subject(s)
Cardiovascular Agents/therapeutic use , Cell Proliferation/drug effects , Heart Failure/therapy , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/transplantation , Regeneration/drug effects , Stem Cell Transplantation , Animals , Cardiovascular Agents/adverse effects , Heart Failure/metabolism , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Recovery of Function , Signal Transduction , Stem Cell Transplantation/adverse effects , Treatment Outcome
9.
Circ Res ; 123(9): 1012-1014, 2018 10 12.
Article in English | MEDLINE | ID: mdl-30355168
10.
Dev Cell ; 44(4): 403-404, 2018 02 26.
Article in English | MEDLINE | ID: mdl-29486188

ABSTRACT

There is mounting circumstantial evidence that ploidy, a cell's relative DNA content, is in part responsible for the differential cardiac regenerative capacity observed between regenerative and non-regenerative organisms. In this issue of Developmental Cell, González-Rosa et al. (2018) provide direct evidence that polyploid cardiomyocytes have reduced proliferative and regenerative potential.


Subject(s)
Heart , Regeneration , Myocytes, Cardiac , Ploidies
SELECTION OF CITATIONS
SEARCH DETAIL
...