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










Publication year range
1.
Cell Death Dis ; 12(12): 1105, 2021 11 26.
Article in English | MEDLINE | ID: mdl-34824192

ABSTRACT

Systemic hypoxia is a common element in most perinatal emergencies and is a known driver of Bnip3 expression in the neonatal heart. Bnip3 plays a prominent role in the evolution of necrotic cell death, disrupting ER calcium homeostasis and initiating mitochondrial permeability transition (MPT). Emerging evidence suggests a cardioprotective role for the prostaglandin E1 analog misoprostol during periods of hypoxia, but the mechanisms for this protection are not completely understood. Using a combination of mouse and cell models, we tested if misoprostol is cardioprotective during neonatal hypoxic injury by altering Bnip3 function. Here we report that hypoxia elicits mitochondrial-fragmentation, MPT, reduced ejection fraction, and evidence of necroinflammation, which were abrogated with misoprostol treatment or Bnip3 knockout. Through molecular studies we show that misoprostol leads to PKA-dependent Bnip3 phosphorylation at threonine-181, and subsequent redistribution of Bnip3 from mitochondrial Opa1 and the ER through an interaction with 14-3-3 proteins. Taken together, our results demonstrate a role for Bnip3 phosphorylation in the regulation of cardiomyocyte contractile/metabolic dysfunction, and necroinflammation. Furthermore, we identify a potential pharmacological mechanism to prevent neonatal hypoxic injury.


Subject(s)
14-3-3 Proteins/metabolism , Heart Diseases/drug therapy , Membrane Proteins/metabolism , Misoprostol/therapeutic use , Mitochondrial Proteins/metabolism , Oxytocics/therapeutic use , Animals , Disease Models, Animal , Humans , Misoprostol/pharmacology , Oxytocics/pharmacology , Rats , Transfection
2.
Wound Repair Regen ; 29(4): 667-677, 2021 07.
Article in English | MEDLINE | ID: mdl-34076932

ABSTRACT

Heart disease with attendant cardiac fibrosis kills more patients in developed countries than any other disease, including cancer. We highlight the recent literature on factors that activate and also deactivate cardiac fibroblasts. Activation of cardiac fibroblasts results in myofibroblasts phenotype which incorporates aSMA to stress fibres, express ED-A fibronectin, elevated PDGFRα and are hypersecretory ECM components. These cells facilitate both acute wound healing (infarct site) and chronic cardiac fibrosis. Quiescent fibroblasts are associated with normal myocardial tissue and provide relatively slow turnover of the ECM. Deactivation of activated myofibroblasts is a much less studied phenomenon. In this context, SKI is a known negative regulator of TGFb1 /Smad signalling, and thus may share functional similarity to PPARγ activation. The discovery of SKI's potent anti-fibrotic role, and its ability to deactivate and/or myofibroblasts is featured and contrasted with PPARγ. While myofibroblasts are typically recruited from pools of potential precursor cells in a variety of organs, the importance of activation of resident cardiac fibroblasts has been recently emphasised. Myofibroblasts deposit ECM components at an elevated rate and contribute to both systolic and diastolic dysfunction with attendant cardiac fibrosis. A major knowledge gap exists as to specific proteins that may signal for fibroblast deactivation. As SKI may be a functionally pluripotent protein, we suggest that it serves as a scaffold to proteins other than R-Smads and associated Smad signal proteins, and thus its anti-fibrotic effects may extend beyond binding R-Smads. While cardiac fibrosis is causal to heart failure, the treatment of cardiac fibrosis is hampered by the lack of availability of effective pharmacological anti-fibrotic agents. The current review will provide an overview of work highlighting novel factors which cause fibroblast activation and deactivation to underscore putative therapeutic avenues for improving disease outcomes in cardiac patients with fibrosed hearts.


Subject(s)
Antifibrotic Agents , Wound Healing , Fibroblasts/pathology , Fibrosis , Humans , Myocardium/pathology , Myofibroblasts/pathology
3.
Methods Mol Biol ; 2299: 171-179, 2021.
Article in English | MEDLINE | ID: mdl-34028743

ABSTRACT

Two-dimensional cell culture is the primary method employed for proof-of-concept studies in most molecular biology labs. While immortalized cell lines are convenient and easy to maintain for extended periods in vitro, their inability to accurately represent genuine cell physiology-or pathophysiology-presents a challenge for drug discovery, as most results are not viable for the transition to clinical trial. The use of primary cells is a more biologically relevant approach to this issue; however, simulating in vitro what is observed in vivo is exigent at best. Primary cardiac fibroblasts are particularly difficult to maintain in a quiescent state, due to their innate phenotypic plasticity, and sensitivity to mechanical and biochemical stimulus. As conventional cell culture methods do not consider these factors, here we describe a method that limits environmental input (i.e., mechanical, nutritional, hormonal) to extend the physiological cardiac fibroblast phenotype in vitro.


Subject(s)
Myocytes, Cardiac/cytology , Myofibroblasts/cytology , Primary Cell Culture/methods , Actins/metabolism , Animals , Cell Differentiation , Cells, Cultured , Humans , Mechanical Phenomena , Mice , Models, Biological , Myocytes, Cardiac/metabolism , Myofibroblasts/metabolism , Phenotype
4.
Basic Res Cardiol ; 116(1): 25, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33847835

ABSTRACT

We have previously shown that overexpression of SKI, an endogenous TGF-ß1 repressor, deactivates the pro-fibrotic myofibroblast phenotype in the heart. We now show that SKI also functions independently of SMAD/TGF-ß signaling, by activating the Hippo tumor-suppressor pathway and inhibiting the Transcriptional co-Activator with PDZ-binding motif (TAZ or WWTR1). The mechanism(s) by which SKI targets TAZ to inhibit cardiac fibroblast activation and fibrogenesis remain undefined. A rat model of post-myocardial infarction was used to examine the expression of TAZ during acute fibrogenesis and chronic heart failure. Results were then corroborated with primary rat cardiac fibroblast cell culture performed both on plastic and on inert elastic substrates, along with the use of siRNA and adenoviral expression vectors for active forms of SKI, YAP, and TAZ. Gene expression was examined by qPCR and luciferase assays, while protein expression was examined by immunoblotting and fluorescence microscopy. Cell phenotype was further assessed by functional assays. Finally, to elucidate SKI's effects on Hippo signaling, the SKI and TAZ interactomes were captured in human cardiac fibroblasts using BioID2 and mass spectrometry. Potential interactors were investigated in vitro to reveal novel mechanisms of action for SKI. In vitro assays on elastic substrates revealed the ability of TAZ to overcome environmental stimuli and induce the activation of hypersynthetic cardiac myofibroblasts. Further cell-based assays demonstrated that SKI causes specific proteasomal degradation of TAZ, but not YAP, and shifts actin cytoskeleton dynamics to inhibit myofibroblast activation. These findings were supported by identifying the bi-phasic expression of TAZ in vivo during post-MI remodeling and fibrosis. BioID2-based interactomics in human cardiac fibroblasts suggest that SKI interacts with actin-modifying proteins and with LIM Domain-containing protein 1 (LIMD1), a negative regulator of Hippo signaling. Furthermore, we found that LATS2 interacts with TAZ, whereas LATS1 does not, and that LATS2 knockdown prevented TAZ downregulation with SKI overexpression. Our findings indicate that SKI's capacity to regulate cardiac fibroblast activation is mediated, in part, by Hippo signaling. We postulate that the interaction between SKI and TAZ in cardiac fibroblasts is arbitrated by LIMD1, an important intermediary in focal adhesion-associated signaling pathways. This study contributes to the understanding of the unique physiology of cardiac fibroblasts, and of the relationship between SKI expression and cell phenotype.


Subject(s)
Fibroblasts/metabolism , Heart Failure/metabolism , Hippo Signaling Pathway , Intracellular Signaling Peptides and Proteins/metabolism , LIM Domain Proteins/metabolism , Myocardial Infarction/metabolism , Myocardium/metabolism , Proto-Oncogene Proteins/metabolism , Ventricular Remodeling , Animals , Cells, Cultured , Disease Models, Animal , Fibroblasts/pathology , Fibrosis , Heart Failure/genetics , Heart Failure/pathology , Heart Failure/physiopathology , Intracellular Signaling Peptides and Proteins/genetics , LIM Domain Proteins/genetics , Male , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phenotype , Proto-Oncogene Proteins/genetics , Rats , Rats, Sprague-Dawley , Transcriptional Coactivator with PDZ-Binding Motif Proteins/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins/metabolism
6.
Sci Rep ; 9(1): 12889, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31501457

ABSTRACT

Primary cardiac fibroblasts are notoriously difficult to maintain for extended periods of time in cell culture, due to the plasticity of their phenotype and sensitivity to mechanical input. In order to study cardiac fibroblast activation in vitro, we have developed cell culture conditions which promote the quiescent fibroblast phenotype in primary cells. Using elastic silicone substrata, both rat and mouse primary cardiac fibroblasts could be maintained in a quiescent state for more than 3 days after isolation and these cells showed low expression of myofibroblast markers, including fibronectin extracellular domain A, non-muscle myosin IIB, platelet-derived growth factor receptor-alpha and alpha-smooth muscle actin. Gene expression was also more fibroblast-like vs. that of myofibroblasts, as Tcf21 was significantly upregulated, while Fn1-EDA, Col1A1 and Col1A2 were markedly downregulated. Cell culture conditions (eg. serum, nutrient concentration) are critical for the control of temporal fibroblast proliferation. We propose that eliminating mechanical stimulus and limiting the nutrient content of cell culture media can extend the quiescent nature of primary cardiac fibroblasts for physiological analyses in vitro.


Subject(s)
Cell Culture Techniques/methods , Myofibroblasts/cytology , Animals , Cell Proliferation , Male , Rats
7.
Adv Exp Med Biol ; 1132: 35-41, 2019.
Article in English | MEDLINE | ID: mdl-31037622

ABSTRACT

Cardiac muscle (the myocardium) is a unique arrangement of atria and ventricles that are spatially and electrically separated by a fibrous border. The spirally-arranged myocytes in both left and right ventricles are tethered by the component molecules of the cardiac extracellular matrix (ECM), including fibrillar collagen types I and III. Loss of normal arrangement of the ECM with either too little (as is observed in acute myocardial infarction) or too much (cardiac fibrosis in chronic post-myocardial infarction) is the primary contributor to cardiac dysfunction and heart failure. Matricellular proteins exist as non-structural signaling moieties in the ECM, and in the context of cardiac hypertrophy and heart failure, secreted 90 kDa periostin protein has attracted intense scrutiny during the past decade. Secreted periostin is now recognized for its important role in ECM development and maturation, as well as cellular adhesion. The novel mechanisms of periostin function include its role as a mediator of cell-to-matrix signaling, cell survival, and epithelial-mesenchymal transition (EMT). A number of recent studies have examined the hypothesis that periostin is a major contributor to ECM remodeling in the heart, and a number of very recent studies underscore its important role. This review examines recent developments in the mechanisms of periostin function in the normal heart and vasculature, and discusses recent advances which underpin its putative role in the development of cardiovascular disease. Periostin expression is very low at baseline in healthy tissues, but is re-expressed in damaged heart and in vessel walls after injury, in activated cardiac myofibroblasts and vascular smooth muscle cells, respectively. For this reason, periostin may be exploited for investigation of mechanisms of cardiac fibrosis , and we speculate that data generated from studies utilizing this approach may shed light on the timing for application of periostin-specific therapies to quell cardiac fibrosis and associated cardiac dysfunction.


Subject(s)
Cell Adhesion Molecules/physiology , Myocardial Infarction , Myofibroblasts/cytology , Ventricular Remodeling , Extracellular Matrix Proteins , Heart Ventricles , Humans , Myocardium , Phenotype
8.
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
9.
Int J Mol Sci ; 19(10)2018 Oct 17.
Article in English | MEDLINE | ID: mdl-30336567

ABSTRACT

Following cardiac injury, fibroblasts are activated and are termed as myofibroblasts, and these cells are key players in extracellular matrix (ECM) remodeling and fibrosis, itself a primary contributor to heart failure. Nutraceuticals have been shown to blunt cardiac fibrosis in both in-vitro and in-vivo studies. However, nutraceuticals have had conflicting results in clinical trials, and there are no effective therapies currently available to specifically target cardiac fibrosis. We have previously shown that expression of the zinc finger E box-binding homeobox 2 (Zeb2) transcription factor increases as fibroblasts are activated. We now show that Zeb2 plays a critical role in fibroblast activation. Zeb2 overexpression in primary rat cardiac fibroblasts is associated with significantly increased expression of embryonic smooth muscle myosin heavy chain (SMemb), ED-A fibronectin and α-smooth muscle actin (α-SMA). We found that Zeb2 was highly expressed in activated myofibroblast nuclei but not in the nuclei of inactive fibroblasts. Moreover, ectopic Zeb2 expression in myofibroblasts resulted in a significantly less migratory phenotype with elevated contractility, which are characteristics of mature myofibroblasts. Knockdown of Zeb2 with siRNA in primary myofibroblasts did not alter the expression of myofibroblast markers, which may indicate that Zeb2 is functionally redundant with other profibrotic transcription factors. These findings add to our understanding of the contribution of Zeb2 to the mechanisms controlling cardiac fibroblast activation.


Subject(s)
Fibroblasts/metabolism , Myocardium/cytology , Zinc Finger E-box Binding Homeobox 2/metabolism , Animals , Biomarkers/metabolism , Cell Movement , Cell Nucleus/metabolism , Gene Knockdown Techniques , Male , Myofibroblasts/metabolism , Phenotype , Protein Transport , RNA, Small Interfering/metabolism , Rats, Sprague-Dawley
10.
Cell Death Differ ; 25(10): 1732-1748, 2018 11.
Article in English | MEDLINE | ID: mdl-29511336

ABSTRACT

Myocardin is a transcriptional co-activator required for cardiovascular development, but also promotes cardiomyocyte survival through an unclear molecular mechanism. Mitochondrial permeability transition is implicated in necrosis, while pore closure is required for mitochondrial maturation during cardiac development. We show that loss of myocardin function leads to subendocardial necrosis at E9.5, concurrent with elevated expression of the death gene Nix. Mechanistically, we demonstrate that myocardin knockdown reduces microRNA-133a levels to allow Nix accumulation, leading to mitochondrial permeability transition, reduced mitochondrial respiration, and necrosis. Myocardin knockdown elicits calcium release from the endo/sarcoplasmic reticulum with mitochondrial calcium accumulation, while restoration of microRNA-133a function, or knockdown of Nix rescues calcium perturbations. We observed reduced myocardin and elevated Nix expression within the infarct border-zone following coronary ligation. These findings identify a myocardin-regulated pathway that maintains calcium homeostasis and mitochondrial function during development, and is attenuated during ischemic heart disease. Given the diverse role of Nix and microRNA-133a, these findings may have broader implications to metabolic disease and cancer.


Subject(s)
Calcium/metabolism , Mitochondria/metabolism , Nuclear Proteins/metabolism , Trans-Activators/metabolism , Animals , Cells, Cultured , Doxorubicin/pharmacology , Gene Expression/drug effects , Heart/drug effects , Isoproterenol/pharmacology , Membrane Potential, Mitochondrial/drug effects , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , MicroRNAs/metabolism , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Permeability/drug effects , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Sarcoplasmic Reticulum/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/genetics
11.
Oncotarget ; 7(48): 78516-78531, 2016 Nov 29.
Article in English | MEDLINE | ID: mdl-27705938

ABSTRACT

The incidence of heart failure with concomitant cardiac fibrosis is very high in developed countries. Fibroblast activation in heart is causal to cardiac fibrosis as they convert to hypersynthetic cardiac myofibroblasts. There is no known treatment for cardiac fibrosis. Myofibroblasts contribute to the inappropriate remodeling of the myocardial interstitium, which leads to reduced cardiac function and ultimately heart failure. Elevated levels of autophagy have been linked to stress-induced ventricular remodeling and other cardiac diseases. Previously, we had shown that TGF-ß1 treatment of human atrial fibroblasts both induced autophagy and enhanced the fibrogenic response supporting a linkage between the myofibroblast phenotype and autophagy. We now demonstrate that with in vitro culture of primary rat cardiac fibroblasts, inhibition of autophagy represses fibroblast to myofibroblast phenoconversion. Culturing unpassaged cardiac fibroblasts for 72 hours on plastic tissue culture plates is associated with elevated α-smooth muscle actin (α-SMA) expression. This activation parallels increased microtubule-associated protein 1A/1B-light chain 3 (LC-3ß II) protein expression. Inhibition of autophagy with bafilomycin-A1 (Baf-A1) and chloroquine (CQ) in cardiac fibroblasts significantly reduces α-SMA and extracellular domain A fibronectin (ED-A FN) protein vs untreated controls. Myofibroblast cell migration and contractility were significantly reduced following inhibition of autophagy. These data support the possibility of a causal link between cardiac fibroblast-to-myofibroblast phenoconversion and autophagy.


Subject(s)
Autophagy/drug effects , Cardiomyopathies/prevention & control , Chloroquine/pharmacology , Fibroblasts/drug effects , Macrolides/pharmacology , Myocardium/pathology , Myofibroblasts/drug effects , Actins/metabolism , Animals , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Movement/drug effects , Cells, Cultured , Fibroblasts/metabolism , Fibroblasts/pathology , Fibronectins/metabolism , Fibrosis , Male , Microtubule-Associated Proteins/metabolism , Myocardium/metabolism , Myofibroblasts/metabolism , Myofibroblasts/pathology , Phenotype , Phosphorylation , Primary Cell Culture , Rats, Sprague-Dawley , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
12.
Biochim Biophys Acta ; 1863(6 Pt A): 1261-8, 2016 Jun.
Article in English | MEDLINE | ID: mdl-27039037

ABSTRACT

Inappropriate cardiac interstitial remodeling is mediated by activated phenoconverted myofibroblasts. The synthesis of matrix proteins by these cells is triggered by both chemical and mechanical stimuli. Ski is a repressor of TGFß1/Smad signaling and has been described as possessing anti-fibrotic properties within the myocardium. We hypothesized that overexpression of Ski in myofibroblasts will induce an apoptotic response, which may either be supported or opposed by autophagic flux. We used primary myofibroblasts (activated fibroblasts) which were sourced from whole heart preparations that were only passaged once. We found that overexpression of Ski results in distinct morphological and biochemical changes within primary cardiac myofibroblasts associated with apoptosis. Ski treatment was associated with the expression of pro-apoptotic factors such as Bax, caspase-7, and -9. Our results indicate that Ski triggers a pro-death mechanism in primary rat cardiac myofibroblasts that is mediated through the intrinsic apoptotic pathway. Myofibroblast survival is prolonged by an autophagic response, as the dataset indicate that apoptosis is hastened when autophagy is inhibited. We suggest that the apoptotic death response of myofibroblasts is working in parallel with the previously observed anti-fibrotic properties of Ski within this cell type. As myofibroblasts are the sole mediators of matrix expansion in heart failure, we suggest that Ski, or a putative Ski-mimetic, may induce graded apoptosis in myofibroblasts within the failing heart and may be a novel therapeutic approach towards controlling cardiac fibrosis. Future studies are needed to examine the potential effects of Ski overexpression on other cell types in the heart.


Subject(s)
Apoptosis/physiology , Autophagy/physiology , Myofibroblasts/metabolism , Proto-Oncogene Proteins/metabolism , Actins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Autophagy/drug effects , Autophagy/genetics , Blotting, Western , Caspases/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Enzyme Inhibitors/pharmacology , Gene Expression , Macrolides/pharmacology , Male , Microscopy, Confocal , Myofibroblasts/cytology , Myofibroblasts/drug effects , Proto-Oncogene Proteins/genetics , Rats, Sprague-Dawley , Staurosporine/pharmacology , Time Factors , Transfection , Vimentin/metabolism
13.
Biochim Biophys Acta ; 1823(12): 2274-86, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23026405

ABSTRACT

Trans fats are not a homogeneous group of molecules and less is known about the cellular effects of individual members of the group. Vaccenic acid (VA) and elaidic acid (EA) are the predominant trans monoenes in ruminant fats and vegetable oil, respectively. Here, we investigated the mechanism of cell death induced by VA and EA on primary rat ventricular myofibroblasts (rVF). The MTT assay demonstrated that both VA and EA (200µM, 0-72 h) reduced cell viability in rVF (P<0.001). The FACS assay confirmed that both VA and EA induced apoptosis in rVF, and this was concomitant with elevation in cleaved caspase-9, -3 and -7, but not caspase-8. VA and EA decreased the expression ratio of Bcl2:Bax, induced Bax translocation to mitochondria and decrease in mitochondrial membrane potential (Δψ). BAX and BAX/BAK silencing in mouse embryonic fibroblasts (MEF) inhibited VA and EA-induced cell death compared to the corresponding wild type cells. Transmission electron microscopy revealed that VA and EA also induced macroautophagosome formation in rVF, and immunoblot analysis confirmed the induction of several autophagy markers: LC3-ß lipidation, Atg5-12 accumulation, and increased beclin-1. Finally, deletion of autophagy genes, ATG3 and ATG5 significantly inhibited VA and EA-induced cell death (P<0.001). Our findings show for the first time that trans fat acid (TFA) induces simultaneous apoptosis and autophagy in rVF. Furthermore, TFA-induced autophagy is required for this pro-apoptotic effect. Further studies to address the effect of TFA on the heart may reveal significant translational value for prevention of TFA-linked heart disease.


Subject(s)
Apoptosis/drug effects , Autophagy , Myofibroblasts/drug effects , Myofibroblasts/pathology , Trans Fatty Acids/pharmacology , Animals , Blotting, Western , Cells, Cultured , Flow Cytometry , Heart Ventricles/drug effects , Heart Ventricles/pathology , Male , Membrane Potential, Mitochondrial/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley
14.
Am J Physiol Heart Circ Physiol ; 301(2): H514-22, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21572008

ABSTRACT

Chemotactic movement of myofibroblasts is recognized as a common means for their sequestration to the site of tissue injury. Following myocardial infarction (MI), recruitment of cardiac myofibroblasts to the infarct scar is a critical step in wound healing. Contractile myofibroblasts express embryonic smooth muscle myosin, α-smooth muscle actin, as well as collagens I and III. We examined the effects of cardiotrophin-1 (CT-1) in the induction of primary rat ventricular myofibroblast motility. Changes in membrane potential (E(m)) and Ca(2+) entry were studied to reveal the mechanisms for induction of myofibroblast migration. CT-1-induced cardiac myofibroblast cell migration, which was attenuated through the inhibition of JAK2 (25 µM AG490), and myosin light chain kinase (20 µM ML-7). Inhibition of K(+) channels (1 mM tetraethylammonium or 100 µM 4-aminopyridine) and nonselective cation channels by 10 µM gadolinium (Gd(3+)) significantly reduced migration in the presence of CT-1. CT-1 treatment caused a significant increase in myosin light chain phosphorylation, which could be inhibited by incubation in Ca(2+)-free conditions or by application of AG490, ML-7, and W7 (100 µM; calmodulin inhibitor). Monitoring myofibroblast membrane potential with potentiometric fluorescent DiBAC(4)(3) dye revealed a biphasic response to CT-1 consisting of an initial depolarization followed by hyperpolarization. Increased intracellular Ca(2+), as assessed by fluo 3, occurred immediately after membrane depolarization and attenuated at the time of maximal hyperpolarization. CT-1 exerts chemotactic effects via multiple parallel signaling modalities in ventricular myofibroblasts, including changes in membrane potential, alterations in intracellular calcium, and activation of a number of intracellular signaling pathways. Further study is warranted to determine the precise role of K(+) currents in this process.


Subject(s)
Chemotaxis , Cytokines/metabolism , Myofibroblasts/enzymology , Myosin-Light-Chain Kinase/metabolism , Analysis of Variance , Animals , Calcium/metabolism , Calmodulin/antagonists & inhibitors , Calmodulin/metabolism , Cardiac Myosins/metabolism , Cells, Cultured , Chemotaxis/drug effects , Gadolinium/metabolism , Heart Ventricles/cytology , Heart Ventricles/enzymology , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Male , Membrane Potentials , Myofibroblasts/drug effects , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/antagonists & inhibitors , Phosphorylation , Potassium/metabolism , Potassium Channel Blockers/pharmacology , Potassium Channels/drug effects , Potassium Channels/metabolism , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Spectrometry, Fluorescence , Time Factors
15.
Am J Physiol Cell Physiol ; 300(1): C176-86, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20943957

ABSTRACT

Cardiac myofibroblasts are key players in chronic remodeling of the cardiac extracellular matrix, which is mediated in part by elevated transforming growth factor-ß1 (TGF-ß1). The c-Ski proto-oncoprotein has been shown to modify TGF-ß1 post-receptor signaling through receptor-activated Smads (R-Smads); however, little is known about how c-Ski regulates fibroblast phenotype and function. We sought to elucidate the function of c-Ski in primary cardiac myofibroblasts using a c-Ski overexpression system. Cardiac myofibroblasts expressed three forms of c-Ski with the predominant band at 105 kDa, and adenoviral c-Ski treatment resulted in overexpression of 95-kDa c-Ski in cellular nuclei. Exogenous c-Ski led to significant inhibition of type I collagen secretion and myofibroblast contractility using two-dimensional semifloating gel contraction assay in both basal and with TGF-ß1 (10 ng/ml for 24 h) stimulation. Overexpressed c-Ski did not inhibit nuclear translocation of phosphorylated R-Smad2, despite their binding, as demonstrated by immunoprecipitation. Acute treatment of primary myofibroblasts with TGF-ß1 in vitro revealed a marked nuclear shuttling of c-Ski at 24 and 48 h following stimulation. Remarkably, overexpression of c-Ski led to a stepwise reduction of the myofibroblast marker α-smooth muscle actin with increasing multiplicity of infection, and these results indicate that 95-kDa c-Ski overexpression may effect a loss of the myofibroblastic phenotype. Furthermore, adenovirus (Ad) for hemagglutinin-tagged c-Ski infection led to a reduction in the number of myofibroblasts versus Ad-LacZ-infected and uninfected controls, due to induction of apoptosis. Finally, we observed a significant increase in 105-kDa c-Ski in the cytosolic fraction of cells of the infarct scar and adjacent remnant myocardium vs. noninfarcted controls.


Subject(s)
Fibrosis/metabolism , Myocardial Contraction/physiology , Myocardium/cytology , Myofibroblasts/cytology , Proto-Oncogene Proteins/metabolism , Active Transport, Cell Nucleus , Animals , Apoptosis/physiology , Cell Differentiation , Cell Survival , Gene Expression Regulation/physiology , Myofibroblasts/classification , Proto-Oncogene Proteins/genetics , Rats , Smad2 Protein/genetics , Smad2 Protein/metabolism , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism
16.
J Nutr ; 140(8): 1438-44, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20554900

ABSTRACT

A high-lipid diet (HLD) may lead to adverse left ventricular (LV) remodeling and endothelial dysfunction in conditions of hemodynamic stress. Although congenital absence of nitric oxide synthase 3 (NOS3) leads to adverse LV remodeling after transverse aortic constriction (TAC), the effects of a HLD in this state remains unknown. Wild-type (WT) and NOS3 knockout mice (NOS3(-/-)) were randomized into the following 4 groups: 1) WT + low-lipid diet (LLD) (10% of energy); 2) WT + HLD (60% of energy); 3) NOS3(-/-) + LLD; and 4) NOS3(-/-) + HLD for a total of 12 wk. After 1 wk of randomization, TAC was performed on all groups. Serial echocardiography revealed a decrease in LV ejection fraction (LVEF) in WT and NOS3(-/-) mice fed the HLD compared with those fed the LLD diet at 12 wk post-TAC. Mice fed the NOS3(-/-) + HLD diet had a lower LVEF compared with mice in the other 3 groups (P < 0.05). There was greater myocyte hypertrophy, interstitial fibrosis, and percentage change in plasma cholesterol concentrations in the NOS3(-/-) + HLD group 12 wk post-TAC compared with the other 3 groups. Although high molecular weight fibroblast growth factor-2, a marker of cardiac hypertrophy, was more upregulated in the NOS3(-/-) + HLD group than in the other groups, markers of the renin-angiotensin system did not differ among them. A HLD potentiates LV dysfunction in NOS3(-/-) mice in a chronic pressure overload state.


Subject(s)
Dietary Fats/administration & dosage , Dietary Fats/adverse effects , Hypertension/complications , Nitric Oxide Synthase Type III/deficiency , Ventricular Dysfunction, Left/etiology , Animals , Aorta , Blood Pressure , Cholesterol/blood , Constriction , Echocardiography , Energy Intake , Fibroblast Growth Factor 2/analysis , Heart Ventricles/pathology , Hypertrophy, Left Ventricular/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Weight , Muscle Cells/pathology , Myocardium/pathology , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/physiology , Stroke Volume , Ventricular Dysfunction, Left/metabolism , Ventricular Dysfunction, Left/pathology
17.
Dev Dyn ; 239(6): 1573-84, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20503355

ABSTRACT

In fibrosing hearts, myofibroblasts are associated with cardiac extracellular matrix remodeling. Expression of key genes in the transition of cardiac fibroblast to myofibroblast phenotype in post-myocardial infarction heart and in vitro has not been well addressed. Contractile, focal adhesion-associated, receptor proteins, fibroblast growth factor-2 (FGF-2) expression, and motility were compared to assess phenotype in adult and neonatal rat cardiac fibroblasts and myofibroblasts. Neonatal and adult fibroblasts undergo phenotypic transition to myofibroblastic cells, marked by increased alpha-smooth muscle actin (alphaSMA), smooth muscle myosin heavy chain (SMemb), extra domain-A (ED-A) fibronectin, paxillin, tensin, FGF-2, and TbetaRII receptor. Elevated ED-A fibronectin confirmed fibroblast to supermature myofibroblastic phenotype transition. Presence of myofibroblasts in vivo was noted in sections of healed infarct scar after myocardial infarction, and their expression is similar to that in culture. Thus, cultured neonatal and adult cardiac fibroblasts transition to myofibroblasts in vitro and share expression profiles of cardiac myofibroblasts in vivo. Reduced motility with in vitro passage reflects enhanced production of focal adhesions.


Subject(s)
Fibroblasts/metabolism , Focal Adhesions/metabolism , Muscle, Smooth/metabolism , Animals , Animals, Newborn , Cell Differentiation/physiology , Cell Movement , Extracellular Matrix/metabolism , Fibroblast Growth Factor 2/metabolism , Fibroblasts/cytology , Fibronectins/metabolism , Fibrosis/metabolism , Fibrosis/pathology , Heart Ventricles/metabolism , Male , Myocardium/metabolism , Myocardium/pathology , Rats , Rats, Sprague-Dawley
18.
J Cell Physiol ; 213(2): 540-51, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17541957

ABSTRACT

Cardiac ventricular myofibroblast motility, proliferation, and contraction contribute to post-myocardial infarct wound healing, infarct scar formation, and remodeling of the ventricle remote to the site of infarction. The Na+-Ca2+ exchanger (NCX1) is involved in altered calcium handling in cardiac myocytes during cardiac remodeling associated with heart failure, however, its role in cardiac myofibroblast cell function is unexplored. In this study we investigated the involvement of NCX1 as well as the role of non-selective-cation channels (NSCC) in cardiac myofibroblast cell function in vitro. Immunofluorescence and Western blots revealed that P1 cells upregulate alpha-smooth muscle actin (alphaSMA) and embryonic smooth muscle myosin heavy chain (SMemb) expression. NCX1 mRNA and proteins as well as Ca(v)1.2a protein are also expressed in P1 myofibroblasts. Myofibroblast motility in the presence of 50 ng/ml PDGF-BB was blocked with AG1296. Myofibroblast motility, contraction, and proliferation were sensitive to KB-R7943, a specific NCX1 reverse-mode inhibitor. In contrast, only proliferation and contraction, but not motility were sensitive to nifedipine, while gadolinium (NSCC blocker) was only associated with decreased motility. ML-7 treatment was associated with inhibition of the chemotactic response and contraction. Thus cardiac myofibroblast chemotaxis, contraction, and proliferation were sensitive to different pharmacologic treatments suggesting that regulation of transplasmalemmal calcium movements may be important in growth factor receptor-mediated processes. NCX1 may represent an important moiety in suppression of myofibroblast functions.


Subject(s)
Cell Movement/physiology , Cell Proliferation , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Sodium-Calcium Exchanger/metabolism , Actins/metabolism , Animals , Anti-Arrhythmia Agents/metabolism , Azepines/metabolism , Becaplermin , Calcium Channels, L-Type/metabolism , Cells, Cultured , Enzyme Inhibitors/metabolism , Gadolinium/metabolism , Male , Myocytes, Cardiac/cytology , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/metabolism , Naphthalenes/metabolism , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , Rats , Rats, Sprague-Dawley , Receptor, Platelet-Derived Growth Factor beta/metabolism , Sodium-Calcium Exchanger/antagonists & inhibitors , Sodium-Calcium Exchanger/genetics , Thiourea/analogs & derivatives , Thiourea/metabolism
19.
Am J Physiol Heart Circ Physiol ; 293(2): H1282-90, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17513491

ABSTRACT

Transforming growth factor-beta(1) (TGF-beta(1)) signal and downstream Smads play an important role in tissue fibrosis and matrix remodeling in various etiologies of heart failure. Inhibitory Smad7 (I-Smad7) is an inducible regulatory Smad protein that antagonizes TGF-beta(1) signal mediated via direct abrogation of R-Smad phosphorylation. The effect of ectopic I-Smad7 on net collagen production was investigated using hydroxyproline assay. Adenovirus-mediated I-Smad7 gene (at 100 multiplicity of infection) transfer was associated with significant decrease of collagen synthesis in the presence and absence of TGF-beta(1) in primary rat cardiac myofibroblasts. In I-Smad7-infected cells, we also observed the ablation of TGF-beta(1)-induced R-Smad2 phosphorylation vs. LacZ controls. Overdriven I-Smad7 was associated with significantly increased expression of immunoreactive 65-kDa matrix metalloproteinase-2 (MMP-2) protein in culture medium of myofibroblast compared with LacZ-infected cells. Expression of the 72-kDa MMP-2 variant, e.g., the inactive form, was not altered by exogenous I-Smad7 transfection/overexpression. Furthermore, I-Smad7 overexpression was associated with a significant increase and decrease in expression of p27 and phospho-Rb protein, respectively, as well as reduced [(3)H]thymidine incorporation vs. Ad-LacZ-infected controls. We suggest that negative modulation of R-Smad phosphorylation by ectopic I-Smad7 may contribute to the downregulation of collagen in cardiac myofibroblasts and may suppress the proliferation of these cells. Thus treatments targeting the collagen deposition by overexpression of I-Smad7 may provide a new therapeutic strategy for cardiac fibrosis.


Subject(s)
Collagen/biosynthesis , Fibroblasts/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Signal Transduction , Smad Proteins, Inhibitory/metabolism , Adenoviridae/genetics , Animals , Cell Proliferation , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Enzyme Activation , Enzyme Induction , Genetic Vectors , Heart Ventricles/cytology , Heart Ventricles/metabolism , Male , Matrix Metalloproteinase 2/biosynthesis , Mitogen-Activated Protein Kinases/metabolism , Myocardium/cytology , Phosphorylation , Rats , Retinoblastoma Protein/metabolism , Smad Proteins, Inhibitory/genetics , Smad Proteins, Receptor-Regulated/metabolism , Smad2 Protein/metabolism , Tissue Inhibitor of Metalloproteinases/biosynthesis , Transfection , Transforming Growth Factor beta1/metabolism , Up-Regulation
20.
Am J Physiol Heart Circ Physiol ; 293(2): H1053-64, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17483238

ABSTRACT

Myofibroblasts respond to an array of signals from mitogens and cytokines during the course of wound healing following a myocardial infarction (MI), and these signals may coordinate ventricular myofibroblast proliferation. Furthermore, myofibroblasts are contractile and contribute to wound contraction by imparting mechanical tension on surrounding extracellular matrix. Although TGF-beta(1), CT-1, and PDGF-BB participate in various stages of post-MI wound healing, their combined net effect(s) on myofibroblast function is unknown. We investigated myofibroblast proliferation, expression of cell cycle proteins, and contractile function of cells treated with TGF-beta(1) and/or CT-1. We confirmed that TGF-beta(1) (10 ng/ml) suppresses proliferation of these cells, whereas CT-1 (10 ng/ml) and, for comparative purposes, PDGF-BB (1 ng/ml) treatments were associated with proliferation. Specific TGF-beta(1) treatment ablated CT-1-induced myofibroblast proliferation. TGF-beta(1) effects were specific, as they were suppressed by either TGF-beta-neutralizing antibody or viral Smad7 overexpression. TGF-beta(1) treatment also increased expression of p27 and decreased expression of cyclin E and Cdk2 in primary cells. CT-1 (10 ng/ml) treatment of myofibroblasts had no effect on collagen gel deformation versus controls, whereas TGF-beta(1) (10 ng/ml) and PDGF (10 ng/ml) treatments were associated with significant cell contraction; again, TGF-beta(1)-mediated contraction was unaffected by CT-1. Alone, CT-1 and TGF-beta(1) treatments exert opposing effects on myofibroblast function, whereas in combination TGF-beta(1)-mediated effects supersede those of CT-1 (and PDGF-BB). Thus TGF-beta(1) and CT-1 exert differential effects on myofibroblast proliferation and contraction in vitro, and we suggest that a balance of these effects may be important for the execution of normal cardiac wound healing.


Subject(s)
Cell Proliferation , Cytokines/metabolism , Fibroblasts/metabolism , Myocardial Contraction , Myocardial Infarction/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Becaplermin , Cell Proliferation/drug effects , Cells, Cultured , Collagen Type I/metabolism , Cyclin E/metabolism , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Cytokines/pharmacology , DNA Replication , Dose-Response Relationship, Drug , Drug Interactions , Fibroblasts/drug effects , Gels , Male , Myocardial Contraction/drug effects , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/cytology , Myocytes, Cardiac/drug effects , Phenotype , Platelet-Derived Growth Factor/metabolism , Proto-Oncogene Proteins c-sis , Rats , Rats, Sprague-Dawley , Smad7 Protein/genetics , Smad7 Protein/metabolism , Time Factors , Transfection , Transforming Growth Factor beta1/pharmacology , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...