Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Front Cell Dev Biol ; 12: 1388378, 2024.
Article in English | MEDLINE | ID: mdl-38699159

ABSTRACT

In heart disease patients, myocyte loss or malfunction invariably leads to fibrosis, involving the activation and accumulation of cardiac fibroblasts that deposit large amounts of extracellular matrix. Apart from the vital replacement fibrosis that follows myocardial infarction, ensuring structural integrity of the heart, cardiac fibrosis is largely considered to be maladaptive. Much work has focused on signaling pathways driving the fibrotic response, including TGF-ß signaling and biomechanical strain. However, currently there are very limited options for reducing cardiac fibrosis, with most patients suffering from chronic fibrosis. The adult heart has very limited regenerative capacity. However, cardiac regeneration has been reported in humans perinatally, and reproduced experimentally in neonatal mice. Furthermore, model organisms such as the zebrafish are able to fully regenerate their hearts following massive cardiac damage into adulthood. Increasing evidence points to a transient immuno-fibrotic response as being key for cardiac regeneration to occur. The mechanisms at play in this context are changing our views on fibrosis, and could be leveraged to promote beneficial remodeling in heart failure patients. This review summarizes our current knowledge of fibroblast properties associated with the healthy, failing or regenerating heart. Furthermore, we explore how cardiac fibroblast activity could be targeted to assist future therapeutic approaches.

2.
Int J Mol Sci ; 24(21)2023 Oct 25.
Article in English | MEDLINE | ID: mdl-37958548

ABSTRACT

Cornelia de Lange Syndrome (CdLS) patients, who frequently carry a mutation in NIPBL, present an increased incidence of outflow tract (OFT)-related congenital heart defects (CHDs). Nipbl+/- mice recapitulate a number of phenotypic traits of CdLS patients, including a small body size and cardiac defects, but no study has specifically focused on the valves. Here, we show that adult Nipbl+/- mice present aortic valve thickening, a condition that has been associated with stenosis. During development, we observed that OFT septation and neural crest cell condensation was delayed in Nipbl+/- embryos. However, we did not observe defects in the deployment of the main lineages contributing to the semilunar valves. Indeed, endocardial endothelial-to-mesenchymal transition (EndMT), analysed via outflow tract explants, and neural crest migration, analysed via genetic lineage tracing, did not significantly differ in Nipbl+/- mice and their wild-type littermates. Our study provides the first direct evidence for valve formation defects in Nipbl+/- mice and points to specific developmental defects as an origin for valve disease in patients.


Subject(s)
Cell Cycle Proteins , De Lange Syndrome , Heart Defects, Congenital , Animals , Humans , Mice , Aortic Valve , Cell Cycle Proteins/genetics , De Lange Syndrome/genetics , Haploinsufficiency , Heart Defects, Congenital/genetics , Mutation
4.
J Mol Cell Biol ; 14(10)2023 03 29.
Article in English | MEDLINE | ID: mdl-36271843

ABSTRACT

Understanding how certain animals are capable of regenerating their hearts will provide much needed insights into how this process can be induced in humans in order to reverse the damage caused by myocardial infarction. Currently, it is becoming increasingly evident that cardiac interstitial cells play crucial roles during cardiac regeneration. To understand how interstitial cells behave during this process, we performed single-cell RNA sequencing of regenerating zebrafish hearts. Using a combination of immunohistochemistry, chemical inhibition, and novel transgenic animals, we were able to investigate the role of cell type-specific mechanisms during cardiac regeneration. This approach allowed us to identify a number of important regenerative processes within the interstitial cell populations. Here, we provide detailed insight into how interstitial cells behave during cardiac regeneration, which will serve to increase our understanding of how this process could eventually be induced in humans.


Subject(s)
Myocardial Infarction , Myocytes, Cardiac , Animals , Humans , Zebrafish , Animals, Genetically Modified , Cell Proliferation
5.
Circ Res ; 126(10): 1330-1342, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32175811

ABSTRACT

RATIONALE: Fibro-fatty infiltration of subepicardial layers of the atrial wall has been shown to contribute to the substrate of atrial fibrillation. OBJECTIVE: Here, we examined if the epicardium that contains multipotent cells is involved in this remodeling process. METHODS AND RESULTS: One hundred nine human surgical right atrial specimens were evaluated. There was a relatively greater extent of epicardial thickening and dense fibro-fatty infiltrates in atrial tissue sections from patients aged over 70 years who had mitral valve disease or atrial fibrillation when compared with patients aged less than 70 years with ischemic cardiomyopathy as indicated using logistic regression adjusted for age and gender. Cells coexpressing markers of epicardial progenitors and fibroblasts were detected in fibro-fatty infiltrates. Such epicardial remodeling was reproduced in an experimental model of atrial cardiomyopathy in rat and in Wilms tumor 1 (WT1)CreERT2/+;ROSA-tdT+/- mice. In the latter, genetic lineage tracing demonstrated the epicardial origin of fibroblasts within fibro-fatty infiltrates. A subpopulation of human adult epicardial-derived cells expressing PDGFR (platelet-derived growth factor receptor)-α were isolated and differentiated into myofibroblasts in the presence of Ang II (angiotensin II). Furthermore, single-cell RNA-sequencing analysis identified several clusters of adult epicardial-derived cells and revealed their specification from adipogenic to fibrogenic cells in the rat model of atrial cardiomyopathy. CONCLUSIONS: Epicardium is reactivated during the formation of the atrial cardiomyopathy. Subsets of adult epicardial-derived cells, preprogrammed towards a specific cell fate, contribute to fibro-fatty infiltration of subepicardium of diseased atria. Our study reveals the biological basis for chronic atrial myocardial remodeling that paves the way of atrial fibrillation.


Subject(s)
Adipose Tissue/pathology , Atrial Fibrillation/etiology , Atrial Remodeling , Cardiomyopathies/complications , Heart Atria/pathology , Myocardium/pathology , Pericardium/pathology , Action Potentials , Adipocytes/metabolism , Adipocytes/pathology , Adipose Tissue/metabolism , Aged , Animals , Atrial Fibrillation/metabolism , Atrial Fibrillation/pathology , Atrial Fibrillation/physiopathology , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cardiomyopathies/physiopathology , Cell Lineage , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Heart Atria/metabolism , Heart Atria/physiopathology , Heart Rate , Humans , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocardium/metabolism , Pericardium/metabolism , Pericardium/physiopathology , Rats, Wistar , Stem Cells/metabolism , Stem Cells/pathology , WT1 Proteins/genetics , WT1 Proteins/metabolism
6.
Nat Commun ; 10(1): 1929, 2019 04 26.
Article in English | MEDLINE | ID: mdl-31028265

ABSTRACT

Genetically modified mice have advanced our understanding of valve development and disease. Yet, human pathophysiological valvulogenesis remains poorly understood. Here we report that, by combining single cell sequencing and in vivo approaches, a population of human pre-valvular endocardial cells (HPVCs) can be derived from pluripotent stem cells. HPVCs express gene patterns conforming to the E9.0 mouse atrio-ventricular canal (AVC) endocardium signature. HPVCs treated with BMP2, cultured on mouse AVC cushions, or transplanted into the AVC of embryonic mouse hearts, undergo endothelial-to-mesenchymal transition and express markers of valve interstitial cells of different valvular layers, demonstrating cell specificity. Extending this model to patient-specific induced pluripotent stem cells recapitulates features of mitral valve prolapse and identified dysregulation of the SHH pathway. Concurrently increased ECM secretion can be rescued by SHH inhibition, thus providing a putative therapeutic target. In summary, we report a human cell model of valvulogenesis that faithfully recapitulates valve disease in a dish.


Subject(s)
Endothelial Cells/pathology , Hedgehog Proteins/genetics , Mitral Valve Prolapse/pathology , Mitral Valve/pathology , Pluripotent Stem Cells/pathology , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers/metabolism , Bone Morphogenetic Protein 2/pharmacology , Cadherin Related Proteins , Cadherins/genetics , Cadherins/metabolism , Cell Differentiation/drug effects , Embryo, Mammalian , Endocardium/metabolism , Endocardium/pathology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Endothelial Cells/transplantation , Epithelial-Mesenchymal Transition/drug effects , GATA5 Transcription Factor/genetics , GATA5 Transcription Factor/metabolism , Gene Expression Profiling , Gene Expression Regulation , Heart Atria/metabolism , Heart Atria/pathology , Hedgehog Proteins/metabolism , Humans , Mice , Mitral Valve/metabolism , Mitral Valve Prolapse/genetics , Mitral Valve Prolapse/metabolism , Mitral Valve Prolapse/therapy , Models, Biological , Pluripotent Stem Cells/drug effects , Pluripotent Stem Cells/metabolism , Primary Cell Culture , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism , Wnt3A Protein/pharmacology
7.
Physiol Rev ; 98(4): 2453-2475, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30156497

ABSTRACT

The heart is the first organ to be functional in the fetus. Heart formation is a complex morphogenetic process regulated by both genetic and epigenetic mechanisms. Congenital heart diseases (CHD) are the most prominent congenital diseases. Genetics is not sufficient to explain these diseases or the impact of them on patients. Epigenetics is more and more emerging as a basis for cardiac malformations. This review brings the essential knowledge on cardiac biology of development. It further provides a broad background on epigenetics with a focus on three-dimensional conformation of chromatin. Then, we summarize the current knowledge of the impact of epigenetics on cardiac cell fate decision. We further provide an update on the epigenetic anomalies in the genesis of CHD.


Subject(s)
Epigenesis, Genetic/genetics , Epigenesis, Genetic/physiology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/physiopathology , Heart/growth & development , Animals , Epigenomics/methods , Fetus/physiology , Humans
8.
Circ Res ; 122(4): 583-590, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29269349

ABSTRACT

RATIONALE: Myocardial infarction is a major cause of adult mortality worldwide. The origin(s) of cardiac fibroblasts that constitute the postinfarct scar remain controversial, in particular the potential contribution of bone marrow lineages to activated fibroblasts within the scar. OBJECTIVE: The aim of this study was to establish the origin(s) of infarct fibroblasts using lineage tracing and bone marrow transplants and a robust marker for cardiac fibroblasts, the Collagen1a1-green fluorescent protein reporter. METHODS AND RESULTS: Using genetic lineage tracing or bone marrow transplant, we found no evidence for collagen-producing fibroblasts derived from hematopoietic or bone marrow lineages in hearts subjected to permanent left anterior descending coronary artery ligation. In fact, fibroblasts within the infarcted area were largely of epicardial origin. Intriguingly, collagen-producing fibrocytes from hematopoietic lineages were observed attached to the epicardial surface of infarcted and sham-operated hearts in which a suture was placed around the left anterior descending coronary artery. CONCLUSIONS: In this controversial field, our study demonstrated that the vast majority of infarct fibroblasts were of epicardial origin and not derived from bone marrow lineages, endothelial-to-mesenchymal transition, or blood. We also noted the presence of collagen-producing fibrocytes on the epicardial surface that resulted at least in part from the surgical procedure.


Subject(s)
Bone Marrow Cells/cytology , Cell Lineage , Myocardial Infarction/therapy , Myofibroblasts/cytology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/adverse effects , Cells, Cultured , Collagen Type I/genetics , Collagen Type I/metabolism , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Pericardium/cytology
9.
Cell Stem Cell ; 20(3): 345-359.e5, 2017 03 02.
Article in English | MEDLINE | ID: mdl-28111199

ABSTRACT

Pericytes are widely believed to function as mesenchymal stem cells (MSCs), multipotent tissue-resident progenitors with great potential for regenerative medicine. Cultured pericytes isolated from distinct tissues can differentiate into multiple cell types in vitro or following transplantation in vivo. However, the cell fate plasticity of endogenous pericytes in vivo remains unclear. Here, we show that the transcription factor Tbx18 selectively marks pericytes and vascular smooth muscle cells in multiple organs of adult mouse. Fluorescence-activated cell sorting (FACS)-purified Tbx18-expressing cells behaved as MSCs in vitro. However, lineage-tracing experiments using an inducible Tbx18-CreERT2 line revealed that pericytes and vascular smooth muscle cells maintained their identity in aging and diverse pathological settings and did not significantly contribute to other cell lineages. These results challenge the current view of endogenous pericytes as multipotent tissue-resident progenitors and suggest that the plasticity observed in vitro or following transplantation in vivo arises from artificial cell manipulations ex vivo.


Subject(s)
Mesenchymal Stem Cells/cytology , Organ Specificity , Pericytes/cytology , Adipocytes/cytology , Aging/genetics , Cell Lineage , Cicatrix/pathology , Fibroblasts/cytology , Gene Expression Regulation, Developmental , Green Fluorescent Proteins/metabolism , Humans , Integrases/metabolism , Mesenchymal Stem Cells/metabolism , Muscle Development , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/metabolism , Neurons/cytology , Pericytes/metabolism , Phenotype , Receptor, Platelet-Derived Growth Factor beta/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
10.
Proc Natl Acad Sci U S A ; 114(5): E771-E780, 2017 01 31.
Article in English | MEDLINE | ID: mdl-28096344

ABSTRACT

The abundance of epicardial adipose tissue (EAT) is associated with atrial fibrillation (AF), the most frequent cardiac arrhythmia. However, both the origin and the factors involved in EAT expansion are unknown. Here, we found that adult human atrial epicardial cells were highly adipogenic through an epithelial-mesenchymal transition both in vitro and in vivo. In a genetic lineage tracing the WT1CreERT2+/-RosatdT+/- mouse model subjected to a high-fat diet, adipocytes of atrial EAT derived from a subset of epicardial progenitors. Atrial myocardium secretome induces the adipogenic differentiation of adult mesenchymal epicardium-derived cells by modulating the balance between mesenchymal Wingless-type Mouse Mammary Tumor Virus integration site family, member 10B (Wnt10b)/ß-catenin and adipogenic ERK/MAPK signaling pathways. The adipogenic property of the atrial secretome was enhanced in AF patients. The atrial natriuretic peptide secreted by atrial myocytes is a major adipogenic factor operating at a low concentration by binding to its natriuretic peptide receptor A (NPRA) receptor and, in turn, by activating a cGMP-dependent pathway. Hence, our data indicate cross-talk between EAT expansion and mechanical function of the atrial myocardium.


Subject(s)
Adipogenesis/physiology , Adipose Tissue/metabolism , Atrial Natriuretic Factor/metabolism , Heart Atria/metabolism , Pericardium/metabolism , Adipocytes/cytology , Aged , Animals , Cells, Cultured , Diet, High-Fat , Epithelial-Mesenchymal Transition , Female , Heart Atria/cytology , Humans , MAP Kinase Signaling System , Male , Mice, Inbred C57BL , Mice, Transgenic , Middle Aged , Myocytes, Cardiac/metabolism , Pericardium/cytology , Proto-Oncogene Proteins/metabolism , Stem Cells/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism
11.
J Mol Cell Cardiol ; 91: 1-5, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26748307

ABSTRACT

Cardiac fibroblasts produce the extracellular matrix (ECM) scaffold within which the various cellular components of the heart are organized. As well as providing structural support, it is becoming evident that the quality and quantity of ECM is a key factor for determining cardiac cell behavior during development and in pathological contexts such as heart failure involving fibrosis. Cardiac fibroblasts have long remained a poorly characterized cardiac lineage. Well characterized markers are now paving the way for a better understanding of the roles of these cells in various developmental and disease contexts. Notably, the relevance of processes including endothelial-tomesenchymal transition and the recruitment of circulating fibroblast progenitors in heart failure has been challenged. This review describes the latest findings on cardiac fibroblast markers and developmental origins, and discusses their importance in myocardial remodeling. Effective modulation of cardiac fibroblast activity would likely contribute to successful treatment of various cardiac disorders.


Subject(s)
Cell Lineage/physiology , Fibroblasts/pathology , Heart Failure/pathology , Myocardium/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Biomarkers/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Discoidin Domain Receptors , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibrosis , Gene Expression , Heart Failure/genetics , Heart Failure/metabolism , Humans , Myocardium/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptor, Platelet-Derived Growth Factor beta/genetics , Receptor, Platelet-Derived Growth Factor beta/metabolism , Receptors, Mitogen/genetics , Receptors, Mitogen/metabolism , T-Box Domain Proteins/genetics , T-Box Domain Proteins/metabolism
12.
Stem Cells ; 34(1): 34-43, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26352327

ABSTRACT

Heart failure is still a major cause of hospitalization and mortality in developed countries. Many clinical trials have tested the use of multipotent stem cells as a cardiac regenerative medicine. The benefit for the patients of this therapeutic intervention has remained limited. Herein, we review the pluripotent stem cells as a cell source for cardiac regeneration. We more specifically address the various challenges of this cell therapy approach. We question the cell delivery systems, the immune tolerance of allogenic cells, the potential proarrhythmic effects, various drug mediated interventions to facilitate cell grafting and, finally, we describe the pathological conditions that may benefit from such an innovative approach. As members of a transatlantic consortium of excellence of basic science researchers and clinicians, we propose some guidelines to be applied to cell types and modes of delivery in order to translate pluripotent stem cell cardiac derivatives into safe and effective clinical trials.


Subject(s)
Heart Failure/therapy , Myocardium/cytology , Pluripotent Stem Cells/cytology , Stem Cell Transplantation , Animals , Cell Differentiation , Disease Models, Animal , Heart Failure/physiopathology , Humans
13.
J Mol Med (Berl) ; 93(8): 823-30, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26169532

ABSTRACT

Cardiac fibroblasts are a major cell population of the heart and are characterized by their capacity to produce extracellular matrix (ECM). In hearts subjected to pressure overload, excessive fibroblast accumulation is responsible for fibrosis of the myocardium, a major clinical issue. Hence, understanding mechanisms generating fibroblasts in this context has become a key question in the cardiovascular field. Recent studies now point to the activation of resident fibroblasts as the underlying cause of fibrosis. However, de novo generation of fibroblasts from endothelium and circulating hematopoietic cells has also been proposed to significantly contribute to fibrosis. Here, we discuss the latest findings on fibroblast origins, with a particular emphasis on the pressure overload model, and the implication of these findings for the development of anti-fibrotic therapies that are currently lacking.


Subject(s)
Fibroblasts/cytology , Fibroblasts/pathology , Heart Failure/pathology , Myocardium/cytology , Myocardium/pathology , Animals , Extracellular Matrix/metabolism , Extracellular Matrix/pathology , Fibroblasts/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Heart/embryology , Heart Failure/metabolism , Humans , Myocardium/metabolism
14.
Genesis ; 53(5): 337-45, 2015 May.
Article in English | MEDLINE | ID: mdl-25950518

ABSTRACT

Heart valve development begins with the endothelial-to-mesenchymal transition (EMT) of endocardial cells. Although lineage studies have demonstrated contributions from cardiac neural crest and epicardium to semilunar and atrioventricular (AV) valve formation, respectively, most valve mesenchyme derives from the endocardial EMT. Specific Cre mouse lines for fate-mapping analyses of valve endocardial cells are limited. Msx1 displayed expression in AV canal endocardium and cushion mesenchyme between E9.5 and E11.5, when EMT is underway. Additionally, previous studies have demonstrated that deletion of Msx1 and its paralog Msx2 results in hypoplastic AV cushions and impaired endocardial signaling. A knock-in tamoxifen-inducible Cre line was recently generated (Msx1CreERT2) and characterized during embryonic development and after birth, and was shown to recapitulate the endogenous Msx1 expression pattern. Here, we further analyze this knock-in allele and track the Msx1-expressing cells and their descendants during cardiac development with a particular focus on their contribution to the valves and their precursors. Thus, Msx1CreERT2 mice represent a useful model for lineage tracing and conditional gene manipulation of endocardial and mesenchymal cushion cells essential to understand mechanisms of valve development and remodeling.


Subject(s)
Alleles , Gene Knock-In Techniques , Heart Valves/embryology , Heart Valves/metabolism , Integrases/genetics , MSX1 Transcription Factor/genetics , Receptors, Estrogen/genetics , Animals , Endocardium/enzymology , Endocardium/metabolism , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Developmental , Integrases/metabolism , MSX1 Transcription Factor/metabolism , Mice , Organogenesis/genetics , Receptors, Estrogen/metabolism
17.
Proc Natl Acad Sci U S A ; 111(32): E3297-305, 2014 Aug 12.
Article in English | MEDLINE | ID: mdl-25074909

ABSTRACT

Hepatic myofibroblasts are activated in response to chronic liver injury of any etiology to produce a fibrous scar. Despite extensive studies, the origin of myofibroblasts in different types of fibrotic liver diseases is unresolved. To identify distinct populations of myofibroblasts and quantify their contribution to hepatic fibrosis of two different etiologies, collagen-α1(I)-GFP mice were subjected to hepatotoxic (carbon tetrachloride; CCl4) or cholestatic (bile duct ligation; BDL) liver injury. All myofibroblasts were purified by flow cytometry of GFP(+) cells and then different subsets identified by phenotyping. Liver resident activated hepatic stellate cells (aHSCs) and activated portal fibroblasts (aPFs) are the major source (>95%) of fibrogenic myofibroblasts in these models of liver fibrosis in mice. As previously reported using other methodologies, hepatic stellate cells (HSCs) are the major source of myofibroblasts (>87%) in CCl4 liver injury. However, aPFs are a major source of myofibroblasts in cholestatic liver injury, contributing >70% of myofibroblasts at the onset of injury (5 d BDL). The relative contribution of aPFs decreases with progressive injury, as HSCs become activated and contribute to the myofibroblast population (14 and 20 d BDL). Unlike aHSCs, aPFs respond to stimulation with taurocholic acid and IL-25 by induction of collagen-α1(I) and IL-13, respectively. Furthermore, BDL-activated PFs express high levels of collagen type I and provide stimulatory signals to HSCs. Gene expression analysis identified several novel markers of aPFs, including a mesothelial-specific marker mesothelin. PFs may play a critical role in the pathogenesis of cholestatic liver fibrosis and, therefore, serve as an attractive target for antifibrotic therapy.


Subject(s)
Liver Cirrhosis/pathology , Liver/pathology , Myofibroblasts/pathology , Animals , Carbon Tetrachloride/toxicity , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury/pathology , Cholestasis/complications , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Disease Models, Animal , GPI-Linked Proteins/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hepatic Stellate Cells/metabolism , Hepatic Stellate Cells/pathology , Liver/metabolism , Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , Mesothelin , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myofibroblasts/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Vitamin A/metabolism
18.
J Clin Invest ; 124(7): 2921-34, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24937432

ABSTRACT

Activation and accumulation of cardiac fibroblasts, which result in excessive extracellular matrix deposition and consequent mechanical stiffness, myocyte uncoupling, and ischemia, are key contributors to heart failure progression. Recently, endothelial-to-mesenchymal transition (EndoMT) and the recruitment of circulating hematopoietic progenitors to the heart have been reported to generate substantial numbers of cardiac fibroblasts in response to pressure overload-induced injury; therefore, these processes are widely considered to be promising therapeutic targets. Here, using multiple independent murine Cre lines and a collagen1a1-GFP fusion reporter, which specifically labels fibroblasts, we found that following pressure overload, fibroblasts were not derived from hematopoietic cells, EndoMT, or epicardial epithelial-to-mesenchymal transition. Instead, pressure overload promoted comparable proliferation and activation of two resident fibroblast lineages, including a previously described epicardial population and a population of endothelial origin. Together, these data present a paradigm for the origins of cardiac fibroblasts during development and in fibrosis. Furthermore, these data indicate that therapeutic strategies for reducing pathogenic cardiac fibroblasts should shift from targeting presumptive EndoMT or infiltrating hematopoietically derived fibroblasts, toward common pathways upregulated in two endogenous fibroblast populations.


Subject(s)
Heart Failure/pathology , Myocardium/pathology , Animals , Biomarkers/metabolism , Blood Pressure , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Lineage , Collagen Type I/genetics , Collagen Type I/metabolism , Collagen Type I, alpha 1 Chain , Endocardium/metabolism , Endocardium/pathology , Epithelial-Mesenchymal Transition , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis , Gene Expression Profiling , Heart Failure/etiology , Heart Failure/physiopathology , Male , Mice , Mice, Knockout , Mice, Transgenic , Myocardium/metabolism , Pericardium/metabolism , Pericardium/pathology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
19.
PLoS Genet ; 10(2): e1004114, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24586179

ABSTRACT

Recent interest has focused on the importance of the nucleus and associated nucleoskeleton in regulating changes in cardiac gene expression in response to biomechanical load. Mutations in genes encoding proteins of the inner nuclear membrane and nucleoskeleton, which cause cardiomyopathy, also disrupt expression of a biomechanically responsive gene program. Furthermore, mutations in the outer nuclear membrane protein Nesprin 1 and 2 have been implicated in cardiomyopathy. Here, we identify for the first time a role for the outer nuclear membrane proteins, Nesprin 1 and Nesprin 2, in regulating gene expression in response to biomechanical load. Ablation of both Nesprin 1 and 2 in cardiomyocytes, but neither alone, resulted in early onset cardiomyopathy. Mutant cardiomyocytes exhibited altered nuclear positioning, shape, and chromatin positioning. Loss of Nesprin 1 or 2, or both, led to impairment of gene expression changes in response to biomechanical stimuli. These data suggest a model whereby biomechanical signals are communicated from proteins of the outer nuclear membrane, to the inner nuclear membrane and nucleoskeleton, to result in changes in gene expression required for adaptation of the cardiomyocyte to changes in biomechanical load, and give insights into etiologies underlying cardiomyopathy consequent to mutations in Nesprin 1 and 2.


Subject(s)
Cardiomyopathies/genetics , Myocardium/metabolism , Nerve Tissue Proteins/genetics , Nuclear Proteins/genetics , Animals , Biomechanical Phenomena , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Nucleus/metabolism , Cytoskeletal Proteins , Gene Expression Regulation , Humans , Mice , Mutation , Myocytes, Cardiac/metabolism , Nerve Tissue Proteins/metabolism , Nuclear Envelope/genetics , Nuclear Envelope/metabolism , Nuclear Matrix/metabolism , Nuclear Proteins/metabolism
20.
J Vis Exp ; (79)2013 Sep 06.
Article in English | MEDLINE | ID: mdl-24056408

ABSTRACT

Cultured neonatal cardiomyocytes have long been used to study myofibrillogenesis and myofibrillar functions. Cultured cardiomyocytes allow for easy investigation and manipulation of biochemical pathways, and their effect on the biomechanical properties of spontaneously beating cardiomyocytes. The following 2-day protocol describes the isolation and culture of neonatal mouse cardiomyocytes. We show how to easily dissect hearts from neonates, dissociate the cardiac tissue and enrich cardiomyocytes from the cardiac cell-population. We discuss the usage of different enzyme mixes for cell-dissociation, and their effects on cell-viability. The isolated cardiomyocytes can be subsequently used for a variety of morphological, electrophysiological, biochemical, cell-biological or biomechanical assays. We optimized the protocol for robustness and reproducibility, by using only commercially available solutions and enzyme mixes that show little lot-to-lot variability. We also address common problems associated with the isolation and culture of cardiomyocytes, and offer a variety of options for the optimization of isolation and culture conditions.


Subject(s)
Cytological Techniques/methods , Myocytes, Cardiac/cytology , Animals , Animals, Newborn , Cell Culture Techniques/methods , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...