1
|
Lim R, Banerjee A, Biswas R, Chari AN, Raghavan S. Mechanotransduction through adhesion molecules: Emerging roles in regulating the stem cell niche. Front Cell Dev Biol 2022; 10:966662. [PMID: 36172276 PMCID: PMC9511051 DOI: 10.3389/fcell.2022.966662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/18/2022] [Indexed: 11/23/2022] Open
Abstract
Stem cells have been shown to play an important role in regenerative medicine due to their proliferative and differentiation potential. The challenge, however, lies in regulating and controlling their potential for this purpose. Stem cells are regulated by growth factors as well as an array of biochemical and mechanical signals. While the role of biochemical signals and growth factors in regulating stem cell homeostasis is well explored, the role of mechanical signals has only just started to be investigated. Stem cells interact with their niche or to other stem cells via adhesion molecules that eventually transduce mechanical cues to maintain their homeostatic function. Here, we present a comprehensive review on our current understanding of the influence of the forces perceived by cell adhesion molecules on the regulation of stem cells. Additionally, we provide insights on how this deeper understanding of mechanobiology of stem cells has translated toward therapeutics.
Collapse
Affiliation(s)
- Ryan Lim
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Avinanda Banerjee
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Ritusree Biswas
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
- Sastra University, Thanjavur, TN, India
| | - Anana Nandakumar Chari
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
| | - Srikala Raghavan
- A∗STAR Skin Research Lab (ASRL), Agency for Science, Technology and Research (A*STAR) 8A Biomedical Grove, Singapore, Singapore
- Institute for Stem Cell Science and Regenerative Medicine (inStem), GKVK Campus, Bangalore, India
| |
Collapse
|
2
|
Tachampa K, Wongtawan T. Unique patterns of cardiogenic and fibrotic gene expression in rat cardiac fibroblasts. Vet World 2020; 13:1697-1708. [PMID: 33061247 PMCID: PMC7522959 DOI: 10.14202/vetworld.2020.1697-1708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 07/02/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND AND AIM Cardiac fibroblasts are important for both normal and pathological states of the heart, but the knowledge in cell physiology and genomics is still poorly understood. The aims of the present study were; first, to investigate the expression of cardiac and fibrotic genes in rat cardiac fibroblasts compared to cardiomyocytes and other fibroblasts (skin and muscle fibroblasts), second, to examine the in vitro effect of serum concentration on fibroblast gene expression. The findings can potentially be applied in ischemia/reperfusion models. MATERIALS AND METHODS Rat cardiac fibroblasts were collected and cultured in different conditions, and their gene expression (21 cardiogenic genes and 16 fibrotic genes) was compared with cardiomyocytes and other fibroblasts using comparative quantitative polymerase chain reaction. We also mimicked myocardial ischemia/reperfusion by depleting and then adding a serum into the culture in conventional culture (10% serum). RESULTS Cardiac fibroblasts expressed most of the cardiogenic genes, but their expression levels were significantly lower than in cardiomyocytes, while almost all fibrotic genes in the cardiac fibroblasts were significantly more highly expressed than in cardiomyocytes, except matrix metallopeptidase 9 (Mmp9) which also had greater expression in other fibroblasts. After mimicking cardiac ischemia and reperfusion in vitro by starving and then adding a serum into the cardiac fibroblast culture, the results revealed that Mmp9 expression was significantly increased (>30 times) after increasing but not reducing the serum in the culture. The expression of most cardiogenic and fibrotic genes in cardiac fibroblasts tended to decrease after increasing the serum in the culture. These changes were specific to cardiac fibroblasts but no other fibroblasts. CONCLUSION Cardiac fibroblasts have a distinct pattern of gene expression from other fibroblasts and cardiomyocytes. They are also sensitive to high serum concentration but not affected by serum depletion, suggesting that the process of developing cardiac fibrosis might be stimulated by reperfusion or overcirculation rather than ischemia. The cell starvation followed the adding of serum may serve as a useful model to study cardiac fibrosis cause by the change of blood flow.
Collapse
Affiliation(s)
- Kittipong Tachampa
- Department of Physiology, Faculty of Veterinary Science, Chulalongkorn University, Henri-Dunant Rd, Pathumwan, Bangkok, 10330, Thailand
| | - Tuempong Wongtawan
- Akkhraratchakumari Veterinary College, Walailak University, Tha Sala, Nakhon Si Thammarat, 80160, Thailand
- Laboratory of Cellular Biomedicine, Faculty of Veterinary Science, Mahidol University, Salaya, Puttamonthon, Nakhon Pathom, 73170, Thailand
| |
Collapse
|
3
|
Nielsen SH, Mouton AJ, DeLeon-Pennell KY, Genovese F, Karsdal M, Lindsey ML. Understanding cardiac extracellular matrix remodeling to develop biomarkers of myocardial infarction outcomes. Matrix Biol 2017; 75-76:43-57. [PMID: 29247693 DOI: 10.1016/j.matbio.2017.12.001] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Revised: 11/02/2017] [Accepted: 12/08/2017] [Indexed: 01/08/2023]
Abstract
Cardiovascular Disease (CVD) is the most common cause of death in industrialized countries, and myocardial infarction (MI) is a major CVD with significant morbidity and mortality. Following MI, the left ventricle (LV) undergoes a wound healing response to ischemia that results in extracellular matrix (ECM) scar formation to replace necrotic myocytes. While ECM accumulation following MI is termed cardiac fibrosis, this is a generic term that does not differentiate between ECM accumulation that occurs in the infarct region to form a scar that is structurally necessary to preserve left ventricle (LV) wall integrity and ECM accumulation that increases LV wall stiffness to exacerbate dilation and stimulate the progression to heart failure. This review focuses on post-MI LV ECM remodeling, targeting the discussion on ECM biomarkers that could be useful for predicting MI outcomes.
Collapse
Affiliation(s)
- Signe Holm Nielsen
- Fibrosis Biology and Biomarkers, Nordic Bioscience, Herlev, Denmark; Disease Systems Immunology, Technical University of Denmark, Kgs. Lyngby, Denmark
| | - Alan J Mouton
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA
| | - Kristine Y DeLeon-Pennell
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA; Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA
| | | | - Morten Karsdal
- Fibrosis Biology and Biomarkers, Nordic Bioscience, Herlev, Denmark
| | - Merry L Lindsey
- Mississippi Center for Heart Research, Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS, USA; Research Service, G.V. (Sonny) Montgomery Veterans Affairs Medical Center, Jackson, MS, USA.
| |
Collapse
|
4
|
Bildyug N. Matrix metalloproteinases: an emerging role in regulation of actin microfilament system. Biomol Concepts 2017; 7:321-329. [PMID: 27763882 DOI: 10.1515/bmc-2016-0022] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2016] [Accepted: 09/20/2016] [Indexed: 12/13/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are implicated in many physiological and pathological processes, including contraction, migration, differentiation, and proliferation. These processes all involve cell phenotype changes, known to be accompanied by reorganization of actin cytoskeleton. Growing evidence indicates a correlation between MMP activity and the dynamics of actin system, suggesting their mutual regulation. Here, data on the influence of MMPs on the actin microfilament system, on the one hand, and the dependence of MMP expression and activation on the organization of actin structures, on the other hand, are reviewed. The different mechanisms of putative actin-MMP regulation are discussed.
Collapse
|
5
|
Bildyug NB, Voronkina IV, Smagina LV, Yudintseva NM, Pinaev GP. Matrix Metalloproteinases in Primary Culture of Cardiomyocytes. BIOCHEMISTRY (MOSCOW) 2016; 80:1318-26. [PMID: 26567576 DOI: 10.1134/s0006297915100132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The highly organized contractile apparatus of cardiomyocytes in heart tissue allows for their continuous contractility, whereas extracellular matrix components are synthesized and spatially organized by fibroblasts and endothelial cells. However, reorganization of the cardiomyocyte contractile apparatus occurs upon their 2D cultivation, which is accompanied by transient loss of their contractility and acquired capability of extracellular matrix synthesis (Bildyug, N. B., and Pinaev, G. P. (2013) Tsitologiya, 55, 713-724). In this study, matrix metalloproteinases were investigated at different times of cardiomyocyte 2D cultivation and 3D cultivation in collagen gels. It was found that cardiomyocytes in 2D culture synthesize matrix metalloproteinases MMP-2 and MMP-9, wherein their amount varies with the cultivation time. The peak MMP-9 amount is at early cultivation time, when the reorganization of cardiomyocyte contractile apparatus occurs, and the MMP-2 peak precedes the recovery of the initial organization of their contractile apparatus. Upon cardiomyocyte cultivation in 3D collagen gels, in which case their contractile apparatus does not rearrange, a steady small amount of MMP-2 and MMP-9 is observed. These data indicate that the cardiomyocyte contractile apparatus reorganization in culture is associated with synthesis and spatial organization of their own extracellular matrix.
Collapse
Affiliation(s)
- N B Bildyug
- Institute of Cytology, Russian Academy of Sciences, Russia.
| | | | | | | | | |
Collapse
|
6
|
Mesenchymal stem cell-derived inflammatory fibroblasts mediate interstitial fibrosis in the aging heart. J Mol Cell Cardiol 2015; 91:28-34. [PMID: 26718722 DOI: 10.1016/j.yjmcc.2015.12.017] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/08/2015] [Accepted: 12/20/2015] [Indexed: 12/24/2022]
Abstract
Pathologic fibrosis in the aging mouse heart is associated with dysregulated resident mesenchymal stem cells (MSC) arising from reduced stemness and aberrant differentiation into dysfunctional inflammatory fibroblasts. Fibroblasts derived from aging MSC secrete higher levels of 1) collagen type 1 (Col1) that directly contributes to fibrosis, 2) monocyte chemoattractant protein-1 (MCP-1) that attracts leukocytes from the blood and 3) interleukin-6 (IL-6) that facilitates transition of monocytes into myeloid fibroblasts. The transcriptional activation of these proteins is controlled via the farnesyltransferase (FTase)-Ras-Erk pathway. The intrinsic change in the MSC phenotype acquired by advanced age is specific for the heart since MSC originating from bone wall (BW-MSC) or fibroblasts derived from them were free of these defects. The potential therapeutic interventions other than clinically approved strategies based on findings presented in this review are discussed as well. This article is a part of a Special Issue entitled "Fibrosis and Myocardial Remodeling".
Collapse
|
7
|
Tyagi SC. Dynamic role of extracellular matrix metalloproteinases in heart failure. Cardiovasc Pathol 2015; 7:153-9. [PMID: 25851222 DOI: 10.1016/s1054-8807(97)00121-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/1997] [Indexed: 10/17/2022] Open
Abstract
In chronic congestive heart failure, an illness affecting more than 4 million Americans, there is extensive myocardial extracellular matrix (ECM) remodeling. Failing human ventricular myocardium contains activated matrix metalloproteinases (MMPs) which are involved in adverse ECM remodeling. Our studies support the concept that impaired ECM remodeling and MMP activation are, in part, responsible for the cardiac structural deformation during heart failure. There is no known program which has declared its aim the investigation of regulation of fibrosis in hypertrophy and disruption of ECM in cardiac dilatation and failure. The development of transgenic technology, and emerging techniques for in vivo gene transfer, suggest a strategy for improving cardiac function by overexpressing or down regulation of the ECM components such as MMPs, tissue inhibitor of metalloproteinases (TIMPs), transforming growth factor β1 (TGFβ), decorin, collagen, and integrins in heart failure.
Collapse
Affiliation(s)
- S C Tyagi
- Department of Physiology and Biophysics, and Center of Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi USA
| |
Collapse
|
8
|
Roche PL, Filomeno KL, Bagchi RA, Czubryt MP. Intracellular Signaling of Cardiac Fibroblasts. Compr Physiol 2015; 5:721-60. [DOI: 10.1002/cphy.c140044] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
9
|
Activation of intracellular matrix metalloproteinase-2 by reactive oxygen–nitrogen species: Consequences and therapeutic strategies in the heart. Arch Biochem Biophys 2013; 540:82-93. [DOI: 10.1016/j.abb.2013.09.019] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 09/20/2013] [Accepted: 09/30/2013] [Indexed: 12/27/2022]
|
10
|
Kinoshita T, Ishikawa Y, Arita M, Akishima-Fukasawa Y, Fujita K, Inomata N, Suzuki T, Namiki A, Mikami T, Ikeda T, Yamazaki J, Ishii T, Akasaka Y. Antifibrotic response of cardiac fibroblasts in hypertensive hearts through enhanced TIMP-1 expression by basic fibroblast growth factor. Cardiovasc Pathol 2013; 23:92-100. [PMID: 24322055 DOI: 10.1016/j.carpath.2013.11.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 10/21/2013] [Accepted: 11/04/2013] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Cardiac fibroblasts (CFs) play a pivotal role in the development of myocardial fibrosis. We previously demonstrated that direct injection of basic fibroblast growth factor (bFGF) into the hypertensive Dahl salt-sensitive (DS) rat heart prevented systolic dysfunction and left ventricular dilation effectively. However, the precise role played by bFGF in fibrotic response of CFs remains unclear. We suggested potential effects of bFGF on the fibrotic response of CFs in vitro. METHODS AND RESULTS Histopathologic assessment of cardiac fibrosis demonstrated a marked decline in the extent of perivascular and interstitial fibrosis in bFGF-injected hypertensive DS rat hearts. CFs harvested from the hearts of noninjected DS rats demonstrated a significantly increased messenger RNA (mRNA) expression of matrix metalloproteinase (MMP)-2, MMP-9, and both collagen I and III. In contrast, bFGF treatment in the CFs induced a marked increase in tissue inhibitor of MMP (TIMP)-1 expression and a marked decline in MMP-9 activation. bFGF also induced a decline in α-smooth muscle actin and collagen I and III mRNA expression in the CFs accompanied by inhibited differentiation of CFs into myofibroblasts. Small interfering RNA targeting FGF receptor 1 confirmed a specific interference of the mRNA expression changes elicited by bFGF. In vivo examination confirmed many TIMP-1-positive CFs in perivascular spaces of bFGF-injected hearts. CONCLUSIONS Up-regulated TIMP-1 expression and down-regulated MMP-9 activation by bFGF in CFs could prevent excessive ECM degradation and collagen deposition in perivascular spaces effectively, leading to prevention of cardiac fibrosis during hypertensive heart failure. SUMMARY Cardiac fibroblasts (CFs) play a pivotal role in myocardial fibrosis. The precise role of CFs in fibrotic response played by growth factors remains unclear. Our results indicates that basic fibroblast growth factor could up-regulate TIMP-1 expression and down-regulate MMP-9 activation in CFs in perivascular spaces, leading to inhibited progression of cardiac fibrosis during hypertensive heart failure.
Collapse
Affiliation(s)
- Toshio Kinoshita
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine, 6-11-1 Omori-Nishi, Ohta-City, Tokyo 143-8541, Japan
| | - Yukio Ishikawa
- Department of Pathology, School of Medicine, Toho University, 5-21-16 Omori-Nishi, Ohta-City, Tokyo 143-8540, Japan
| | - Michitsune Arita
- Department of Molecular Immunology, School of Medicine, Toho University, 5-21-16 Omori-Nishi, Ohta-City, Tokyo 143-8540, Japan
| | - Yuri Akishima-Fukasawa
- Department of Pathology, School of Medicine, Toho University, 5-21-16 Omori-Nishi, Ohta-City, Tokyo 143-8540, Japan
| | - Kazuko Fujita
- Department of Pathology, School of Medicine, Toho University, 5-21-16 Omori-Nishi, Ohta-City, Tokyo 143-8540, Japan
| | - Naomi Inomata
- Department of Plastic and Reconstructive Surgery, School of Medicine, Toho University, 5-21-16 Omori-Nishi, Ohta-City, Tokyo 143-8540, Japan
| | - Takeya Suzuki
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine, 6-11-1 Omori-Nishi, Ohta-City, Tokyo 143-8541, Japan
| | - Atsushi Namiki
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine, 6-11-1 Omori-Nishi, Ohta-City, Tokyo 143-8541, Japan
| | - Tetuo Mikami
- Department of Pathology, School of Medicine, Toho University, 5-21-16 Omori-Nishi, Ohta-City, Tokyo 143-8540, Japan
| | - Takanori Ikeda
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine, 6-11-1 Omori-Nishi, Ohta-City, Tokyo 143-8541, Japan
| | - Junichi Yamazaki
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine, 6-11-1 Omori-Nishi, Ohta-City, Tokyo 143-8541, Japan
| | - Toshiharu Ishii
- Department of Pathology, School of Medicine, Toho University, 5-21-16 Omori-Nishi, Ohta-City, Tokyo 143-8540, Japan
| | - Yoshikiyo Akasaka
- Division of Chronic Inflammatory Diseases, Advanced Medical Research Center, Toho University Graduate School of Medicine, 5-21-16 Omori-Nishi, Ohta-City, Tokyo 143-8540, Japan.
| |
Collapse
|
11
|
Lindsey ML, Zamilpa R. Temporal and spatial expression of matrix metalloproteinases and tissue inhibitors of metalloproteinases following myocardial infarction. Cardiovasc Ther 2010; 30:31-41. [PMID: 20645986 DOI: 10.1111/j.1755-5922.2010.00207.x] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Following a myocardial infarction (MI), the homeostatic balance between matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) is disrupted as part of the left ventricle (LV) response to injury. The full complement of responses to MI has been termed LV remodeling and includes changes in LV size, shape and function. The following events encompass the LV response to MI: (1) inflammation and LV wall thinning and dilation, (2) infarct expansion and necrotic myocyte resorption, (3) accumulation of fibroblasts and scar formation, and (4) endothelial cell activation and neovascularization. In this review, we will summarize MMP and TIMP roles during these events, focusing on the spatiotemporal localization and MMP and TIMP effects on cellular and tissue-level responses. We will review MMP and TIMP structure and function, and discuss specific MMP roles during both the acute and chronic phases post-MI, which may provide insight into novel therapeutic targets to limit adverse remodeling in the MI setting.
Collapse
Affiliation(s)
- Merry L Lindsey
- Division of Cardiology, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX 78229-3900, USA
| | | |
Collapse
|
12
|
Rizzo A, Minoia G, Trisolini C, Mutinati M, Spedicato M, Jirillo F, Sciorsci RL. Reactive oxygen species (ROS): involvement in bovine follicular cysts etiopathogenesis. Immunopharmacol Immunotoxicol 2010; 31:631-5. [PMID: 19874233 DOI: 10.3109/08923970902932962] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Ovulation is compared to an acute inflammatory process during which vasoactive agents, prostanoids, leukotrienes and Reactive Oxygen Species (ROS) develop. The aim of this study was to evaluate the levels of ROS in cystic and follicular fluid, in order to establish their involvement in the etiopathogenesis of Cystic Ovarian Follicle (COF) in dairy cows. The study was conducted in 30 healthy cows (group C) and 30 cows affected by COF (group COF). The fluid of follicular cysts and of preovulatory follicles was drawn by means of ultrasound guided aspiration from the cows of both groups. The fluid obtained was analyzed by a photometric analytical system to detect ROS level. ROS concentration was statistically lower in the cystic fluid than in the follicular one (62.4 +/- 13.36 U.Carr vs. 84.89 +/- 26.99 U.Carr) (p<0.05), thus suggesting that an alteration of the cascade responsible for ROS production may be implicated in the complex etipathogenesis of COF.
Collapse
Affiliation(s)
- Annalisa Rizzo
- Department of Animal Production, University of Bari, Bari, Italy
| | | | | | | | | | | | | |
Collapse
|
13
|
Cardiac fibroblasts: at the heart of myocardial remodeling. Pharmacol Ther 2009; 123:255-78. [PMID: 19460403 DOI: 10.1016/j.pharmthera.2009.05.002] [Citation(s) in RCA: 763] [Impact Index Per Article: 50.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2009] [Accepted: 05/05/2009] [Indexed: 12/24/2022]
Abstract
Cardiac fibroblasts are the most prevalent cell type in the heart and play a key role in regulating normal myocardial function and in the adverse myocardial remodeling that occurs with hypertension, myocardial infarction and heart failure. Many of the functional effects of cardiac fibroblasts are mediated through differentiation to a myofibroblast phenotype that expresses contractile proteins and exhibits increased migratory, proliferative and secretory properties. Cardiac myofibroblasts respond to proinflammatory cytokines (e.g. TNFalpha, IL-1, IL-6, TGF-beta), vasoactive peptides (e.g. angiotensin II, endothelin-1, natriuretic peptides) and hormones (e.g. noradrenaline), the levels of which are increased in the remodeling heart. Their function is also modulated by mechanical stretch and changes in oxygen availability (e.g. ischaemia-reperfusion). Myofibroblast responses to such stimuli include changes in cell proliferation, cell migration, extracellular matrix metabolism and secretion of various bioactive molecules including cytokines, vasoactive peptides and growth factors. Several classes of commonly prescribed therapeutic agents for cardiovascular disease also exert pleiotropic effects on cardiac fibroblasts that may explain some of their beneficial outcomes on the remodeling heart. These include drugs for reducing hypertension (ACE inhibitors, angiotensin receptor blockers, beta-blockers), cholesterol levels (statins, fibrates) and insulin resistance (thiazolidinediones). In this review, we provide insight into the properties of cardiac fibroblasts that underscores their importance in the remodeling heart, including their origin, electrophysiological properties, role in matrix metabolism, functional responses to environmental stimuli and ability to secrete bioactive molecules. We also review the evidence suggesting that certain cardiovascular drugs can reduce myocardial remodeling specifically via modulatory effects on cardiac fibroblasts.
Collapse
|
14
|
Polymorphisms of MMP-2 Gene are Associated With Systolic Heart Failure Risk in Han Chinese. Am J Med Sci 2009; 337:344-8. [DOI: 10.1097/maj.0b013e31818eb2a2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
15
|
Polymorphisms of MMP-2 gene are associated with systolic heart failure prognosis. Clin Chim Acta 2009; 404:119-23. [PMID: 19332048 DOI: 10.1016/j.cca.2009.03.030] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2008] [Revised: 03/09/2009] [Accepted: 03/10/2009] [Indexed: 11/21/2022]
Abstract
BACKGROUND MMP-2 is a proteolytic enzyme involved in myocardial remodeling that occurs in congestive heart failure (HF). We hypothesized MMP-2 genetic variations could influence the prognosis of systolic HF. METHODS To test our hypothesis, we performed a follow-up study of 605 patients with systolic HF. Three single nucleotide polymorphisms (SNPs) of MMP-2 (rs243864, rs243866, rs17859821) were analyzed by restriction fragment length polymorphism (RFLP) methods. RESULTS Totally 526 patients (86.9%) were followed up. At follow up (median 24 months), 116 patients (22.1%) died, 102 patients (19.4%) were readmitted because of HF. One, two, three and four year survival rate was 86.9%, 81%, 77.9% and 77.9%. MMP-2 rs17859821 A allele carriers had lower all cause death rate, cardiac death rate and MACE rate than did GG genotype carriers (OR = 0.655, 0.580, 0.705; P = 0.030, 0.008, 0.011). After adjustment for age, bundle branch block, LVEF and NYHA grade by using cox regression analysis, MMP-2 A allele carriers had lower cardiac death rate and MACE rate than did GG genotype carriers (OR = 0.643 and 0.746; P < 0.05). However, the genotypes had no association with plasma levels of proMMP-2. Haplotype analysis had confirmed the above results. MMP-2 rs243866, rs243864 had no association with systolic HF prognosis. CONCLUSION The findings of the present study suggest that MMP-2 rs17859821 A allele was associated with better prognosis of systolic HF in the northern Han Chinese population.
Collapse
|
16
|
Wan R, Mo Y, Zhang X, Chien S, Tollerud DJ, Zhang Q. Matrix metalloproteinase-2 and -9 are induced differently by metal nanoparticles in human monocytes: The role of oxidative stress and protein tyrosine kinase activation. Toxicol Appl Pharmacol 2008; 233:276-85. [PMID: 18835569 PMCID: PMC2633358 DOI: 10.1016/j.taap.2008.08.022] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Revised: 08/22/2008] [Accepted: 08/26/2008] [Indexed: 11/22/2022]
Abstract
Recently, many studies have shown that nanoparticles can translocate from the lungs to the circulatory system. As a particulate foreign body, nanoparticles could induce host responses such as reactive oxygen species (ROS) generation, inflammatory cytokine and matrix metalloproteinase (MMP) release which play a major role in tissue destruction and remodeling. However, the direct effects of nanoparticles on leukocytes, especially monocytes, are still unclear. The objective of the present study was to compare the ability of Nano-Co and Nano-TiO(2) to cause alteration of transcription and activity of MMPs and to explore possible mechanisms. We hypothesized that non-toxic doses of some transition metal nanoparticles stimulate an imbalance of MMP/TIMP that cause MMP production that may contribute to their health effects. To test this hypothesis, U937 cells were treated with Nano-Co and Nano-TiO(2) and cytotoxic effects and ROS generation were measured. The alteration of MMP-2 and MMP-9 expression and activity of MMP-2 and MMP-9 after exposure to these metal nanoparticles were subsequently determined. To investigate the potential signaling pathways involved in the Nano-Co-induced MMP activation, the ROS scavengers or inhibitors, AP-1 inhibitor, and protein tyrosine kinase (PTK) inhibitors were also used to pre-treat U937 cells. Our results demonstrated that exposure of U937 cells to Nano-Co, but not to Nano-TiO(2), at a dose that does not cause cytotoxicity, resulted in ROS generation and up-regulation of MMP-2 and MMP-9 mRNA expression(..) Our results also showed dose- and time-related increases in pro-MMP-2 and pro-MMP-9 gelatinolytic activities in conditioned media after exposure of U937 cells to Nano-Co, but not to Nano-TiO(2). Nano-Co-induced pro-MMP-2 and pro-MMP-9 activity increases were inhibited by pre-treatment with ROS scavengers or inhibitors. We also demonstrated dose- and time-related decreases in tissue inhibitors of metalloproteinases 2 (TIMP-2) in U937 cells after exposure to Nano-Co, but not to Nano-TiO(2). However, neither Nano-Co nor Nano-TiO(2) exposure led to any transcriptional change of TIMP-1. The decrease of TIMP-2 after exposure to Nano-Co was also inhibited by pre-treatment with ROS scavengers or inhibitors. Our results also showed that pre-treatment of U937 cells with AP-1 inhibitor, curcumin, or the PTK specific inhibitor, herbimycin A or genistein, prior to exposure to Nano-Co, significantly abolished Nano-Co-induced pro-MMP-2 and-9 activity. Our results suggest that Nano-Co causes an imbalance between the expression and activity of MMPs and their inhibitors which is mediated by the AP-1 and tyrosine kinase pathways due to oxidative stress.
Collapse
Affiliation(s)
- Rong Wan
- Department of Environmental Health and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 555 South Floyd Street, Louisville, KY 40202, USA
- Department of Pathology, Fujian Medical University, Fujian, PR China
| | - Yiqun Mo
- Department of Environmental Health and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 555 South Floyd Street, Louisville, KY 40202, USA
| | - Xing Zhang
- Department of Hygiene, Zhejiang Academy of Medical Sciences, Zhejiang, PR China
| | - Sufan Chien
- Department of Surgery, University of Louisville, Louisville, KY, USA
| | - David J. Tollerud
- Department of Environmental Health and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 555 South Floyd Street, Louisville, KY 40202, USA
| | - Qunwei Zhang
- Department of Environmental Health and Occupational Health Sciences, School of Public Health and Information Sciences, University of Louisville, 555 South Floyd Street, Louisville, KY 40202, USA
| |
Collapse
|
17
|
Spinale FG. Myocardial Matrix Remodeling and the Matrix Metalloproteinases: Influence on Cardiac Form and Function. Physiol Rev 2007; 87:1285-342. [DOI: 10.1152/physrev.00012.2007] [Citation(s) in RCA: 855] [Impact Index Per Article: 50.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
It is now becoming apparent that dynamic changes occur within the interstitium that directly contribute to adverse myocardial remodeling following myocardial infarction (MI), with hypertensive heart disease and with intrinsic myocardial disease such as cardiomyopathy. Furthermore, a family of matrix proteases, the matrix metalloproteinases (MMPs) and the tissue inhibitors of MMPs (TIMPs), has been recognized to play an important role in matrix remodeling in these cardiac disease states. The purpose of this review is fivefold: 1) to examine and redefine the myocardial matrix as a critical and dynamic entity with respect to the remodeling process encountered with MI, hypertension, or cardiomyopathic disease; 2) present the remarkable progress that has been made with respect to MMP/TIMP biology and how it relates to myocardial matrix remodeling; 3) to evaluate critical translational/clinical studies that have provided a cause-effect relationship between alterations in MMP/TIMP regulation and myocardial matrix remodeling; 4) to provide a critical review and analysis of current diagnostic, prognostic, and pharmacological approaches that utilized our basic understanding of MMP/TIMPs in the context of cardiac disease; and 5) most importantly, to dispel the historical belief that the myocardial matrix is a passive structure and supplant this belief that the regulation of matrix protease pathways such as the MMPs and TIMPs will likely yield a new avenue of diagnostic and therapeutic strategies for myocardial remodeling and the progression to heart failure.
Collapse
|
18
|
Zhong Y, Gao J, Ye W, Huang P, Cheng Y, Jiao Q. Effect of Latanoprost Acid and Pilocarpine on Cultured Rabbit Ciliary Muscle Cells. Ophthalmic Res 2007; 39:232-40. [PMID: 17622744 DOI: 10.1159/000104832] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2006] [Accepted: 05/09/2007] [Indexed: 12/29/2022]
Abstract
OBJECTIVE To study the interaction between latanoprost and pilocarpine on cultured rabbit ciliary muscle (RCM) cells, and investigate the time courses of the two drugs, when given alone or in combination. METHODS Cultured RCM cells were treated for 24 h with different concentrations of latanoprost acid, pilocarpine and mixtures of latanoprost acid and pilocarpine. RNA was extracted, expressions of matrix metalloproteinase 1 (MMP-1), tissue inhibitor of metalloproteinase 1 (TIMP-1) and TIMP-2 were detected by reverse transcription-polymerase chain reaction (RT-PCR), and the optimum concentrations of those drugs were found. Then the cells were treated with the optimum concentrations of those drugs for various periods. RNA was extracted after the treatment and expressions of MMP-1, TIMP-1 and TIMP-2 were detected by RT-PCR again. Changes in [Ca(2+)](i) were estimated by fluorescence measurement using the Ca(2+) indicator Fluo-3 AM with a laser scanning confocal microscope. [Ca(2+)](i) of each cell was monitored continually after administration of the drugs. Gray values at 5 s and 2, 4, 6, 8 and 10 min were chosen for statistical analysis, and the influence and time-effect relationship of those drugs on [Ca(2+)](i) of the cultured cells were evaluated. RESULTS Exposure of the cells to increasing concentrations of latanoprost acid induced increased MMP-1 mRNA and decreased TIMP-1 and TIMP-2 mRNA in a dose-dependent manner. After 24 h of treatment, the optimum concentration of latanoprost acid for maximal changes in MMP-1 and TIMP-2 expression was 2 x 10(-7)M, and for maximal changes in TIMP-1 expression, the optimum concentration was 5 x 10(-7)M. When the optimum concentrations of latanoprost acid were chosen to treat the cells for various periods, the optimum time of the peak MMP-1 expression and trough TIMP-1 expression was 24 h, and of the trough TIMP-2 expression, it was 36 h after initiation of treatment. No significant expression changes of MMP-1, TIMP-1 and TIMP-2 mRNA were found when the cells were treated with pilocarpine at any concentration or at any time. Exposure of the cells to the mixtures of latanoprost acid and pilocarpine induced the same changes and time course of MMP-1, TIMP-1, and TIMP-2 mRNA expression as exposure of the cells to latanoprost acid alone. Exposure of ciliary muscle cells to pilocarpine induced an increase in [Ca(2+)](i), with the peak of increase observed at 5 s after initiation of treatment; then [Ca(2+)](i) gradually decreased near to baseline level within 10 min. Exposure of the cells to latanoprost acid did not significantly change [Ca(2+)](i). Exposure of the cells to the mixtures of latanoprost acid and pilocarpine induced the same [Ca(2+)](i) change as exposure to pilocarpine alone. CONCLUSION Latanoprost and pilocarpine have no interaction in their various effects on the cultured RCM cells.
Collapse
Affiliation(s)
- Yisheng Zhong
- Department of Ophthalmology, Ruijin Hospital Affiliated with Shanghai Jiaotong University, Shanghai, China.
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
Cardiac hypertrophy and heart failure are major causes of morbidity and mortality in Western societies. Many factors have been implicated in cardiac remodeling, including alterations in gene expression in myocytes, cardiomyocytes apoptosis, cytokines and growth factors that influence cardiac dynamics, and deficits in energy metabolism as well as alterations in cardiac extracellular matrix composition. Many therapeutic means have been shown to prevent or reverse cardiac hypertrophy. New concepts for characterizing the pathophysiology of cardiac hypertrophy have been drawn from various aspects, including medical therapy and gene therapy, or use of stem cells for tissue regeneration. In this review, we focus on various types of cardiac hypertrophy, defining the causes of hypertrophy, describing available animal models of hypertrophy, discussing the mechanisms for development of hypertrophy and its transition to heart failure, and presenting the potential use of novel promising therapeutic strategies derived from new advances in basic scientific research.
Collapse
Affiliation(s)
- Sudhiranjan Gupta
- Department of Molecular Cardiology, Lerner Research Institute, The Cleveland Clinic, Cleveland, Ohio 44195, USA
| | | | | |
Collapse
|
20
|
Tyagi N, Ovechkin AV, Lominadze D, Moshal KS, Tyagi SC. Mitochondrial mechanism of microvascular endothelial cells apoptosis in hyperhomocysteinemia. J Cell Biochem 2006; 98:1150-62. [PMID: 16514665 PMCID: PMC3182486 DOI: 10.1002/jcb.20837] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
An elevated level of homocysteine (Hcy) limits the growth and induces apoptosis. However, the mechanism of Hcy-induced programmed cell death in endothelial cells is largely unknown. We hypothesize that Hcy induces intracellular reactive oxygen species (ROS) production that leads to the loss of transmembrane mitochondrial potential (Deltapsi(m)) accompanied by the release of cytochrome-c from mitochondria. Cytochrome-c release contributes to caspase activation, such as caspase-9, caspase-6, and caspase-3, which results in the degradation of numerous nuclear proteins including poly (ADP-ribose) polymerase (PARP), which subsequently leads to the internucleosomal cleavage of DNA, resulting cell death. In this study, rat heart microvascular endothelial cells (MVEC) were treated with different doses of Hcy at different time intervals. Apoptosis was measured by DNA laddering and transferase-mediated dUTP nick-end labeling (TUNEL) assay. ROS production and MP were determined using fluorescent probes (2,7-dichlorofluorescein (DCFH-DA) and 5,5',6,6'-tetrachloro-1,1',3,3'-tetraethyl-benzamidazolocarbocyanin iodide (JC-1), respectively, by confocal microscopy. Differential gene expression for apoptosis was analyzed by cDNA array. The results showed that Hcy-mediated ROS production preceded the loss of MP, the release of cytochrome-c, and the activation of caspase-9 and -3. Moreover the Hcy treatment resulted in a decrease in Bcl(2)/Bax ratio, evaluated by mRNA levels. Caspase-9 and -3 were activated, causing cleavage of PARP, a hallmark of apoptosis and internucleosomal DNA fragmentation. The cytotoxic effect of Hcy was blocked by using small interfering RNA (siRNA)-mediated suppression of caspase-9 in MVEC. Suppressing the activation of caspase-9 inhibited the activation of caspase -3 and enhanced the cell viability and MP. Our data suggested that Hcy-mediated ROS production promotes endothelial cell death in part by disturbing MP, which results in subsequent release of cytochrome-c and activation of caspase-9 and 3, leading to cell death.
Collapse
Affiliation(s)
| | | | | | | | - Suresh C. Tyagi
- Correspondence to: Suresh C. Tyagi, PhD, Department of Physiology and Biophysics, University of Louisville School of Medicine, A-1115, 500 S Preston Street, Louisville, KY 40202.
| |
Collapse
|
21
|
Shastry S, Tyagi N, Moshal KS, Lominadze D, Hayden MR, Tyagi SC. GABA receptors ameliorate Hcy-mediated integrin shedding and constrictive collagen remodeling in microvascular endothelial cells. Cell Biochem Biophys 2006; 45:157-65. [PMID: 16757816 PMCID: PMC3182507 DOI: 10.1385/cbb:45:2:157] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/25/2023]
Abstract
Mammalian endothelial cells are deficient in cystathionine beta synthetase (CBS) activity, which is responsible for homocysteine (Hcy) clearance. This deficiency makes the endothelium the prime target for Hcy toxicity. Hcy induces integrin shedding in microvascular endothelial cells (MVEC) by increasing matrix metalloproteinase (MMP). Hcy competes with inhibitory neurotransmitter gamma aminobutyric acid (GABA)-A receptor. We hypothesized that Hcy transduces MVEC remodeling by increasing metalloproteinase activity and shedding beta-1 integrin by inactivating the GABA-A/B receptors, thus behaving as an excitatory neurotransmitter. MVEC were isolated from mouse brain. The presence of GABA-A receptor was determined by immunolabeling. It was induced by muscimol, an agonist of GABA-A receptor as measured by Western blot analysis. Hcy induced MMP-2 activity in a dose- and time-dependent manner, measured by zymography. GABA-A/B receptors ameliorated the Hcymediated MMP-2 activation. Hcy selectively increased the levels of tissue inhibitor of metalloproteinase (TIMP)-1 and TIMP-3 but decreased the levels of TIMP-4. Treatment with muscimol decreased the levels of TIMP-1 and TIMP-3 and increased the levels of TIMP-4 to control. Hcy caused a robust increase in the levels of a disintegrin and metalloproteinase (ADAM)-12. In the medium of MVEC treated with Hcy, the levels of beta-1 integrin were significantly increased. Treatment with muscimol or baclofen (GABA-B receptor agonist) ameliorated the levels of beta-1 integrin in the medium. These results suggested that Hcy induced ADAM-12. Significantly, Hcy facilitated the beta-1 integrin shedding. Treatment of MVEC with muscimol or baclofen during Hcy administration ameliorated the expression of metalloproteinase, integrin-shedding, and constrictive collagen remodeling, suggesting a role of Hcy in GABA receptor-mediated cerebrovascular remodeling.
Collapse
Affiliation(s)
| | | | | | | | | | - Suresh C. Tyagi
- Author to whom all correspondence and reprint requests should be addressed.
| |
Collapse
|
22
|
Moshal KS, Singh M, Sen U, Rosenberger DSE, Henderson B, Tyagi N, Zhang H, Tyagi SC. Homocysteine-mediated activation and mitochondrial translocation of calpain regulates MMP-9 in MVEC. Am J Physiol Heart Circ Physiol 2006; 291:H2825-35. [PMID: 16877562 DOI: 10.1152/ajpheart.00377.2006] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyperhomocysteinemia (HHcy) is associated with atherosclerosis, stroke, and dementia. Hcy causes extracellular matrix remodeling by the activation of matrix metalloproteinase-9 (MMP-9), in part, by inducing redox signaling and modulating the intracellular calcium dynamics. Calpains are the calcium-dependent cysteine proteases that are implicated in mitochondrial damage via oxidative burst. Mitochondrial abnormalities have been identified in HHcy. The mechanism of Hcy-induced extracellular matrix remodeling by MMP-9 activation via mitochondrial pathway is largely unknown. We report a novel role of calpains in mitochondrial-mediated MMP-9 activation by Hcy in cultured rat heart microvascular endothelial cells. Our observations suggested that calpain regulates Hcy-induced MMP-9 expression and activity. We showed that Hcy activates calpain-1, but not calpain-2, in a calcium-dependent manner. Interestingly, the enhanced calpain activity was not mirrored by the decreased levels of its endogenous inhibitor calpastatin. We presented evidence that Hcy induces the translocation of active calpain from cytosol to mitochondria, leading to MMP-9 activation, in part, by causing intramitochondrial oxidative burst. Furthermore, studies with pharmacological inhibitors of calpain (calpeptin and calpain-1 inhibitor), ERK (PD-98059) and the mitochondrial uncoupler FCCP suggested that calpain and ERK-1/2 are the major events within the Hcy/MMP-9 signal axis and that intramitochondrial oxidative stress regulates MMP-9 via ERK-1/2 signal cascade. Taken together, these findings determine the novel role of mitochondrial translocation of calpain-1 in MMP-9 activation during HHcy, in part, by increasing mitochondrial oxidative stress.
Collapse
Affiliation(s)
- Karni S Moshal
- Dept. of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40202, USA
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Rude MK, Duhaney TAS, Kuster GM, Judge S, Heo J, Colucci WS, Siwik DA, Sam F. Aldosterone Stimulates Matrix Metalloproteinases and Reactive Oxygen Species in Adult Rat Ventricular Cardiomyocytes. Hypertension 2005; 46:555-61. [PMID: 16043662 DOI: 10.1161/01.hyp.0000176236.55322.18] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Matrix metalloproteinases (MMPs), aldosterone, and reactive oxygen species (ROS) are implicated in myocardial remodeling. Although ROS, cytokines, and neurohormones regulate MMP in cardiac fibroblasts, it is unknown whether aldosterone regulates MMP in cardiomyocytes. Therefore, we tested the hypothesis that aldosterone regulates MMP in cultured adult rat ventricular myocytes (ARVMs). ARVMs were treated with aldosterone for 24 hours, and MMP-2 and MMP-9 activities were measured by zymography. Aldosterone (50 nmol/L) increased MMP-2 (43+/-5%) and MMP-9 (55+/-15%; P<0.001 for both) activities. Pretreatment with spironolactone (100 nmol/L) abolished the aldosterone-induced increase in MMP activities. Aldosterone (50 nmol/L; 30 minutes) increased mitogen/extracellular signal-regulated kinase (MEK) (31+/-3%) and extracellular signal-regulated kinase 1/2 (ERK1/2; 41+/-7%; P<0.001 for both) phosphorylation. U0126 (10 micromol/L), an MEK1/2 inhibitor, abolished the aldosterone-induced increase in MMP activities. Aldosterone increased intracellular ROS as assessed by dichlorofluorescein diacetate (27+/-4%; P<0.05). This increase was inhibited by apocynin, an NADPH oxidase inhibitor. Apocynin likewise inhibited aldosterone-induced ERK1/2 phosphorylation and the increase in MMP activities. Furthermore, the antioxidants MnTMPyP and N-acetylcysteine inhibited the aldosterone-induced increase in ERK1/2 phosphorylation and MMP activities, respectively. Protein kinase C (PKC) is implicated in the nongenomic effects of aldosterone. To test the role of PKC, ARVMs were pretreated with chelerythrine, a PKC inhibitor. Chelerythrine prevented the aldosterone-induced increase in ERK1/2 phosphorylation and MMP activities. Thus, aldosterone induces MMP activity in ARVM via activation of the mineralocorticoid receptor, PKC, and ROS-dependent activation of the MEK/ERK pathway. NADPH oxidase is a likely source of ROS in this system.
Collapse
Affiliation(s)
- Mary K Rude
- Whitaker Cardiovascular Institute, Boston University Medical Center, Massachusetts, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Tyagi N, Sedoris KC, Steed M, Ovechkin AV, Moshal KS, Tyagi SC. Mechanisms of homocysteine-induced oxidative stress. Am J Physiol Heart Circ Physiol 2005; 289:H2649-56. [PMID: 16085680 DOI: 10.1152/ajpheart.00548.2005] [Citation(s) in RCA: 285] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyperhomocysteinemia decreases vascular reactivity and is associated with cardiovascular morbidity and mortality. However, pathogenic mechanisms that increase oxidative stress by homocysteine (Hcy) are unsubstantiated. The aim of this study was to examine the molecular mechanism by which Hcy triggers oxidative stress and reduces bioavailability of nitric oxide (NO) in cardiac microvascular endothelial cells (MVEC). MVEC were cultured for 0-24 h with 0-100 microM Hcy. Differential expression of protease-activated receptors (PARs), thioredoxin, NADPH oxidase, endothelial NO synthase, inducible NO synthase, neuronal NO synthase, and dimethylarginine-dimethylaminohydrolase (DDAH) were measured by real-time quantitative RT-PCR. Reactive oxygen species were measured by using a fluorescent probe, 2',7'-dichlorofluorescein diacetate. Levels of asymmetric dimethylarginine (ADMA) were measured by ELISA and NO levels by the Griess method in the cultured MVEC. There were no alterations in the basal NO levels with 0-100 microM Hcy and 0-24 h of treatment. However, Hcy significantly induced inducible NO synthase and decreased endothelial NO synthase without altering neuronal NO synthase levels. There was significant accumulation of ADMA, in part because of reduced DDAH expression by Hcy in MVEC. Nitrotyrosine expression was increased significantly by Hcy. The results suggest that Hcy activates PAR-4, which induces production of reactive oxygen species by increasing NADPH oxidase and decreasing thioredoxin expression and reduces NO bioavailability in cultured MVEC by 1) increasing NO2-tyrosine formation and 2) accumulating ADMA by decreasing DDAH expression.
Collapse
Affiliation(s)
- Neetu Tyagi
- Dept. of Physiology and Biophysics, School of Medicine, 500 S. Preston St., 1115-A, Univ. of Louisville, Louisville, KY 40202, USA
| | | | | | | | | | | |
Collapse
|
25
|
Moshal KS, Tyagi N, Moss V, Henderson B, Steed M, Ovechkin A, Aru GM, Tyagi SC. Early induction of matrix metalloproteinase-9 transduces signaling in human heart end stage failure. J Cell Mol Med 2005; 9:704-13. [PMID: 16202218 PMCID: PMC6741634 DOI: 10.1111/j.1582-4934.2005.tb00501.x] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Extracellular matrix (ECM) turnover is regulated by matrix metalloproteinases (MMPs) and plays an important role in cardiac remodeling. Previous studies from our lab demonstrated an increase in gelatinolytic-MMP-2 and -9 activities in endocardial tissue from ischemic cardiomyopathic (ICM) and idiopathic dilated cardiomyopathic (DCM) hearts. The signaling mechanism responsible for the left ventricular (LV) remodeling, however, is unclear. Administration of cardiac specific inhibitor of metalloproteinase (CIMP) prevented the activation of MMP-2 and -9 in ailing to failing myocardium. Activation of MMP-2 and -9 leads to induction of proteinase activated receptor-1 (PAR-1). We hypothesize that the early induction of MMP-9 is a key regulator for modulating intracellular signaling through activation of PAR and various downstream events which are implicated in development of cardiac fibrosis in an extracellular receptor mediated kinase-1 (ERK-1) and focal adhesion kinase (FAK) dependent manner. To test this hypothesis, explanted human heart tissues from ICM and DCM patients were obtained at the time of orthotopic cardiac transplants. Quantitative analysis of MMP-2 and -9 gelatinolytic activities was made by real-time quantitative zymography. Gel phosphorylation staining for PAR-1 showed a significant increase in ICM hearts. Western blot and RT-PCR analysis and in-situ labeling, showed significant increased expression of PAR-1, ERK-1and FAK in ICM and DCM. These observations suggest that the enhanced expression and potentially increased activity of LV myocardial MMP-9 triggers the signal cascade instigating cardiac remodeling. This early mechanism for the initiation of LV remodeling appears to have a role in end-stage human heart failure.
Collapse
Affiliation(s)
- Karni S Moshal
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Moshal KS, Tyagi N, Henderson B, Ovechkin AV, Tyagi SC. Protease-activated receptor and endothelial-myocyte uncoupling in chronic heart failure. Am J Physiol Heart Circ Physiol 2005; 288:H2770-7. [PMID: 15681708 DOI: 10.1152/ajpheart.01146.2004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
We examined the hypothesis that oxidants generated nitroso derivatives, activated latent matrix metalloproteinase (MMP), and induced proteinase-activated receptor 1 (PAR-1), leading to disconnection between the endothelium and myocytes. Administration of cardiospecific tissue inhibitor of metalloproteinase-4 (TIMP-4/CIMP) ameliorated the oxidative-proteolytic stress and endothelial-myocyte uncoupling in chronic heart failure (CHF) in mice. Aortic-vena cava fistula (AVF) was created in 30 male mice (C57BL/6J) and studied at 0-, 2-, and 8-wk AVF. To reverse cardiac remodeling, as measured by MMP activation, purified CIMP was administered by an osmotic minipump subcutaneously after 8-wk AVF, and groups of mice ( n = 6 mice/group) were examined after 12 and 16 wk. Levels of PAR-1 in the left ventricle (LV) were increased at 2 and 8 wk (compared with 0 wk of no CIMP treatment) but were normal at 12 and 16 wk after CIMP treatment, as measured by Western blot analysis. Similar results were obtained for LV levels of nitrotyrosine, MMP-2 and -9 activities, and TIMP-1 and -3. However, the levels of TIMP-4, endothelial cell density, and responses of cardiac rings to acetylcholine and bradykinin were attenuated at 2 and 8 wk and normalized after CIMP administration in AVF mice. CIMP induced nitric oxide in microvascular endocardial endothelial cells. The results suggest that nitro generation activated MMP and PAR-1, leading to endothelial-myocyte uncoupling. CIMP treatment normalized PAR-1 expression and ameliorated endothelial-myocyte uncoupling by decreasing oxidant-mediated proteolytic stress in CHF.
Collapse
Affiliation(s)
- Karni S Moshal
- Univ. of Louisville School of Medicine, A-1115, Dept. of Physiology and Biophysics, 500 S. Preston St., Louisville, KY 40202, USA
| | | | | | | | | |
Collapse
|
27
|
Lovelock JD, Baker AH, Gao F, Dong JF, Bergeron AL, McPheat W, Sivasubramanian N, Mann DL. Heterogeneous effects of tissue inhibitors of matrix metalloproteinases on cardiac fibroblasts. Am J Physiol Heart Circ Physiol 2005; 288:H461-8. [PMID: 15650153 DOI: 10.1152/ajpheart.00402.2004] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The balance between matrix metalloproteinases (MMPs) and their natural inhibitors, the tissue inhibitors of metalloproteinases (TIMPs), plays a critical role in cardiac remodeling. Although a number of studies have characterized the pathophysiological role of MMPs in the heart, very little is known with respect to the role of TIMPs in the heart. To delineate the role of TIMPs in the heart we examined the effects of adenovirus-mediated overexpression of TIMP-1, -2, -3, and -4 in cardiac fibroblasts. Infection of cardiac fibroblasts with adenoviral constructs containing human recombinant TIMP (AdTIMP-1, -2, -3, and -4) provoked a significant (P < 0.0001) 1.3-fold in increase in bromodeoxyuridine (BrdU) incorporation. Similarly, treatment of cardiac fibroblasts with AdTIMP-1-, -2-, -3-, and -4-conditioned medium led to a 1.2-fold increase in BrdU incorporation (P < 0.0001) that was abolished by pretreatment with anti-TIMP-1, -2, -3, and -4 antibodies. The effects of TIMPs were not mimicked by treating the cells with RS-130830, a broad-based MMP inhibitor, suggesting that the effects of TIMPs were independent of their ability to inhibit MMPs. Infection with AdTIMP-1, -2, -3, and -4 led to a significant increase in alpha-smooth muscle actin staining, consistent with TIMP-induced phenotypic differentiation into myofibroblasts. Finally, infection with AdTIMP-2 resulted in a significant increase in collagen synthesis, whereas infection with AdTIMP-3 resulted in a significant increase in fibroblast apoptosis. TIMPs exert overlapping as well as diverse effects on isolated cardiac fibroblasts. The observation that TIMPs stimulate fibroblast proliferation as well as phenotypic differentiation into myofibroblasts suggests that TIMPs may play an important role in tissue repair in the heart that extends beyond their traditional role as MMP inhibitors.
Collapse
Affiliation(s)
- Joshua D Lovelock
- Winters Center for Heart Failure Research, Michael E. DeBakey Department of Veterans Affairs Medical Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Kizaki K, Okada M, Ito R, Yoshioka K, Hashizume K, Mutoh KI, Hara Y. Induction of Heparanase Gene Expression in Ventricular Myocardium of Rats with Isoproterenol-Induced Cardiac Hypertrophy. Biol Pharm Bull 2005; 28:2331-4. [PMID: 16327176 DOI: 10.1248/bpb.28.2331] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Gene expression of heparanase, matrix metalloproteinases (MMP)-2 and MMP-9 were examined in ventricles after chronic treatment with isoproterenol (ISO) induced cardiac hypertrophy in rats. Rats were treated with ISO (4 mg/kg, intraperitoneal) twice daily for 4 d. Ventricle weight of the heart and the ventricle weight/body weight ratio were increased respectively by 22% and 25% compared with control rats. Histology showed considerable cardiomyocyte hypertrophy in the ISO-treated rats in comparison to control rats. Northern blot hybridization revealed that heparanase and MMP-2 gene transcripts increased significantly in the ventricles of ISO-treated rats, whereas MMP-9 gene expression was not induced. Thus, heparanase and MMP-2 gene expressions are induced in the ventricle after chronic treatment with ISO, indicating that they might play an important role in development of ISO-induced cardiac hypertrophy.
Collapse
MESH Headings
- Animals
- Blotting, Northern/methods
- Cardiomegaly/chemically induced
- Cardiomegaly/enzymology
- Disease Models, Animal
- Drug Administration Schedule
- Enzyme Induction/drug effects
- Extracellular Matrix
- Female
- Gene Expression Regulation, Enzymologic/drug effects
- Glucuronidase/biosynthesis
- Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism
- Hypertrophy, Left Ventricular/enzymology
- Hypertrophy, Right Ventricular/enzymology
- Injections, Intraperitoneal
- Isoproterenol
- Male
- Matrix Metalloproteinase 2/biosynthesis
- Myocardium/enzymology
- Myocytes, Cardiac/enzymology
- RNA, Complementary/genetics
- RNA, Complementary/metabolism
- Rats
- Rats, Wistar
Collapse
Affiliation(s)
- Keiichiro Kizaki
- Department of Veterinary Pharmacology, School of Veterinary Medicine and Animal Sciences, Kitasato University, Towada, Aomori 034-8628, Japan.
| | | | | | | | | | | | | |
Collapse
|
29
|
Lalu MM, Pasini E, Schulze CJ, Ferrari-Vivaldi M, Ferrari-Vivaldi G, Bachetti T, Schulz R. Ischaemia–reperfusion injury activates matrix metalloproteinases in the human heart. Eur Heart J 2004; 26:27-35. [PMID: 15615796 DOI: 10.1093/eurheartj/ehi007] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
AIMS Matrix metalloproteinases (MMPs) and tissue inhibitors of metalloproteinases (TIMPs) regulate matrix remodelling in the heart and play a pivotal role in myocardial dysfunction immediately following ischaemia-reperfusion injury ex vivo in rats. We investigated the changes in MMPs and TIMPs in acute myocardial ischaemia-reperfusion injury in humans. METHODS AND RESULTS Fifteen patients with stable angina undergoing coronary artery bypass graft surgery with cardiopulmonary bypass were enrolled. Left ventricular stroke work index was monitored prior to bypass and for 24 h following reperfusion. Left atrial biopsy samples were obtained at the start of bypass before cardioplegia and within 10 min after removal of the aortic cross-clamp. Plasma samples were collected from the radial artery and coronary sinus 1, 5, and 10 min following removal of the cross-clamp. In cardiac biopsies there was a marked increase in 72 kDa MMP-2 and 92 kDa MMP-9 activities, and a decrease in TIMP-1 upon reperfusion. Increased MMP activity correlated positively with cross-clamp duration and inversely with cardiac mechanical function 3 h following reperfusion. TIMP-1 correlated inversely with cross-clamp time and positively with cardiac mechanical function. Plasma samples revealed a significant increase in both 92 kDa MMP-9 and 64 kDa MMP-2 activities 1 min following removal of cross-clamp. CONCLUSION Reperfusion following cardioplegia activates MMPs in the myocardium and plasma of patients undergoing coronary artery bypass grafting. This is the first correlation of MMP myocardial activity with cardiac function in humans. The early increase in MMP activity produces a proteolytic environment that may contribute to myocardial stunning injury in humans.
Collapse
Affiliation(s)
- Manoj M Lalu
- Department of Pharmacology, University of Alberta, Edmonton, Canada
| | | | | | | | | | | | | |
Collapse
|
30
|
Olkowski AA, Wojnarowicz C, Rathgeber BM, Abbott JA, Classen HL. Lesions of the pericardium and their significance in the aetiology of heart failure in broiler chickens. Res Vet Sci 2003; 74:203-11. [PMID: 12726738 DOI: 10.1016/s0034-5288(03)00004-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The present study focuses on lesions of the pericardium commonly observed in fast growing broilers. These lesions are examined in the context of electrophysiological and functional changes associated with cardiac performance and patho-physiology in broilers succumbing to acute or chronic heart failure. Typical lesions involving the pericardium in fast growing broiler chickens included: (1) excessive pericardial effusion, (2) locally extensive or focal adhesions between parietal and visceral components of the pericardium, (3) fibrous deposits on visceral pericardium, and (4) thickened pericardium. Echocardiographic evidence indicated that severe pericardial effusion and/or adhesions may have a restrictive effect on heart pump function, where both diastolic and systolic function of the heart may be affected. Electrocardiographic data showed a strong trend indicating that pericardial adhesions may be associated with ventricular arrhythmia and increased risk of sudden death in fast growing broilers. Relatively high levels of matrix metalloproteinase MMP-2 activity have been found in pericardial effusions from affected chickens, suggesting a possible involvement of this enzyme in the aetiology of pericardial lesions. The present results indicate that pericardial lesions may be associated with biochemical, morphological, electrophysiological, and functional changes occurring in the hearts of broilers succumbing to acute or chronic heart failure and ascites.
Collapse
Affiliation(s)
- A A Olkowski
- Department of Animal and Poultry Science, University of Saskatchewan, 6D34 Agriculture Building, 51 Campus Drive, Saskatoon, SK, Canada S7N 5A8.
| | | | | | | | | |
Collapse
|
31
|
Hunt MJ, Tyagi SC. Peroxisome proliferators compete and ameliorate Hcy-mediated endocardial endothelial cell activation. Am J Physiol Cell Physiol 2002; 283:C1073-9. [PMID: 12225971 DOI: 10.1152/ajpcell.00152.2002] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To determine whether homocysteine (Hcy)-mediated activation of endocardial endothelial (EE) cells is ameliorated by peroxisome proliferator-activated receptor (PPAR), we isolated EE cells from mouse endocardium. Matrix metalloproteinase (MMP) activity and intercellular adhesion molecule (ICAM)-1 in EE cells were measured in the presence and absence of Hcy, and ciprofibrate (CF; PPAR-alpha agonist) or 15-deoxy-Delta(12,14)-prostaglandin J(2) (PGJ(2); PPAR-gamma agonist) by zymography and Western blot analyses, respectively. Results suggest that Hcy-mediated MMP activation and ICAM-1 expression are ameliorated by CF and PGJ(2). To test the hypothesis that Hcy competes with other ligands for binding to PPARalpha and -gamma, we prepared cardiac nuclear extracts. Extracts were loaded onto an Hcy-cellulose affinity column. Bound proteins were eluted with CF and PGJ(2). To determine conformational changes in PPAR upon binding to Hcy, we measured PPAR fluorescence at 334 nm. Dose-dependent increase in PPAR fluorescence demonstrated a primary binding affinity of 0.32 +/- 0.06 microM. There was dose-dependent quenching of PPAR fluorescence by fluorescamine-homocysteine (F-Hcy). PPAR-alpha fluorescence quenching was abrogated by the addition of CF but not by PGJ(2). PPAR-gamma fluorescence quenching was abrogated by the addition of PGJ(2) but not by CF. These results suggest that Hcy competes with CF and PGJ(2) for binding to PPAR-alpha and -gamma, respectively, indicating a role of PPAR in amelioration of Hcy-mediated EE dysfunction.
Collapse
Affiliation(s)
- Matthew J Hunt
- Department of Physiology and Biophysics, University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| | | |
Collapse
|
32
|
Lalu MM, Csonka C, Giricz Z, Csont T, Schulz R, Ferdinandy P. Preconditioning decreases ischemia/reperfusion-induced release and activation of matrix metalloproteinase-2. Biochem Biophys Res Commun 2002; 296:937-41. [PMID: 12200138 DOI: 10.1016/s0006-291x(02)02019-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Release and activation of matrix metalloproteinases (MMPs) significantly contribute to myocardial stunning injury immediately after ischemia and reperfusion, however, their role in preconditioning remains unknown. We therefore examined the effects of preconditioning and subsequent ischemia/reperfusion on MMP activity in isolated rat hearts. Hearts were subjected to a preconditioning protocol (three consecutive 5-min periods of global ischemia interspersed with 5 min of reperfusion) followed by 30 min ischemia and 5 min reperfusion. To measure MMP release, coronary effluent was collected: (a) during aerobic perfusion, (b) in reperfusion following each preconditioning ischemia, and (c) during the final reperfusion following test ischemia. MMP-2 activities could be detected by gelatin zymography in the ventricles and coronary effluent samples from the perfused hearts. The levels of MMP-2 activity in the effluent were markedly increased in effluent following test ischemia from control hearts without preconditioning. This was accompanied by a decrease in corresponding tissue MMP activities. Preconditioning significantly decreased the MMP-2 activity in the coronary effluent following test ischemia/reperfusion and preserved the MMP-2 protein content and activity in the myocardium. Our results demonstrate that classic preconditioning inhibits ischemia/reperfusion induced release and activation of MMP-2. These results suggest that preconditioning may exert part of its cardioprotective effects through the reduction of MMP-2 release.
Collapse
Affiliation(s)
- Manoj M Lalu
- Cardiovascular Research Group, Department of Pharmacology, University of Alberta, Edmonton, AB, Canada T6G 2S2
| | | | | | | | | | | |
Collapse
|
33
|
Mujumdar VS, Tummalapalli CM, Aru GM, Tyagi SC. Mechanism of constrictive vascular remodeling by homocysteine: role of PPAR. Am J Physiol Cell Physiol 2002; 282:C1009-15. [PMID: 11940516 DOI: 10.1152/ajpcell.00353.2001] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To test the hypothesis that homocysteine induces constrictive vascular remodeling by inactivating peroxisome proliferator-activated receptor (PPAR), aortic endothelial cells (ECs) and smooth muscle cells (SMCs) were isolated. Collagen gels were prepared, and ECs or SMCs (10(5)) or SMCs + ECs (10(4)) were incorporated into the gels. To characterize PPAR, agonists of PPAR-alpha [ciprofibrate (CF)] and PPAR-gamma [15-deoxy-12,14-prostaglandin J(2) (PGJ(2))] were used. To determine the role of disintegrin metalloproteinase (DMP), cardiac inhibitor of metalloproteinase (CIMP) was used in collagen gels. Gel diameter at 0 h was 14.1 +/- 0.2 mm and was unchanged up to 24 h as measured by a digital micrometer. SMCs reduce gel diameter to 10.5 +/- 0.4 mm at 24 h. Addition of homocysteine to SMCs reduces further the gel diameter to 8.0 +/- 0.2 mm, suggesting that SMCs induce contraction and that the contraction is further enhanced by homocysteine. Addition of ECs and SMCs reduces gel diameter to 12.0 +/- 0.3 mm, suggesting that ECs play a role in collagen contraction. Only PGJ(2), not CF, inhibits SMC contraction. However, both PGJ(2) and CF inhibit contraction of ECs and SMCs + ECs. Addition of anti-DMP blocks SMC- as well as homocysteine-mediated contraction. However, CIMP inhibits only homocysteine-mediated contraction. The results suggest that homocysteine may enhance vascular constrictive remodeling by inactivating PPAR-alpha and -gamma in ECs and PPAR-gamma in SMCs.
Collapse
Affiliation(s)
- Vibhas S Mujumdar
- Department of Physiology and Biophysics, School of Medicine, The University of Mississippi Medical Center, Jackson, Mississippi 39216, USA
| | | | | | | |
Collapse
|
34
|
Olkowski AA, Rathgeber BM, Sawicki G, Classen HL. Ultrastructural and molecular changes in the left and right ventricular myocardium associated with ascites syndrome in broiler chickens raised at low altitude. JOURNAL OF VETERINARY MEDICINE. A, PHYSIOLOGY, PATHOLOGY, CLINICAL MEDICINE 2001; 48:1-14. [PMID: 11515307 DOI: 10.1046/j.1439-0442.2001.00329.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The present study examines ultrastructural and molecular changes in ventricular myocardium associated with ascites cases in fast-growing broilers raised at low altitude. Extensive ultrastructural lesions were seen in the left and right ventricular myocardium of broilers with fulminant heart failure and ascites. Significant changes included lesions in the myofibril contractile apparatus, altered mitochondria, marked reduction in the myofibril component, and changes in the extracellular matrix (ECM) architecture. No lesions were observed in hearts of slow growing broilers, but mild to moderate changes (predominantly in the left ventriculum) were apparent in the hearts from some clinically normal, fast-growing broilers. SDS-PAGE profiles of washed myofibrils showed several distinctly different bands in preparations from left ventricular myocardium of ascitic birds. Western blot analysis of these samples revealed several fragments of myosin heavy chain, M-protein, and titin. Based on gelatinolytic activity, matrix metalloproteinases (MMP) in the cytosolic fraction of ventricular myocardium homogenates were identified as MMP-2. The relative activity of this enzyme appears to be considerably higher in preparations from broilers, particularly in the preparations from the left ventriculum of fast-growing broilers, in comparison to leghorns or slow growing broilers. The nature and distribution of the changes in the heart indicate that chronic cardiomyopathic process in the left ventricular myocardium occurs during the development of ascites. It is postulated that progressive deterioration of the left heart pump function caused by initial lesions in the left ventricular myocardium is a significant factor in the development of pulmonary hypertension and the pathogenesis of ascites in broilers raised at low altitude.
Collapse
Affiliation(s)
- A A Olkowski
- Department of Animal and Poultry Science, University of Saskatchewan, 72 Campus Drive, Saskatoon, SK, S7N 5B5, Canada.
| | | | | | | |
Collapse
|
35
|
Tummalapalli CM, Heath BJ, Tyagi SC. Tissue inhibitor of metalloproteinase-4 instigates apoptosis in transformed cardiac fibroblasts. J Cell Biochem 2001; 80:512-21. [PMID: 11169735 DOI: 10.1002/1097-4644(20010315)80:4<512::aid-jcb1005>3.0.co;2-n] [Citation(s) in RCA: 67] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Tumor cells become malignant, in part, because of their activation of matrix metalloproteinases (MMPs) and inactivation of tissue inhibitor of metalloproteinases (TIMPs). Myocardial tumors are rarely malignant. This raises the possibility that the MMPs and TIMPs are differentially regulated in the heart compared to other tissues. Therefore, we hypothesized that a tissue specific tumor suppressor exists in the heart. To test this hypothesis we prepared cardiac tissue extracts from normal (n = 4), ischemic cardiomypathic (ICM) [n = 5], and dilated cardiomyopathic (DCM) [n = 8] human heart end-stage explants. The level of cardiospecific TIMP-4 was determined by SDS-PAGE and Western-blot analysis. The results suggested reduced levels of TIMP-4 in ICM and DCM as compared to normal heart. TIMP-4 was purified by reverse phase HPLC and gelatin-sepharose affinity chromatography. Collagenase inhibitory activity of chromatographic peaks was determined using fluorescein-conjugated collagen as substrate and fluorescence spectroscopy. The activity of TIMP-4 (27 kDa) was characterized by reverse zymography. The role of TIMP-4 in cardiac fibroblast cell migration was examined using Boyden chamber analysis. The results suggested that TIMP-4 inhibited cardiac fibroblast cells migration and collagen gel invasion. To test whether TIMP-4 induces apoptosis, we cultured cardiac normal and polyomavirus transformed fibroblast cells in the presence and absence of TIMP-4. The number of cells were measured and DNA laddering was determined. The results suggested that TIMP-4 controlled normal cardiac fibroblast transformation and induced apoptosis in transformed cells. Cardiospecific TIMP-4 plays a significant role in regulating the normal cell phenotype. The reduced levels of TIMP-4 elicit cellular transformation and may lead to adverse extracellular matrix degradation (remodeling), cardiac hypertrophy and failure. This study suggests a possible protective role of TIMP-4 in other organs which are susceptible to malignancy.
Collapse
Affiliation(s)
- C M Tummalapalli
- Department of Physiology and Biophysics, The University of Mississippi Medical Center, 2500 North State Street, Jackson, MS 39216, USA
| | | | | |
Collapse
|
36
|
Siwik DA, Pagano PJ, Colucci WS. Oxidative stress regulates collagen synthesis and matrix metalloproteinase activity in cardiac fibroblasts. Am J Physiol Cell Physiol 2001; 280:C53-60. [PMID: 11121376 DOI: 10.1152/ajpcell.2001.280.1.c53] [Citation(s) in RCA: 459] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Oxidative stress has been implicated in the pathophysiology of myocardial failure. We tested the hypothesis that oxidative stress can regulate extracellular matrix in cardiac fibroblasts. Neonatal and adult rat cardiac fibroblasts in vitro were exposed to H(2)O(2) (0.05-5 microM) or the superoxide-generating system xanthine (500 microM) plus xanthine oxidase (0.001-0.1 mU/ml) (XXO) for 24 h. In-gel zymography demonstrated that H(2)O(2) and XXO each increased gelatinase activity corresponding to matrix metalloproteinases (MMP) MMP-13, MMP-2, and MMP-9. H(2)O(2) and XXO decreased collagen synthesis (collagenase-sensitive [(3)H]proline incorporation) without affecting total protein synthesis ([(3)H]leucine incorporation). H(2)O(2) and XXO decreased the expression of procollagen alpha(1)(I), alpha(2)(I), and alpha(1)(III) mRNA but increased the expression of fibronectin mRNA, suggesting a selective transcriptional effect on collagen synthesis. H(2)O(2), but not XXO, also decreased the expression of nonfibrillar procollagen alpha(1)(IV) and alpha(2)(IV) mRNA. To determine the role of endogenous antioxidant systems, cells were treated with the superoxide dismutase (SOD) inhibitor diethyldithiocarbamic acid (DDC, 100 microM) to increase intracellular superoxide or with the glucose-6-phosphate dehydrogenase inhibitor dehydroisoandrosterone 3-acetate (DHEA; 10 microM) to increase intracellular H(2)O(2). DDC and DHEA decreased collagen synthesis and increased MMP activity, and both effects were inhibited by an SOD/catalase mimetic. Thus increased oxidative stress activates MMPs and decreases fibrillar collagen synthesis in cardiac fibroblasts. Oxidative stress may play a role in the pathogenesis of myocardial remodeling by regulating the quantity and quality of extracellular matrix.
Collapse
Affiliation(s)
- D A Siwik
- Myocardial Biology Unit, Whitaker Cardiovascular Institute, Boston University School of Medicine, Massachusetts 02118, USA
| | | | | |
Collapse
|
37
|
Mujumdar VS, Aru GM, Tyagi SC. Induction of oxidative stress by homocyst(e)ine impairs endothelial function. J Cell Biochem 2001; 82:491-500. [PMID: 11500925 DOI: 10.1002/jcb.1175] [Citation(s) in RCA: 132] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Previous studies have demonstrated a relationship between hyperhomocysteinemia and endothelial dysfunction, reduced bioavailability of nitric oxide, elastinolysis and, vascular muscle cell proliferation. In vivo decreased nitric oxide production is associated with increased matrix metalloproteinase (MMP) activity and formation of nitrotyrosine. To test the hypothesis that homocysteine neutralizes vascular endothelial nitric oxide, activates metalloproteinase, causes elastinolysis and vascular hypertrophy, we isolated aortas from normotensive Wistar rats and cultured them in medium containing homocysteine, and calf serum for 14 days. Homocysteine-mediated impairment of endothelial-dependent vasodilatation was reversed by co-incubation of homocysteine with nicotinamide (an inhibitor of peroxinitrite and nitrotyrosine), suggesting a role of homocysteine in redox-mediating endothelial dysfunction and nitrotyrosine formation. The Western blot analysis, using anti-nitrotyrosine antibody, on aortic tissue homogeneates demonstrated decreased nitrotyrosine in hyperhomocysteinemic vessels treated with nicotinamide. Zymographic analysis revealed increased elastinolytic gelatinase A and B (MMP-2, -9) in homocysteine treated vessels and the treatment with nicotinamide decreases the homocysteine-induced MMP activation. Morphometric analyses revealed significant medial hypertrophic thickening (1.4 +/- 0.2-fold of control, P = 0.03) and elastin disruption in homocysteine-treated vessels as compared to control. To determine whether homocysteine causes endothelial cell injury, cross-sections of aortas were analyzed for caspase activity by incubating with Ac-YVAD-AMC (substrate for apoptotic enzyme, caspase). The endothelium of homocysteine treated vessels, and endothelial cells treated with homocysteine, showed marked labeling for caspase. The length-tension relationship of homocysteine treated aortas was shifted to the left as compared to untreated aortas, indicating reduced vascular elastic compliance in homocysteine-treated vessels. Co-incubation of homocysteine and inhibitors of MMP, tissue inhibitor of metalloproteinase-4 (TIMP-4), and caspase, YVAD-CHO, improved vascular function. The results suggest that alteration in vascular elastin/collagen ratio and activation of MMP-2 are associated with decreased NO production in hyperhomocysteinemia.
Collapse
Affiliation(s)
- V S Mujumdar
- Department of Physiology and Biophysics, School of Medicine, The University of Mississippi Medical Center, Jackson, Mississippi 39216, USA
| | | | | |
Collapse
|
38
|
Abstract
Although a considerable amount of effort has been placed on discovering the etiologies of cancer, the majority of the basic cancer research existing today has focused on understanding the molecular mechanism of tumor formation and metastasis. Metastatic spread of tumors continues to be a major obstacle to successful treatment of malignant tumors. Approximately 30% of those patients diagnosed with a solid tumor have a clinically detectable metastasis and for the remaining 70%, metastases are continually being formed throughout the life of the tumor. Even after the tumor is excised, the threat of death is attributable to the metastasis that may occur through the remaining tumor cells. In addition, treating the metastasis often proves futile since metastasis often vary in size, composition, and anatomical location. New treatments blocking the formation of metastasis will provide greater chances of survival for cancer patients. One family of enzymes that has been shown over the years to play a role in tumor progression is the matrix metalloproteinase (MMP) family. The main function of MMPs, also known as matrixins, is degradation of the extracellular matrix physiologic function involving MMPs include wound healing, bone resorption and mammary involution. MMPs, however, also contribute to pathological conditions including rheumatoid arthritis, coronary artery disease, and cancer. Tumor cells are believed to utilize the matrix degrading capability of these enzymes to spread to distant sites. In addition, MMPs also are thought to promote the growth of these tumor cells once they have metastasized. This review will discuss the role of MMPs and their inhibitors in tumor invasion, angiogenesis and metastasis with special emphasis on the gelatinases, MMP-2 and MMP-9.
Collapse
Affiliation(s)
- A John
- Medical College of Pennsylvania and Hahnemann University School of Medicine, Department of Pathology, Philadelphia 19102, USA
| | | |
Collapse
|
39
|
Ma G, Mamaril JL, Young DB. Increased potassium concentration inhibits stimulation of vascular smooth muscle proliferation by PDGF-BB and bFGF. Am J Hypertens 2000; 13:1055-60. [PMID: 11041158 DOI: 10.1016/s0895-7061(99)00278-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
Abstract
The effects of changes in extracellular potassium concentration on the rate of vascular smooth muscle cell proliferation stimulated by cytokines and serum were analyzed in vitro. To analyze the DNA synthesis response, cells from swine coronary artery were grown in DMEM medium containing 3, 4, 5, or 6 mmol/L potassium together with 20 ng/mL platelet-derived growth factor BB (PDGF-BB), 25 ng/mL basic fibroblast growth factor (bFGF), or 5% fetal bovine serum (FBS), with [methyl 3H] thymidine added, for 24 h. Proliferation responses were analyzed in cells grown in medium with potassium concentrations of 3, 4, 5, or 6 mmol/L, together with either 20 ng/mL PDGF-BB, 25 ng/mL bFGF, or 5% FBS, for 7 days, then harvested and counted. Highly significant inverse relationships were observed between potassium concentration and 3H-thymidine incorporation stimulated by each of the three agonists (P < .01 for each, ANOVA), and between potassium concentration and proliferation (all P < .01, ANOVA). Elevation of potassium concentration within the physiologic range inhibits vascular smooth muscle cell DNA synthesis and proliferation.
Collapse
Affiliation(s)
- G Ma
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson 39216-4505, USA.
| | | | | |
Collapse
|
40
|
Siwik DA, Chang DL, Colucci WS. Interleukin-1beta and tumor necrosis factor-alpha decrease collagen synthesis and increase matrix metalloproteinase activity in cardiac fibroblasts in vitro. Circ Res 2000; 86:1259-65. [PMID: 10864917 DOI: 10.1161/01.res.86.12.1259] [Citation(s) in RCA: 343] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We tested the hypothesis that the inflammatory cytokines can regulate fibroblast extracellular matrix metabolism. Neonatal and adult rat cardiac fibroblasts cultures in vitro were exposed to interleukin (IL)-1beta (4 ng/mL), tumor necrosis factor-alpha (TNF-alpha; 100 ng/mL), IL-6 (10 ng/mL), or interferon-gamma (IFN-gamma; 500 U/mL) for 24 hours. IL-1beta, and to a lesser extent TNF-alpha, decreased collagen synthesis, which was measured as collagenase-sensitive [(3)H]proline incorporation, but had no effect on cell number or total protein synthesis. IL-1beta decreased the expression of procollagen alpha(1)(I), alpha(2)(I), and alpha1(III) mRNA, but increased the expression of procollagen alpha(1)(IV), alpha(2)(IV), and fibronectin mRNA, indicating a selective transcriptional downregulation of fibrillar collagen synthesis. IL-1beta and TNF-alpha each increased total matrix metalloproteinase (MMP) activity as measured by in-gel zymography, causing specific increases in the bands corresponding to MMP-13, MMP-2, and MMP-9. IL-1beta increased the expression of proMMP-2 and proMMP-3 mRNA, suggesting that increased metalloproteinase activity is due, at least in part, to increased transcription. The effects of IL-1beta were not dependent on NO production. Thus, IL-1beta and TNF-alpha decrease collagen synthesis and activate MMPs that degrade collagen. These observations suggest that IL-1beta and TNF-alpha may contribute to ventricular dilation and myocardial failure by promoting the remodeling of interstitial collagen.
Collapse
Affiliation(s)
- D A Siwik
- Myocardial Biology Unit, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA. USA
| | | | | |
Collapse
|
41
|
Abstract
Homocysteine found in the plasma of patients with coronary heart disease, induces vascular smooth muscle cell (VSMC) proliferation and increases deposition of extracellular matrix (ECM) components. Yet, the mechanism by which homocysteine mediates this effect and its role in vascular disease is largely unknown. We hypothesized that homocysteine induces ECM production via intracellular calcium release in VSMC. To test this hypothesis, aortic VSMC from Sprague-Dawley rats were isolated and characterized by positive labeling for vascular smooth muscle alpha-actin. Early passage cells (p2-3) were grown in monolayer on coverslips. Calcium transients were quantified with fura2/AM spectrofluorometry. Homocysteine induced intracellular calcium [Ca(2+)](i) transients with an EC(50) of 60 +/- 5 nM. The EC(50) for glutathione and cysteine were 10 and 100-fold lower, respectively. Depleting extracellular calcium did not alter the homocysteine effect on intracellular calcium; however, thapsigargin pretreatment, which depletes intracellular Ca(2+) stores, abolished the homocysteine effect, demonstrating its dependence on intracellular Ca(2+) stores. Extracellular sodium depletion significantly (P < 0.05) increased [Ca(2+)](i) also suggesting a possible role of sodium-calcium exchange in the process. To begin to elucidate the intracellular pathways by which homocysteine might act, VSMC were pretreated with specific inhibitors and stimulators prior to homocysteine stimulation. Staurosporine and phorbol myrisate acetate (PMA), potent simulators of protein kinase C, augmented the release of Ca(2+) by homocysteine. Interestingly, pretreatment with the nitric oxide synthase inhibitor N-nitro-L-arginine methyl ester (L-NAME) greatly exacerbated the sensitivity of VSMC to homocysteine. In contrast, pretreatment with either the phospholipase A(2) activator neomycin, the antioxidant and hepatic hydroxymethyl glutaryl coenzyme A (HMG CoA) reductase inhibitor, pravastatin, the tyrosine kinase inhibitor genestein, or the calcium channel blocker, felodipine completely inhibited the homocysteine-induced Ca(2+) signal in VSMC. This suggests the role of multiple signaling pathways in the homocysteine effect on VSMC Ca(2+). Effects of homocysteine on collagen production, as ascertained by immunoblot analysis, correlated with its effect in intracellular calcium. Regardless of the signaling pathways involved, homocysteine, by virtue of its role on VSMC proliferation and ECM deposition, has the potential to affect vascular reactivity. To determine the effect of homocysteine on the ability of VSMC to react to potent agonist such as angiotensin II, VSMC were pretreated with homocysteine and exposed to a range of angiotensin II concentrations which normally have no effect on intracellular Ca(2+). After homocysteine pretreatment, VSMC were extremely responsive to angiotensin II at concentrations well below the physiologic range. These data taken together suggested that an initial effect of homocysteine is to induce release of intracellular Ca(2+) in VSMC and may induce vascular reactivity. The transient in Ca(2+) correlates with the effect on ECM associated with homocysteine.
Collapse
Affiliation(s)
- V S Mujumdar
- Department of Physiology and Biophysics, Center of Excellence in Cardiovascular-Renal Research, University of Mississippi Medical Center, Jackson, Mississippi 39216-4505, USA
| | | | | |
Collapse
|
42
|
Ma G, Mason DP, Young DB. Inhibition of vascular smooth muscle cell migration by elevation of extracellular potassium concentration. Hypertension 2000; 35:948-51. [PMID: 10775567 DOI: 10.1161/01.hyp.35.4.948] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The effect of potassium on the migration of vascular smooth muscle cells was analyzed in media made with extracellular potassium concentrations of 3, 4, 5, and 6 mmol/L. The migration of cultured porcine coronary artery cells was stimulated with platelet-derived growth factor (PDGF)-BB. In the first study, cells were exposed to PDGF-BB at concentrations of 0, 10, or 20 ng/mL for 5 hours with the use of a Boyden chamber. Cells were quiescent overnight in 0.5% fetal bovine serum in Dulbecco's modified Eagle's medium with an extracellular potassium concentration of 4 mmol/L. With increasing potassium concentration, migration was significantly inhibited (P<0. 02, 2-way ANOVA). In the cells exposed to 10 ng/mL PDGF-BB, migration ranged from 500+/-86% to 294+/-44% (value in wells with 0 ng/mL PDGF-BB and 4 mmol/L potassium concentration=100%) in medium containing 3 to 6 mmol/L extracellular potassium concentration (P<0. 03). Long-term potassium exposure was investigated in cells grown in 5% serum in Dulbecco's modified Eagle's medium with an extracellular potassium concentration of 3, 4, 5, or 6 mmol/L for 3 to 4 weeks. Migration was assessed with 0 or 20 ng/mL PDGF-BB. Migration was significantly inhibited by the elevation of extracellular potassium concentration (P<0.01, 2-way ANOVA). With 20 ng/mL PDGF-BB, the migration rates ranged from 152+/-11% in medium with 3 mmol/L potassium to 69+/-5% in 6 mmol/L potassium (P<0.01). Increases in extracellular potassium concentration within the physiological range significantly and directly inhibit vascular smooth muscle cell migration.
Collapse
Affiliation(s)
- G Ma
- Department of Physiology and Biophysics , University of Mississippi Medical Center, Jackson, MS 39216-4505, USA
| | | | | |
Collapse
|
43
|
|
44
|
Coker ML, Doscher MA, Thomas CV, Galis ZS, Spinale FG. Matrix metalloproteinase synthesis and expression in isolated LV myocyte preparations. THE AMERICAN JOURNAL OF PHYSIOLOGY 1999; 277:H777-87. [PMID: 10444506 DOI: 10.1152/ajpheart.1999.277.2.h777] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In several cardiac disease states, alterations in myocyte and extracellular matrix (ECM) structure occur with left ventricular (LV) remodeling and are associated with changes in matrix metalloproteinase (MMP) activity. Although nonmyocyte cell types have been implicated as sites for synthesis and expression of MMPs within the ECM, whether the LV myocyte itself expresses specific types and active forms of MMPs remains unknown. Accordingly, isolated Ca(2+)-tolerant LV porcine myocytes (10(5) cells/ml) in which selective disaggregation and resuspension was performed (13 independent experiments) were plated on basement membrane substrates including Matrigel, collagen IV, laminin, and fibronectin as well as poly-L-lysine. After 24-h incubation, LV myocyte conditioned media were subjected to zymography, a specific MMP-2 proteolytic capture assay, immunoblotting, and ELISA for detection of MMP activity and relative content of the 72-kDa gelatinase MMP-2. Although robust zymographic activity [(pixels. mm(2))/cell] was observed in conditioned media from LV myocytes plated on collagen IV (1,673 +/- 297), fibronectin (1,530 +/- 281), and poly-L-lysine (2,545 +/- 560), proteolytic activity appeared to be lower in conditioned media from LV myocytes plated on Matrigel (842 +/- 83) and laminin (1,329 +/- 238). MMP-2 proteolytic activity was increased by approximately eightfold in conditioned media taken from LV myocytes plated on poly-L-lysine compared with that of Matrigel. With respect to each of the adhesion substrates, MMP-2 content was at least 50% lower in LV myocyte conditioned media taken from Matrigel and laminin. Immunofluorescent labeling of LV myocytes yielded a strong signal for MMP-2 within the myocyte and along the sarcolemmal surface. In conclusion, this study demonstrated for the first time that adult LV myocytes synthesize and express members of the MMP family and thus may potentially participate in the LV remodeling process through synthesis and secretion of MMPs.
Collapse
Affiliation(s)
- M L Coker
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, South Carolina 29425, USA
| | | | | | | | | |
Collapse
|
45
|
Bugno M, Witek B, Bereta J, Bereta M, Edwards DR, Kordula T. Reprogramming of TIMP-1 and TIMP-3 expression profiles in brain microvascular endothelial cells and astrocytes in response to proinflammatory cytokines. FEBS Lett 1999; 448:9-14. [PMID: 10217399 DOI: 10.1016/s0014-5793(99)00323-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cytokine-dependent regulation of tissue inhibitors of metalloproteinases (TIMPs) expression provides an important mechanism for controlling the activity of matrix metalloproteinases. We present data indicating that during inflammatory processes TIMP-1 and TIMP-3 may be involved in the proteolytic remodeling of subendothelial basement membrane of the brain microvascular system, a key step during leukocyte migration into the brain perivascular tissue. In brain endothelial cells the expression of TIMP-1 is dramatically up-regulated by major proinflammatory cytokines, with the combination of interleukin-1beta (IL-1beta) and tumor necrosis factor-alpha (TNF alpha) exhibiting the strongest synergistic stimulation. Simultaneously, IL-1beta/TNF alpha almost completely blocks TIMP-3 expression. Both synergistic effects are dose-dependent within the concentration range 0.05-5 ng/ml of both cytokines and correlate with the expression of inducible nitric oxide synthase, an endothelial cell activation marker. Down-regulation of TIMP-3 expression is also detected in astrocytes treated with TNF alpha or IFN-gamma whereas oncostatin M as well as TNF alpha up-regulate TIMP-1 mRNA level. We propose that the cytokine-modified balance between TIMP-1 and TIMP-3 expression provides a potential mechanism involved in the regulation of microvascular basement membrane proteolysis.
Collapse
Affiliation(s)
- M Bugno
- Institute of Molecular Biology, Jagiellonian University, Cracow, Poland.
| | | | | | | | | | | |
Collapse
|
46
|
Tyagi SC, Lewis K, Pikes D, Marcello A, Mujumdar VS, Smiley LM, Moore CK. Stretch-induced membrane type matrix metalloproteinase and tissue plasminogen activator in cardiac fibroblast cells. J Cell Physiol 1998; 176:374-82. [PMID: 9648925 DOI: 10.1002/(sici)1097-4652(199808)176:2<374::aid-jcp16>3.0.co;2-3] [Citation(s) in RCA: 71] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
In the normal heart, cardiomyocytes are surrounded by extracellular matrix (ECM) and latent matrix metalloproteinases (MMPs), which are produced primarily by cardiac fibroblasts. An activator of latent MMPs might be induced by ischemic conditions or pressure-induced stretching. To test the hypothesis that an activator of latent MMP is induced in the ischemic heart during transformation of a compensatory hypertrophic response to a decompensatory failing response in cardiac fibroblast cells, we stretched the human cardiac fibroblasts at 25 cycles/min in serum-free or 5% serum culture condition. The membrane type (MT)-MMP activity in stretched cells was measured by zymography and immuno-blot analyses using MT-MMP-2 antibody. The MT-MMP activity was further characterized by transverse-urea gradient (TUG)-zymography. The results suggested that stretch induced a membrane MMP in the fibroblasts that was similar to the MT-MMP induced in ischemic heart. Furthermore, we observed that membrane MMP has distinct mobility in TUG-zymography. To localize the MT-MMP and tissue plasminogen activator (tPA) of latent MMPs, the membrane and cytosol were separated by a method employing a detergent and sedimentation. The MT-MMP and tPA activities of cytosol and membrane fractions were measured by gelatin- and plasminogen-zymography, respectively. Differential-display mRNA analysis was performed on control and stretched cells. In situ immuno-labelling was performed to localize the MT-MMP. The results indicate that induction of MT-MMP occurred in the membrane fractions. The secretion of tPA was elevated in the stretched cells. The MT-MMP activity was inhibited by prior incubation with an antibody generated to membrane MMP. The tPA activity was inhibited by using tPA antibody. These results suggest that, under stretched conditions, neutral transmembrane matrix proteinases are induced in the cardiac fibroblasts. This may lead to activation of adverse ECM remodeling, cardiac dilatation, and failure.
Collapse
Affiliation(s)
- S C Tyagi
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson 39216-4505, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Coker ML, Spinale FG. Myocardial extracellular matrix remodeling with the development of pacing induced congestive heart failure contributory mechanisms. Cardiovasc Pathol 1998; 7:161-8. [PMID: 25851223 DOI: 10.1016/s1054-8807(97)00120-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/1997] [Indexed: 11/18/2022] Open
Abstract
The myocardial fibrillar collagens ensure structural integrity of adjoining myocytes, provide the means by which myocyte shortening is translated into overall left ventricular (LV) pump function, and have been postulated to be essential for maintaining alignment of myofibrils within the myocyte through a collagen-integrin-cytoskeletal-myofibril relation. This laboratory has performed a series of studies in order to examine the relationship between changes in myocardial collagen matrix components to LV function and geometry which occurred in a model of congestive heart failure (CHF) induced by chronic rapid pacing. In this model of CHF, indices of LV pump function are reduced and accompanied by significant dilation. LV fibrillar collagen concentration was reduced and salt extractable collagen, which reflects collagen cross-linking, was increased with the development of CHF. LV myocyte adhesion capacity to basement membrane substrates was reduced with pacing CHF. Results from a recently completed series of studies have demonstrated alterations in the expression and activity of the collagenases, or matrix metalloproteinases (MMPs) occur during the progression of CHF. Increased LV myocardial MMP abundance and activity occurred with pacing CHF and were associated with the development of LV dilation, wall thinning, and pump dysfunction. These results suggest that changes within the myocardial extracellular space are a dynamic process and accompany the LV remodeling and dysfunction which occurs with the development of a CHF process. Future studies which define the contributory role of MMP synthesis and activation in the LV remodeling process which occurs in the setting of CHF will likely identify unique therapeutic modalities to slow the progression of this disease process.
Collapse
Affiliation(s)
- M L Coker
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, South Carolina USA
| | - F G Spinale
- Division of Cardiothoracic Surgery, Medical University of South Carolina, Charleston, South Carolina USA
| |
Collapse
|
48
|
|
49
|
Tyagi SC. Homocysteine redox receptor and regulation of extracellular matrix components in vascular cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:C396-405. [PMID: 9486129 DOI: 10.1152/ajpcell.1998.274.2.c396] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Dynamic changes in the reduction-oxidation (redox) state of the tissue lead to the pathophysiological condition. Reduced homocysteine causes dysfunctions in endothelium. The proliferation of smooth muscle cells may lead to occlusive vascular disease, ischemia, and heart failure, but whether fibrosis and hypertension are a consequence of smooth muscle proliferation is unclear. Redox changes during hyper-homocyst(e)inemia may be one of the causes of premature atherosclerotic heart disease. To examine the effect of homocystine on human vascular smooth muscle cells (HVSMC), we isolated HVSMC from idiopathic dilated cardiomyopathic hearts. Coronaries in these hearts were apparently normal. HVSMC numbers in culture were measured by hemocytometer in the presence and absence of homocystine. Results show that homocystine induced cellular proliferation. This proliferation was reversed by the addition of the antioxidant N-acetylcysteine (NAC). Homocystine induces collagen expression in a dose- and time-dependent manner, as measured by Northern blot (mRNA) analysis. The 50% inhibitory concentration of 5 microM for collagen was estimated. The induction of collagen was reversed by the addition of NAC and reduced glutathione. To localize the receptor for homocystine on HVSMC, we synthesized fluorescamine-labeled homocystine conjugate. Incubation of labeled homocystine with HVSMC demonstrated membrane and cytosol localization of homocystine binding. The receptor-ligand binding was disrupted by NAC. Based on sodium dodecyl sulfate-polyacrylamide gel electrophoresis fluorography, we observed a 40- to 25-kDa homocystine redox receptor in HVSMC. Our results suggested that the redox homocysteine induces HVSMC proliferation by binding to the redox receptor and may exacerbate atherosclerotic lesion formation by inducing collagen expression.
Collapse
Affiliation(s)
- S C Tyagi
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson 39216, USA
| |
Collapse
|
50
|
Tyagi SC. Dynamic extracellular matrix remodeling in the heart failure: cardiac hypertrophy, dilatation and fibrosis. PATHOPHYSIOLOGY 1997. [DOI: 10.1016/s0928-4680(97)00019-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|