Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Am J Physiol Cell Physiol ; 325(5): C1326-C1335, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37781738

ABSTRACT

Muscular dystrophy exerts significant and dramatic impacts on affected patients, including progressive muscle wasting leading to lung and heart failure, and results in severely curtailed lifespan. Although the focus for many years has been on the dysfunction induced by the loss of function of dystrophin or related components of the striated muscle costamere, recent studies have demonstrated that accompanying pathologies, particularly muscle fibrosis, also contribute adversely to patient outcomes. A significant body of research has now shown that therapeutically targeting these accompanying pathologies via their underlying molecular mechanisms may provide novel approaches to patient management that can complement the current standard of care. In this review, we discuss the interplay between muscle fibrosis and muscular dystrophy pathology. A better understanding of these processes will contribute to improved patient care options, restoration of muscle function, and reduced patient morbidity and mortality.


Subject(s)
Heart Failure , Muscular Dystrophy, Duchenne , Humans , Muscular Dystrophy, Duchenne/pathology , Muscle, Skeletal/pathology , Fibrosis , Myofibrils/pathology , Heart Failure/pathology
2.
Eur Heart J ; 43(45): 4739-4750, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36200607

ABSTRACT

AIMS: In response to pro-fibrotic signals, scleraxis regulates cardiac fibroblast activation in vitro via transcriptional control of key fibrosis genes such as collagen and fibronectin; however, its role in vivo is unknown. The present study assessed the impact of scleraxis loss on fibroblast activation, cardiac fibrosis, and dysfunction in pressure overload-induced heart failure. METHODS AND RESULTS: Scleraxis expression was upregulated in the hearts of non-ischemic dilated cardiomyopathy patients, and in mice subjected to pressure overload by transverse aortic constriction (TAC). Tamoxifen-inducible fibroblast-specific scleraxis knockout (Scx-fKO) completely attenuated cardiac fibrosis, and significantly improved cardiac systolic function and ventricular remodelling, following TAC compared to Scx+/+ TAC mice, concomitant with attenuation of fibroblast activation. Scleraxis deletion, after the establishment of cardiac fibrosis, attenuated the further functional decline observed in Scx+/+ mice, with a reduction in cardiac myofibroblasts. Notably, scleraxis knockout reduced pressure overload-induced mortality from 33% to zero, without affecting the degree of cardiac hypertrophy. Scleraxis directly regulated transcription of the myofibroblast marker periostin, and cardiac fibroblasts lacking scleraxis failed to upregulate periostin synthesis and secretion in response to pro-fibrotic transforming growth factor ß. CONCLUSION: Scleraxis governs fibroblast activation in pressure overload-induced heart failure, and scleraxis knockout attenuated fibrosis and improved cardiac function and survival. These findings identify scleraxis as a viable target for the development of novel anti-fibrotic treatments.


Subject(s)
Heart Failure , Ventricular Remodeling , Mice , Animals , Fibrosis , Myofibroblasts/metabolism , Cardiomegaly/metabolism , Fibroblasts/metabolism , Heart Failure/pathology , Myocardium/pathology , Mice, Inbred C57BL
3.
Cells ; 11(14)2022 07 13.
Article in English | MEDLINE | ID: mdl-35883637

ABSTRACT

Hypoxia exerts broad effects on cardiomyocyte function and viability, ranging from altered metabolism and mitochondrial physiology to apoptotic or necrotic cell death. The transcriptional coactivator peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) is a key regulator of cardiomyocyte metabolism and mitochondrial function and is down-regulated in hypoxia; however, the underlying mechanism is incompletely resolved. Using primary rat cardiomyocytes coupled with electrophoretic mobility shift and luciferase assays, we report that hypoxia impaired mitochondrial energetics and resulted in an increase in nuclear localization of the Nuclear Factor-κB (NF-κB) p65 subunit, and the association of p65 with the PGC-1α proximal promoter. Tumor necrosis factor α (TNFα), an activator of NF-κB signaling, similarly reduced PGC-1α expression and p65 binding to the PGC-1α promoter in a dose-dependent manner, and TNFα-mediated down-regulation of PGC-1α expression could be reversed by the NF-κB inhibitor parthenolide. RNA-seq analysis revealed that cardiomyocytes isolated from p65 knockout mice exhibited alterations in genes associated with chromatin remodeling. Decreased PGC-1α promoter transactivation by p65 could be partially reversed by the histone deacetylase inhibitor trichostatin A. These results implicate NF-κB signaling, and specifically p65, as a potent inhibitor of PGC-1α expression in cardiac myocyte hypoxia.


Subject(s)
Hypoxia , Myocytes, Cardiac , NF-kappa B , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Animals , Hypoxia/metabolism , Mice , Myocytes, Cardiac/metabolism , NF-kappa B/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Rats , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/pharmacology
4.
Cells ; 11(15)2022 07 23.
Article in English | MEDLINE | ID: mdl-35892569

ABSTRACT

Fibroblasts are stromal cells found in virtually every tissue and organ of the body. For many years, these cells were often considered to be secondary in functional importance to parenchymal cells. Over the past 2 decades, focused research into the roles of fibroblasts has revealed important roles for these cells in the homeostasis of healthy tissue, and has demonstrated that activation of fibroblasts to myofibroblasts is a key step in disease initiation and progression in many tissues, with fibrosis now recognized as not only an outcome of disease, but also a central contributor to tissue dysfunction, particularly in the heart and lungs. With a growing understanding of both fibroblast and myofibroblast heterogeneity, and the deciphering of the humoral and mechanical cues that impact the phenotype of these cells, fibroblast biology is rapidly becoming a major focus in biomedical research. In this review, we provide an overview of fibroblast and myofibroblast biology, particularly in the heart, and including a discussion of pathophysiological processes such as fibrosis and scarring. We then discuss the central role of Canadian researchers in moving this field forwards, particularly in cardiac fibrosis, and highlight some of the major contributions of these individuals to our understanding of fibroblast and myofibroblast biology in health and disease.


Subject(s)
Fibroblasts , Myocardium , Biology , Canada , Cell Differentiation , Fibroblasts/pathology , Fibrosis , Humans , Myocardium/pathology
5.
Cells ; 11(9)2022 04 27.
Article in English | MEDLINE | ID: mdl-35563778

ABSTRACT

Fibrosis is an energy-intensive process requiring the activation of fibroblasts to myofibroblasts, resulting in the increased synthesis of extracellular matrix proteins. Little is known about the transcriptional control of energy metabolism in cardiac fibroblast activation, but glutaminolysis has been implicated in liver and lung fibrosis. Here we explored how pro-fibrotic TGFß and its effector scleraxis, which drive cardiac fibroblast activation, regulate genes involved in glutaminolysis, particularly the rate-limiting enzyme glutaminase (GLS1). The GLS1 inhibitor CB-839 attenuated TGFß-induced fibroblast activation. Cardiac fibroblast activation to myofibroblasts by scleraxis overexpression increased glutaminolysis gene expression, including GLS1, while cardiac fibroblasts from scleraxis-null mice showed reduced expression. TGFß induced GLS1 expression and increased intracellular glutamine and glutamate levels, indicative of increased glutaminolysis, but in scleraxis knockout cells, these measures were attenuated, and the response to TGFß was lost. The knockdown of scleraxis in activated cardiac fibroblasts reduced GLS1 expression by 75%. Scleraxis transactivated the human GLS1 promoter in luciferase reporter assays, and this effect was dependent on a key scleraxis-binding E-box motif. These results implicate scleraxis-mediated GLS1 expression as a key regulator of glutaminolysis in cardiac fibroblast activation, and blocking scleraxis in this process may provide a means of starving fibroblasts of the energy required for fibrosis.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors , Glutaminase , Pulmonary Fibrosis , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Fibroblasts/metabolism , Glutaminase/genetics , Mice , Myofibroblasts/metabolism , Pulmonary Fibrosis/metabolism , Transforming Growth Factor beta/metabolism
6.
Cell Mol Life Sci ; 79(4): 193, 2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35298717

ABSTRACT

Aberrant insulin-like growth factor 1 (IGF-1) signaling has been proposed as a contributing factor to the development of neurodegenerative disorders including diabetic neuropathy, and delivery of exogenous IGF-1 has been explored as a treatment for Alzheimer's disease and amyotrophic lateral sclerosis. However, the role of autocrine/paracrine IGF-1 in neuroprotection has not been well established. We therefore used in vitro cell culture systems and animal models of diabetic neuropathy to characterize endogenous IGF-1 in sensory neurons and determine the factors regulating IGF-1 expression and/or affecting neuronal health. Single-cell RNA sequencing (scRNA-Seq) and in situ hybridization analyses revealed high expression of endogenous IGF-1 in non-peptidergic neurons and satellite glial cells (SGCs) of dorsal root ganglia (DRG). Brain cortex and DRG had higher IGF-1 gene expression than sciatic nerve. Bidirectional transport of IGF-1 along sensory nerves was observed. Despite no difference in IGF-1 receptor levels, IGF-1 gene expression was significantly (P < 0.05) reduced in liver and DRG from streptozotocin (STZ)-induced type 1 diabetic rats, Zucker diabetic fatty (ZDF) rats, mice on a high-fat/ high-sugar diet and db/db type 2 diabetic mice. Hyperglycemia suppressed IGF-1 gene expression in cultured DRG neurons and this was reversed by exogenous IGF-1 or the aldose reductase inhibitor sorbinil. Transcription factors, such as NFAT1 and CEBPß, were also less enriched at the IGF-1 promoter in DRG from diabetic rats vs control rats. CEBPß overexpression promoted neurite outgrowth and mitochondrial respiration, both of which were blunted by knocking down or blocking IGF-1. Suppression of endogenous IGF-1 in diabetes may contribute to neuropathy and its upregulation at the transcriptional level by CEBPß can be a promising therapeutic approach.


Subject(s)
Aging/metabolism , Axons/pathology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetes Mellitus, Experimental/pathology , Energy Metabolism , Insulin-Like Growth Factor I/metabolism , Sensory Receptor Cells/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Axons/drug effects , Axons/metabolism , Base Sequence , CCAAT-Enhancer-Binding Protein-beta/genetics , Cell Respiration/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/pathology , Diabetes Mellitus, Type 2/genetics , Diabetes Mellitus, Type 2/pathology , Energy Metabolism/drug effects , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Gene Expression Regulation/drug effects , Glycolysis/drug effects , HEK293 Cells , Humans , Insulin-Like Growth Factor I/genetics , Liver/metabolism , Male , Mitochondria/drug effects , Mitochondria/metabolism , NFATC Transcription Factors/metabolism , Neuronal Outgrowth/drug effects , Polymers/metabolism , Promoter Regions, Genetic/genetics , Protein Transport/drug effects , Rats, Sprague-Dawley , Sensory Receptor Cells/pathology , Signal Transduction/drug effects
7.
Cell Signal ; 85: 110066, 2021 09.
Article in English | MEDLINE | ID: mdl-34146658

ABSTRACT

Cardiac fibrosis is characteristic of the end stage in nearly all forms of heart disease. Accumulation of extracellular matrix in the myocardium leads to increased risk of arrhythmia and impaired cardiac function, and ultimately progression to heart failure. Despite the critical need to slow or reverse development of cardiac fibrosis to maintain cardiac function, there are no approved therapies that directly target the extracellular matrix. Research into the underlying causes and therapeutic targets has been hampered, in part, by the lack of a clear marker for cardiac fibroblasts - the cells responsible for regulating extracellular matrix turnover. Lineage tracing studies as well as single-cell RNA sequencing studies have provided new insights into cardiac fibroblast origins and heterogeneity. Moreover, a greater understanding of pathways governing fibroblast activation during ischemic and non-ischemic cardiac remodeling and their communication with other inflammatory and cardiac cells may lead to novel therapeutic targets to slow or reverse fibrotic remodeling. The special issue of Cellular Signaling entitled "Cardiac Fibrosis: Pathobiology and Therapeutic Targets" is comprised of review articles in which these topics, as well as important open questions for future investigation, are discussed.


Subject(s)
Fibroblasts , Myocardium , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibrosis , Heart , Humans , Myocardium/metabolism
8.
Cell Tissue Res ; 385(3): 753-768, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34057573

ABSTRACT

Fibroblast growth factor 2 (FGF2), produced as high (Hi-) and low (Lo-) molecular weight isoforms, is implicated in cardiac response to injury. The role of endogenous FGF2 isoforms during chronic stress is not well defined. We investigated the effects of endogenous Hi-FGF2 in a mouse model of simulated pressure-overload stress achieved by transverse aortic constriction (TAC) surgery. Hi-FGF2 knockout mice, expressing only Lo-FGF2, FGF2(Lo), and wild-type mice, FGF2(WT), expressing both Hi-FGF2 and Lo-FGF2, were used. By echocardiography, a decline in systolic function was observed in FGF2(WT) but not FGF2(Lo) mice compared to corresponding sham-operated animals at 4-8 weeks post-TAC surgery. TAC surgery increased markers of myocardial stress/damage including B-type natriuretic peptide (BNP) and the pro-cell death protein BCL2/adenovirus E1B 19 kDa protein-interacting protein-3 (Bnip3) in FGF2(WT) but not FGF2(Lo) mice. In FGF2(Lo) mice, cardiac levels of activated FGF receptor 1 (FGFR1), and downstream signals, including phosphorylated mTOR and p70S6 kinase, were elevated post-TAC. Finally, NR1D1 (nuclear receptor subfamily 1 group D member 1), implicated in cardioprotection from pressure-overload stress, was downregulated or upregulated in the presence or absence, respectively, of Hi-FGF2 expression, post-TAC surgery. In wild-type cardiomyocyte cultures, endothelin-1 (added to simulate pressure-overload signals) caused NR1D1 downregulation and BNP upregulation, similar to the effect of TAC surgery on the FGF2(WT) mice. The NR1D1 agonist SR9009 prevented BNP upregulation, simulating post-TAC findings in FGF2(Lo) mice. We propose that elimination of Hi-FGF2 is cardioprotective during pressure-overload by increasing FGFR1-associated signaling and NR1D1 expression.


Subject(s)
Blood Pressure/genetics , Fibroblast Growth Factor 2/therapeutic use , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , Animals , Male , Mice , Mice, Knockout , Rats , Signal Transduction
9.
Cell Signal ; 80: 109903, 2021 04.
Article in English | MEDLINE | ID: mdl-33370581

ABSTRACT

Angiotensin II (Ang II) is a primary mediator of profibrotic signaling in the heart and more specifically, the cardiac fibroblast. Ang II-mediated cardiomyocyte hypertrophy in combination with cardiac fibroblast proliferation, activation, and extracellular matrix production compromise cardiac function and increase mortality in humans. Profibrotic actions of Ang II are mediated by increasing production of fibrogenic mediators (e.g. transforming growth factor beta, scleraxis, osteopontin, and periostin), recruitment of immune cells, and via increased reactive oxygen species generation. Drugs that inhibit Ang II production or action, collectively referred to as renin angiotensin system (RAS) inhibitors, are first line therapeutics for heart failure. Moreover, transient RAS inhibition has been found to persistently alter hypertensive cardiac fibroblast responses to injury providing a useful tool to identify novel therapeutic targets. This review summarizes the profibrotic actions of Ang II and the known impact of RAS inhibition on cardiac fibroblast phenotype and cardiac remodeling.


Subject(s)
Fibroblasts/metabolism , Renin-Angiotensin System , Angiotensin II/genetics , Angiotensin II/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Epigenesis, Genetic , Fibroblasts/cytology , Fibrosis , Humans , Renin-Angiotensin System/drug effects , Signal Transduction , Transforming Growth Factor beta/metabolism
10.
Matrix Biol ; 91-92: 92-108, 2020 09.
Article in English | MEDLINE | ID: mdl-32422329

ABSTRACT

Fibrosis is characterized by excessive deposition of extracellular matrix components such as collagen in tissues or organs. Fibrosis can develop in the heart, kidneys, liver, skin or any other body organ in response to injury or maladaptive reparative processes, reducing overall function and leading eventually to organ failure. A variety of cellular and molecular signaling mechanisms are involved in the pathogenesis of fibrosis. The renin-angiotensin-aldosterone system (RAAS) interacts with the potent Transforming Growth Factor ß (TGFß) pro-fibrotic pathway to mediate fibrosis in many cell and tissue types. RAAS consists of both classical and alternative pathways, which act to potentiate or antagonize fibrotic signaling mechanisms, respectively. This review provides an overview of recent literature describing the roles of RAAS in the pathogenesis of fibrosis, particularly in the liver, heart, kidney and skin, and with a focus on RAAS interactions with TGFß signaling. Targeting RAAS to combat fibrosis represents a promising therapeutic approach, particularly given the lack of strategies for treating fibrosis as its own entity, thus animal and clinical studies to examine the impact of natural and synthetic substances to alter RAAS signaling as a means to treat fibrosis are reviewed as well.


Subject(s)
Extracellular Matrix Proteins/genetics , Extracellular Matrix/metabolism , Fibroblasts/drug effects , Fibrosis/prevention & control , Molecular Targeted Therapy/methods , Renin-Angiotensin System/drug effects , Amides/therapeutic use , Angiotensins/antagonists & inhibitors , Angiotensins/genetics , Angiotensins/metabolism , Animals , Benzimidazoles/therapeutic use , Biphenyl Compounds/therapeutic use , Extracellular Matrix/chemistry , Extracellular Matrix Proteins/metabolism , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/genetics , Fibrosis/metabolism , Fibrosis/pathology , Fumarates/therapeutic use , Gene Expression Regulation , Humans , Kidney/drug effects , Kidney/metabolism , Kidney/pathology , Liver/drug effects , Liver/metabolism , Liver/pathology , Myocardium/metabolism , Myocardium/pathology , Pyridones/therapeutic use , Renin-Angiotensin System/genetics , Signal Transduction , Skin/drug effects , Skin/metabolism , Skin/pathology , Tetrazoles/therapeutic use , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
11.
Can J Physiol Pharmacol ; 98(7): 459-465, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32027517

ABSTRACT

Interstitial fibrosis is a histopathological hallmark of hypertrophic cardiomyopathy (HCM). Although extracellular matrix (ECM) biomarkers, including matrix metalloproteinases, are overexpressed in HCM patients, they do not correlate with sudden cardiac death (SCD) risk. The objective of this study was to determine whether scleraxis, a transcription factor that regulates collagen gene expression, is detectable in HCM patients and correlates with disease burden. Between 2017 and 2018, a total of 46 HCM patients were enrolled (58 ± 14 years (31 males, 15 females)) with a mean 5 year SCD risk of 2.3% ± 1.3%. Cardiac MRI confirmed HCM in all patients with a mean interventricular septal thickness of 20 ± 2 mm. Late gadolinium enhancement (LGE) was present in 32 (70%) study participants occupying 18% ± 7% of the left ventricular (LV) myocardium. Serum scleraxis levels were significantly higher in the HCM patients by approximately twofold as compared to controls (0.76 ± 0.06 versus 0.32 ± 0.02 ng/mL, p < 0.05). No correlation was demonstrated between serum scleraxis levels and markers of disease severity in HCM patients, including maximum LV wall thickness, %LGE, and SCD risk factors. Serum scleraxis is elevated in the HCM population. Future studies are warranted to evaluate the prognostic value of scleraxis in identifying high-risk HCM patients who require aggressive management for prevention of SCD.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/blood , Cardiomyopathy, Hypertrophic/diagnosis , Heart Ventricles/pathology , Myocardium/pathology , Adult , Aged , Biomarkers/blood , Cardiomyopathy, Hypertrophic/blood , Cardiomyopathy, Hypertrophic/pathology , Contrast Media/administration & dosage , Echocardiography, Doppler, Color , Female , Fibrosis , Gadolinium DTPA/administration & dosage , Heart Ventricles/diagnostic imaging , Humans , Magnetic Resonance Imaging/methods , Male , Middle Aged , Prognosis , Risk Factors , Severity of Illness Index
12.
J Cardiovasc Dev Dis ; 6(3)2019 Aug 16.
Article in English | MEDLINE | ID: mdl-31426390

ABSTRACT

Fibrosis occurs when the synthesis of extracellular matrix outpaces its degradation, and over time can negatively impact tissue and organ function. In the case of cardiac fibrosis, contraction and relaxation of the heart can be impaired to the point of precipitating heart failure, while at the same time fibrosis can result in arrhythmias due to altered electrical properties of the myocardium. The critical event in the evolution of cardiac fibrosis is the phenotype conversion of cardiac fibroblasts to their overly-active counterparts, myofibroblasts: cells demarked by their expression of novel markers such as periostin, by their gain of contractile activity, and by their pronounced and prolonged increase in the production of extracellular matrix components such as collagens. The phenotype change is dramatic, and can be triggered by many stimuli, including mechanical force, inflammatory cytokines, and growth factors. This review will explore fibroblast to myofibroblast transition mechanisms and will consider the therapeutic potential of targeting this process as a means to arrest or even reverse cardiac fibrosis.

13.
Can J Physiol Pharmacol ; 97(6): 493-497, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30468625

ABSTRACT

Fibroblasts have long been recognized as important stromal cells, playing key roles in synthesizing and maintaining the extracellular matrix, but historically were treated as a relatively uniform cell type. Studies in recent years have revealed a surprising level of heterogeneity of fibroblasts across tissues, and even within organs such as the skin and heart. This heterogeneity may have functional consequences, including during stress and disease. While the field has moved forward quickly to begin to address the scientific import of this heterogeneity, the descriptive language used for these cells has not kept pace, particularly when considering the phenotype changes that occur as fibroblasts convert to myofibroblasts in response to injury. We discuss here the nature and sources of the heterogeneity of fibroblasts, and review how our understanding of the complexity of the fibroblast to myofibroblast phenotype conversion has changed with increasing scrutiny. We propose that the time is opportune to reevaluate how we name and describe these cells, particularly as they transition to myofibroblasts through discrete stages. A standardized nomenclature is essential to address the confusion that currently exists in the literature as to the usage of terms like myofibroblast and the description of fibroblast phenotype changes in disease.


Subject(s)
Fibroblasts/cytology , Phenotype , Terminology as Topic , Animals , Humans
14.
Compr Physiol ; 9(1): 75-125, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30549015

ABSTRACT

Cardiovascular disease leading to heart failure (HF) remains a leading cause of morbidity and mortality worldwide. Improved pharmacological and interventional coronary procedures have led to improved outcomes following acute myocardial infarction. This success has translated into an unforeseen increased incidence in HF. This review summarizes the signaling pathways implicated in the transition to HF following cardiac injury. In addition, we provide an update on cell death signaling and discuss recent advances in cardiac fibrosis as an independent event leading to HF. Finally, we discuss cell-based therapies and their possible use to avert the deteriorating nature of HF. © 2019 American Physiological Society. Compr Physiol 9:75-125, 2019.


Subject(s)
Heart Failure/metabolism , Regenerative Medicine/methods , Signal Transduction , Tissue Engineering/methods , Animals , Heart Failure/pathology , Heart Failure/therapy , Humans , Myocardium/metabolism , Myocardium/pathology
15.
Physiol Rep ; 6(22): e13897, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30488595

ABSTRACT

Many etiologies of heart disease are characterized by expansion and remodeling of the cardiac extracellular matrix (ECM or matrix) which results in cardiac fibrosis. Cardiac fibrosis is mediated in cardiac fibroblasts by TGF-ß1 /R-Smad2/3 signaling. Matrix component proteins are synthesized by activated resident cardiac fibroblasts known as myofibroblasts (MFB). These events are causal to heart failure with diastolic dysfunction and reduced cardiac filling. We have shown that exogenous Ski, a TGF-ß1 /Smad repressor, localizes in the cellular nucleus and deactivates cardiac myofibroblasts. This deactivation is associated with reduction of myofibroblast marker protein expression in vitro, including alpha smooth muscle actin (α-SMA) and extracellular domain-A (ED-A) fibronectin. We hypothesize that Ski also acutely regulates MMP expression in cardiac MFB. While acute Ski overexpression in cardiac MFB in vitro was not associated with any change in intracellular MMP-9 protein expression versus LacZ-treated controls,exogenous Ski caused elevated MMP-9 mRNA expression and increased MMP-9 protein secretion versus controls. Zymographic analysis revealed increased MMP-9-specific gelatinase activity in myofibroblasts overexpressing Ski versus controls. Moreover, Ski expression was attended by reduced paxillin and focal adhesion kinase phosphorylation (FAK - Tyr 397) versus controls. As myofibroblasts are hypersecretory and less motile relative to fibroblasts, Ski's reduction of paxillin and FAK expression may reflect the relative deactivation of myofibroblasts. Thus, in addition to its known antifibrotic effects, Ski overexpression elevates expression and extracellular secretion/release of MMP-9 and thus may facilitate internal cytoskeletal remodeling as well as extracellular ECM components. Further, as acute TGF-ß1 treatment of primary cardiac MFB is known to cause rapid translocation of Ski to the nucleus, our data support an autoregulatory role for Ski in mediating cardiac ECM accumulation.


Subject(s)
Matrix Metalloproteinase 9/metabolism , Myocardium/cytology , Myofibroblasts/metabolism , Proto-Oncogene Proteins/metabolism , Actins/genetics , Actins/metabolism , Animals , Cell Movement , Cells, Cultured , Fibronectins/genetics , Fibronectins/metabolism , Male , Matrix Metalloproteinase 9/genetics , Myocardium/metabolism , Myofibroblasts/physiology , Paxillin/genetics , Paxillin/metabolism , Proto-Oncogene Proteins/genetics , Rats , Rats, Sprague-Dawley
16.
Am J Physiol Heart Circ Physiol ; 315(3): H658-H668, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29906225

ABSTRACT

Numerous physiological and pathological events, from organ development to cancer and fibrosis, are characterized by an epithelial-to-mesenchymal transition (EMT), whereby adherent epithelial cells convert to migratory mesenchymal cells. During cardiac development, proepicardial organ epithelial cells undergo EMT to generate fibroblasts. Subsequent stress or damage induces further phenotype conversion of fibroblasts to myofibroblasts, causing fibrosis via synthesis of an excessive extracellular matrix. We have previously shown that the transcription factor scleraxis is both sufficient and necessary for the conversion of cardiac fibroblasts to myofibroblasts and found that scleraxis knockout reduced cardiac fibroblast numbers by 50%, possibly via EMT attenuation. Scleraxis induced expression of the EMT transcriptional regulators Twist1 and Snai1 via an unknown mechanism. Here, we report that scleraxis binds to E-box consensus sequences within the Twist1 and Snai1 promoters to transactivate these genes directly. Scleraxis upregulates expression of both genes in A549 epithelial cells and in cardiac myofibroblasts. Transforming growth factor-ß induces EMT, fibrosis, and scleraxis expression, and we found that transforming growth factor-ß-mediated upregulation of Twist1 and Snai1 completely depends on the presence of scleraxis. Snai1 knockdown upregulated the epithelial marker E-cadherin; however, this effect was lost after scleraxis overexpression, suggesting that scleraxis may repress E-cadherin expression. Together, these results indicate that scleraxis can regulate EMT via direct transactivation of the Twist1 and Snai1 genes. Given the role of scleraxis in also driving the myofibroblast phenotype, scleraxis appears to be a critical controller of fibroblast genesis and fate in the myocardium and thus may play key roles in wound healing and fibrosis. NEW & NOTEWORTHY The molecular mechanism by which the transcription factor scleraxis mediates Twist1 and Snai1 gene expression was determined. These results reveal a novel means of transcriptional regulation of epithelial-to-mesenchymal transition and demonstrate that transforming growth factor-ß-mediated epithelial-to-mesenchymal transition is dependent on scleraxis, providing a potential target for controlling this process.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Epithelial-Mesenchymal Transition , Nuclear Proteins/metabolism , Snail Family Transcription Factors/metabolism , Twist-Related Protein 1/metabolism , 3T3 Cells , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line, Tumor , Cells, Cultured , Humans , Male , Mice , Myofibroblasts/metabolism , Nuclear Proteins/genetics , Promoter Regions, Genetic , Protein Binding , Rats , Rats, Sprague-Dawley , Snail Family Transcription Factors/genetics , Twist-Related Protein 1/genetics
17.
J Mol Cell Cardiol ; 120: 64-73, 2018 07.
Article in English | MEDLINE | ID: mdl-29750994

ABSTRACT

Remodeling of the cardiac extracellular matrix is responsible for a number of the detrimental effects on heart function that arise secondary to hypertension, diabetes and myocardial infarction. This remodeling consists both of an increase in new matrix protein synthesis, and an increase in the expression of matrix metalloproteinases (MMPs) that degrade existing matrix structures. Previous studies utilizing knockout mice have demonstrated clearly that MMP2 plays a pathogenic role during matrix remodeling, thus it is important to understand the mechanisms that regulate MMP2 gene expression. We have shown that the transcription factor scleraxis is an important inducer of extracellular matrix gene expression in the heart that may also control MMP2 expression. In the present study, we demonstrate that scleraxis directly transactivates the proximal MMP2 gene promoter, resulting in increased histone acetylation, and identify a specific E-box sequence in the promoter to which scleraxis binds. Cardiac myo-fibroblasts isolated from scleraxis knockout mice exhibited dramatically decreased MMP2 expression; however, scleraxis over-expression in knockout cells could rescue this loss. We further show that regulation of MMP2 gene expression by the pro-fibrotic cytokine TGFß occurs via a scleraxis-dependent mechanism: TGFß induces recruitment of scleraxis to the MMP2 promoter, and TGFß was unable to up-regulate MMP2 expression in cells lacking scleraxis due to either gene knockdown or knockout. These results reveal that scleraxis can exert control over both extracellular matrix synthesis and breakdown, and thus may contribute to matrix remodeling in wound healing and disease.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Gene Expression Regulation , Matrix Metalloproteinase 2/genetics , Myocardium/cytology , Myofibroblasts/physiology , Analysis of Variance , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , E-Box Elements/physiology , Extracellular Matrix/genetics , Extracellular Matrix/metabolism , Genetic Vectors , Humans , Male , Mice , Mice, Knockout , NIH 3T3 Cells , Promoter Regions, Genetic , Rats , Rats, Sprague-Dawley , Transcriptional Activation , Transfection , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
18.
Can J Physiol Pharmacol ; 95(10): 1067-1077, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28727928

ABSTRACT

In the vascular system, ageing is accompanied by the accrual of senescent cells and is associated with an increased risk of vascular disease. Endothelial cell (EC) dysfunction is a hallmark of vascular disease and is characterized by decreased angiogenic potential, reduced nitric oxide bioavailability, impaired vasodilation, increased production of ROS, and enhanced inflammation. In ECs, the major producer of nitric oxide is the endothelial nitric oxide synthase (eNOS) enzyme that is encoded by the NOS3 gene. NOS3/eNOS function is tightly regulated at both the transcriptional and post-transcriptional levels to maintain normal vascular function. A key transcriptional regulator of eNOS expression is p53, which has been shown to play a central role in mediating cellular senescence and thereby vascular dysfunction. Herein, we show that, in ECs, the MEOX homeodomain transcription factors decrease the expression of genes involved in angiogenesis, repress eNOS expression at the mRNA and protein levels, and increase the expression of p53. These findings support a role for the MEOX proteins in promoting endothelial dysfunction.


Subject(s)
Aging/metabolism , Blood Vessels/metabolism , Endothelium, Vascular/metabolism , Hemodynamics , Homeodomain Proteins/metabolism , Vascular Diseases/metabolism , Age Factors , Aging/genetics , Animals , Blood Vessels/physiopathology , Cellular Senescence , Endothelium, Vascular/physiopathology , Gene Expression Regulation , Homeodomain Proteins/genetics , Humans , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Signal Transduction , Sirtuin 1/genetics , Sirtuin 1/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Vascular Diseases/genetics , Vascular Diseases/physiopathology
19.
Trends Pharmacol Sci ; 38(5): 448-458, 2017 05.
Article in English | MEDLINE | ID: mdl-28365093

ABSTRACT

In response to myocardial infarction (MI), the wound healing response of the left ventricle (LV) comprises overlapping inflammatory, proliferative, and maturation phases, and the cardiac fibroblast is a key cell type involved in each phase. It has recently been appreciated that, early post-MI, fibroblasts transform to a proinflammatory phenotype and secrete cytokines and chemokines as well as matrix metalloproteinases (MMPs). Later post-MI, fibroblasts are activated to anti-inflammatory and proreparative phenotypes and generate anti-inflammatory and proangiogenic factors and extracellular matrix (ECM) components that form the infarct scar. Additional studies are needed to systematically examine how fibroblast activation shifts over the timeframe of the MI response and how modulation at different activation stages could alter wound healing and LV remodeling in distinct ways. This review summarizes current fibroblast knowledge as the foundation for a discussion of existing knowledge gaps.


Subject(s)
Myocardial Infarction/pathology , Myofibroblasts/pathology , Animals , Humans
20.
Can J Physiol Pharmacol ; 95(10): 1091-1099, 2017 Oct.
Article in English | MEDLINE | ID: mdl-28273426

ABSTRACT

Cardiac fibrosis is a significant global health problem that is closely associated with multiple forms of cardiovascular disease, including myocardial infarction, dilated cardiomyopathy, and diabetes. Fibrosis increases myocardial wall stiffness due to excessive extracellular matrix deposition, causing impaired systolic and diastolic function, and facilitating arrhythmogenesis. As a result, patient morbidity and mortality are often dramatically elevated compared with those with cardiovascular disease but without overt fibrosis, demonstrating that fibrosis itself is both a pathologic response to existing disease and a significant risk factor for exacerbation of the underlying condition. The lack of any specific treatment for cardiac fibrosis in patients suffering from cardiovascular disease is a critical gap in our ability to care for these individuals. Here we provide an overview of the development of cardiac fibrosis, and discuss new research directions that have recently emerged and that may lead to the creation of novel treatments for patients with cardiovascular diseases. Such treatments would, ideally, complement existing therapy by specifically focusing on amelioration of fibrosis.


Subject(s)
Cardiomyopathies/pathology , Extracellular Matrix/pathology , Fibroblasts/pathology , Myocardium/pathology , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/physiopathology , Cardiomyopathies/therapy , Extracellular Matrix/metabolism , Fibroblasts/metabolism , Fibrosis , Humans , Myocardium/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Signal Transduction , Ventricular Remodeling
SELECTION OF CITATIONS
SEARCH DETAIL
...