Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Clin Invest ; 133(10)2023 05 15.
Article in English | MEDLINE | ID: mdl-37183820

ABSTRACT

Despite the prevalence of pericytes in the microvasculature of the heart, their role during ischemia-induced remodeling remains unclear. We used multiple lineage-tracing mouse models and found that pericytes migrated to the injury site and expressed profibrotic genes, coinciding with increased vessel leakage after myocardial infarction (MI). Single-cell RNA-Seq of cardiac pericytes at various time points after MI revealed the temporally regulated induction of genes related to vascular permeability, extracellular matrix production, basement membrane degradation, and TGF-ß signaling. Deleting TGF-ß receptor 1 in chondroitin sulfate proteoglycan 4-expressing (Cspg4-expressing) cells reduced fibrosis following MI, leading to a transient improvement in the cardiac ejection fraction. Furthermore, genetic ablation of Cspg4-expressing cells resulted in excessive vascular permeability, a decline in cardiac function, and increased mortality in the second week after MI. These data reveal an essential role for cardiac pericytes in the control of vascular homeostasis and the fibrotic response after acute ischemic injury, information that will help guide the development of novel strategies to preserve vascular integrity and attenuate pathological cardiac remodeling.


Subject(s)
Myocardial Infarction , Pericytes , Mice , Animals , Pericytes/metabolism , Myocardial Infarction/metabolism , Heart , Fibrosis , Extracellular Matrix/metabolism , Ventricular Remodeling/genetics , Myocardium/metabolism
3.
JACC Basic Transl Sci ; 5(5): 432-443, 2020 May.
Article in English | MEDLINE | ID: mdl-32478206

ABSTRACT

Cardiac fibrosis is a pathological process associated with various forms of heart failure. This study identified latent transforming growth factor-ß binding protein 2, cartilage oligomeric matrix protein, and cartilage intermediate layer protein 1 as potential biomarkers for cardiac fibrosis. All 3 encoded proteins showed increased expression in fibroblasts after transforming growth factor-ß stimulation in vitro and localized specifically to fibrotic regions in vivo. Of the 3, only the full-length cartilage intermediate layer protein 1 showed a significant decrease in circulating levels in patients with heart failure compared with healthy volunteers. Further studies on these 3 proteins will lead to a better understanding of their biomarker potential for cardiac fibrosis.

4.
Transl Res ; 209: 121-137, 2019 07.
Article in English | MEDLINE | ID: mdl-30930180

ABSTRACT

Cardiovascular disease is a leading cause of mortality in the world and is exacerbated by the presence of cardiac fibrosis, defined by the accumulation of noncontractile extracellular matrix proteins. Cardiac fibrosis is directly linked to cardiac dysfunction and increased risk of arrhythmia. Despite its prevalence, there is a lack of efficacious therapies for inhibiting or reversing cardiac fibrosis, largely due to the complexity of the cell types and signaling pathways involved. Ongoing research has aimed to understand the mechanisms of cardiac fibrosis and develop new therapies for treating scar formation. Major approaches include preventing the formation of scar tissue and replacing fibrous tissue with functional cardiomyocytes. While targeting the renin-angiotensin-aldosterone system is currently used as the standard line of therapy for heart failure, there has been increased interest in inhibiting the transforming growth factor-ß signaling pathway due its established role in cardiac fibrosis. Significant advances in cell transplantation therapy and biomaterials engineering have also demonstrated potential in regenerating the myocardium. Novel techniques, such as cellular direct reprogramming, and molecular targets, such as noncoding RNAs and epigenetic modifiers, are uncovering novel therapeutic options targeting fibrosis. This review provides an overview of current approaches and discuss future directions for treating cardiac fibrosis.


Subject(s)
Molecular Targeted Therapy , Myocardium/pathology , Animals , Biocompatible Materials/pharmacology , Fibrosis , Humans , Renin-Angiotensin System , Signal Transduction
5.
Circulation ; 138(12): 1224-1235, 2018 09 18.
Article in English | MEDLINE | ID: mdl-29950403

ABSTRACT

BACKGROUND: Genetic diversity and the heterogeneous nature of cardiac fibroblasts (CFbs) have hindered characterization of the molecular mechanisms that regulate cardiac fibrosis. The Hybrid Mouse Diversity Panel offers a valuable tool to examine genetically diverse cardiac fibroblasts and their role in fibrosis. METHODS: Three strains of mice (C57BL/6J, C3H/HeJ, and KK/HlJ) were selected from the Hybrid Mouse Diversity Panel and treated with either isoproterenol (ISO) or saline by an intraperitoneally implanted osmotic pump. After 21 days, cardiac function and levels of fibrosis were measured by echocardiography and trichrome staining, respectively. Activation and proliferation of CFbs were measured by in vitro and in vivo assays under normal and injury conditions. RNA sequencing was done on isolated CFbs from each strain. Results were analyzed by Ingenuity Pathway Analysis and validated by reverse transcription-qPCR, immunohistochemistry, and ELISA. RESULTS: ISO treatment in C57BL/6J, C3H/HeJ, and KK/HlJ mice resulted in minimal, moderate, and extensive levels of fibrosis, respectively (n=7-8 hearts per condition). Isolated CFbs treated with ISO exhibited strain-specific increases in the levels of activation but showed comparable levels of proliferation. Similar results were found in vivo, with fibroblast activation, and not proliferation, correlating with the differential levels of cardiac fibrosis after ISO treatment. RNA sequencing revealed that CFbs from each strain exhibit unique gene expression changes in response to ISO. We identified Ltbp2 as a commonly upregulated gene after ISO treatment. Expression of LTBP2 was elevated and specifically localized in the fibrotic regions of the myocardium after injury in mice and in human heart failure patients. CONCLUSIONS: This study highlights the importance of genetic variation in cardiac fibrosis by using multiple inbred mouse strains to characterize CFbs and their response to ISO treatment. Our data suggest that, although fibroblast activation is a response that parallels the extent of scar formation, proliferation may not necessarily correlate with levels of fibrosis. In addition, by comparing CFbs from multiple strains, we identified pathways as potential therapeutic targets and LTBP2 as a marker for fibrosis, with relevance to patients with underlying myocardial fibrosis.


Subject(s)
Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cell Proliferation , Fibroblasts/pathology , Genetic Variation , Latent TGF-beta Binding Proteins/genetics , Animals , Cardiomyopathies/chemically induced , Cardiomyopathies/metabolism , Cells, Cultured , Disease Models, Animal , Female , Fibroblasts/metabolism , Fibrosis , Genetic Predisposition to Disease , Isoproterenol , Latent TGF-beta Binding Proteins/metabolism , Mice, Inbred C3H , Mice, Inbred C57BL , Phenotype , Species Specificity , Transcriptome
6.
Nat Commun ; 9(1): 754, 2018 02 21.
Article in English | MEDLINE | ID: mdl-29467410

ABSTRACT

The cellular mechanisms driving cardiac tissue formation remain poorly understood, largely due to the structural and functional complexity of the heart. It is unclear whether newly generated myocytes originate from cardiac stem/progenitor cells or from pre-existing cardiomyocytes that re-enter the cell cycle. Here, we identify the source of new cardiomyocytes during mouse development and after injury. Our findings suggest that cardiac progenitors maintain proliferative potential and are the main source of cardiomyocytes during development; however, the onset of αMHC expression leads to reduced cycling capacity. Single-cell RNA sequencing reveals a proliferative, "progenitor-like" population abundant in early embryonic stages that decreases to minimal levels postnatally. Furthermore, cardiac injury by ligation of the left anterior descending artery was found to activate cardiomyocyte proliferation in neonatal but not adult mice. Our data suggest that clonal dominance of differentiating progenitors mediates cardiac development, while a distinct subpopulation of cardiomyocytes may have the potential for limited proliferation during late embryonic development and shortly after birth.


Subject(s)
Heart Injuries/pathology , Heart/growth & development , Myocytes, Cardiac/cytology , Animals , Animals, Newborn , Cell Differentiation , Cell Lineage , Cell Proliferation , Embryonic Stem Cells/cytology , Female , Fetal Heart/cytology , Fetal Heart/growth & development , Heart Injuries/genetics , Male , Mice , Mice, Transgenic , Myoblasts, Cardiac/cytology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/metabolism , Pericardium/cytology , Pericardium/embryology , Pericardium/growth & development , Pregnancy , Sequence Analysis, RNA
7.
Genesis ; 55(8)2017 08.
Article in English | MEDLINE | ID: mdl-28589709

ABSTRACT

Nkx2-5 is a homeobox-containing transcriptional regulator that serves as one of the earliest markers of cardiac lineage commitment. To study the role of Nkx2-5-expressing progenitors at specific time points in cardiac development, we have generated a novel and inducible NKX2-5 mouse line by knocking in a CreER cassette into the Nkx2-5 genomic locus, while preserving the endogenous Nkx2-5 gene to avoid haploinsufficiency. We evaluated the specificity and efficiency of CreER activity after 4-OHT injection by crossing Nkx2-5CreER/+ mice with a Rosa26tdT/+ reporter strain. Our immunohistochemistry results confirmed Cre-induced tdTomato expression specifically in cells expressing Nkx2-5. These cells were mainly cardiomyocytes and were observed in the embryonic heart as early as day 9.5. Additionally, quantitative polymerase chain reaction on postnatal hearts showed enriched expression of Nkx2-5 in isolated tdTomato-expressing cells. No tdTomato expression was observed in Nkx2-5CreER/+ ;Rosa26tdT/+ mice in the absence of 4-OHT, confirming the inducible nature of CreER activity. The Nkx2-5/CreER mouse model described in this article will serve as an invaluable tool to trace myocardial lineage and to temporally induce genetic manipulation in a selective population of cardiac progenitors during embryonic development and in the adult heart.


Subject(s)
Gene Targeting/methods , Genetic Engineering/methods , Heart/embryology , Homeobox Protein Nkx-2.5/genetics , Myocytes, Cardiac/metabolism , Animals , Cell Lineage , Homeobox Protein Nkx-2.5/metabolism , Integrases/genetics , Integrases/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Myocytes, Cardiac/cytology , Transgenes
8.
Mol Biol Cell ; 26(4): 594-604, 2015 Feb 15.
Article in English | MEDLINE | ID: mdl-25518935

ABSTRACT

Apolipoprotein (apo) B is an obligatory component of very low density lipoprotein (VLDL), and its cotranslational and posttranslational modifications are important in VLDL synthesis, secretion, and hepatic lipid homeostasis. ApoB100 contains 25 cysteine residues and eight disulfide bonds. Although these disulfide bonds were suggested to be important in maintaining apoB100 function, neither the specific oxidoreductase involved nor the direct role of these disulfide bonds in apoB100-lipidation is known. Here we used RNA knockdown to evaluate both MTP-dependent and -independent roles of PDI1 in apoB100 synthesis and lipidation in McA-RH7777 cells. Pdi1 knockdown did not elicit any discernible detrimental effect under normal, unstressed conditions. However, it decreased apoB100 synthesis with attenuated MTP activity, delayed apoB100 oxidative folding, and reduced apoB100 lipidation, leading to defective VLDL secretion. The oxidative folding-impaired apoB100 was secreted mainly associated with LDL instead of VLDL particles from PDI1-deficient cells, a phenotype that was fully rescued by overexpression of wild-type but not a catalytically inactive PDI1 that fully restored MTP activity. Further, we demonstrate that PDI1 directly interacts with apoB100 via its redox-active CXXC motifs and assists in the oxidative folding of apoB100. Taken together, these findings reveal an unsuspected, yet key role for PDI1 in oxidative folding of apoB100 and VLDL assembly.


Subject(s)
Apolipoprotein B-100/chemistry , Protein Disulfide-Isomerases/physiology , Animals , Apolipoprotein B-100/biosynthesis , Cell Line , Endoplasmic Reticulum/metabolism , Gene Knockdown Techniques , Homeostasis , Lipid Metabolism , Mice , Oxidative Stress , Protein Disulfide-Isomerases/chemistry , Protein Disulfide-Isomerases/genetics , Rats
SELECTION OF CITATIONS
SEARCH DETAIL
...