1
|
Wolosowicz M, Prokopiuk S, Kaminski TW. The Complex Role of Matrix Metalloproteinase-2 (MMP-2) in Health and Disease. Int J Mol Sci 2024; 25:13691. [PMID: 39769454 PMCID: PMC11728377 DOI: 10.3390/ijms252413691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
Matrix metalloproteinase-2 (MMP-2), a zinc-dependent enzyme, plays a critical role in the degradation and remodeling of the extracellular matrix (ECM). As a member of the gelatinase subgroup of matrix metalloproteinases, MMP-2 is involved in a variety of physiological processes, including tissue repair, wound healing, angiogenesis, and embryogenesis. It is primarily responsible for the degradation of type IV and V collagen, fibronectin, laminin, and elastin, which are essential components of the ECM. MMP-2 is secreted as an inactive pro-enzyme (proMMP-2) and activated through proteolytic cleavage, with its activity being precisely regulated by tissue inhibitors of metalloproteinases (TIMPs). Dysregulation of MMP-2 has been linked to a variety of pathological conditions, including cardiovascular diseases, diabetic complications, kidney diseases, and cancer. In cardiovascular diseases, it contributes to vascular remodeling, atherosclerosis, and aneurysms, while in fibrotic diseases, it mediates excessive ECM degradation leading to tissue scarring. In diabetes, elevated MMP-2 activity exacerbates complications such as nephropathy, retinopathy, and cardiovascular disease. In cancer, MMP-2 facilitates tumor invasion and metastasis by degrading ECM components and promoting angiogenesis. Despite its essential roles in both physiological and pathological processes, targeting MMP-2 for therapeutic purposes presents challenges due to its dual functions in tissue remodeling and repair, raising concerns about unplanned consequences such as impaired tissue healing or excessive tissue damage. These challenges underscore the need for future research to focus on developing selective modulators that can precisely balance their activity under specific disease environments. Clinical trials targeting MMP-2 modulation highlight the potential of gelatinase inhibitors, including those targeting MMP-2, to reduce tumor progression in fibrosarcoma, breast, and lung cancers. This paper reviews the structure, function, and regulation of MMP-2, its involvement in disease pathogenesis, and the potential challenges in the therapeutic implications of modulating its activity.
Collapse
Affiliation(s)
- Marta Wolosowicz
- Department of Pharmacology & Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Slawomir Prokopiuk
- Faculty of Health Sciences, University of Lomza, 14 Akademicka St., 18-400 Łomża, Poland;
| | - Tomasz W. Kaminski
- Pittsburgh Heart, Lung and Blood Vascular Medicine Institute (VMI), University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
- Thrombosis and Hemostasis Program, VERSITI Blood Research Institute, Milwaukee, WI 53226, USA
| |
Collapse
|
2
|
Garcia T, Petrera A, Hauck SM, Baber R, Wirkner K, Kirsten H, Pott J, Tönjes A, Henger S, Loeffler M, Peters A, Scholz M. Relationship of proteins and subclinical cardiovascular traits in the population-based LIFE-Adult study. Atherosclerosis 2024; 398:118613. [PMID: 39340936 DOI: 10.1016/j.atherosclerosis.2024.118613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 09/06/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
BACKGROUND AND AIMS Understanding molecular processes of the early phase of atherosclerotic cardiovascular disease conditions is of utmost importance for early prediction and intervention measures. METHODS We measured 92 cardiovascular-disease-related proteins (Olink, Cardiovascular III) in 2024 elderly participants of the population-based LIFE-Adult study. We analysed the impact of 27 covariables on these proteins including blood counts, cardiovascular risk factors and life-style-related parameters. We also analysed protein associations with 13 subclinical cardiovascular traits comprising carotid intima media thickness, plaque burden, three modes of Vicorder-based pulse-wave velocities, ankle-brachial index and ECLIA-based N-terminal prohormone of brain natriuretic peptide (NT-proBNP). RESULTS Estimated glomerular filtration rate, triglycerides and sex where the most relevant covariables explaining more than 1 % variance of 49, 22 and 20 proteins, respectively. A total of 43 proteins were significantly associated with at least one of the analysed subclinical cardiovascular traits. NT-pro-BNP, brachial-ankle pulse-wave velocity (baPWV) and parameters of carotid plaque burden accounted for the largest number of associations. Association overlaps were relatively sparse. Only growth/differentiation factor 15, low density lipoprotein receptor and interleukin-1 receptor type 2 are associated with these three different cardiovascular traits. We confirmed several literature findings and found yet unreported associations for carotid plaque presence (von-Willebrand factor, galectin 4), carotid intima-media thickness (carboxypeptidase A1 andB1), baPWV (cathepsin D) and NT-proBNP (cathepsin Z, low density lipoprotein receptor, neurogenic locus homolog protein 3, trem-like transcript 2). Sex-interaction effects were observed, e.g. for spondin-1 and growth/differentiation factor 15 likely regulated by androgen response elements. CONCLUSIONS We extend the catalogue of proteome biomarkers possibly involved in early stages of cardiovascular disease pathologies providing targets for early risk prediction or intervention strategies.
Collapse
Affiliation(s)
- Tarcyane Garcia
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Agnese Petrera
- Metabolomics and Proteomics Core, Helmholtz Zentrum Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - Stefanie M Hauck
- Metabolomics and Proteomics Core, Helmholtz Zentrum Munich - German Research Center for Environmental Health (GmbH), Neuherberg, Germany; German Center for Diabetes Research (DZD), München, Neuherberg, Germany
| | - Ronny Baber
- LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany; Institute of Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Kerstin Wirkner
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Holger Kirsten
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Janne Pott
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; MRC Biostatistics Unit, University of Cambridge, Cambridge, UK
| | - Anke Tönjes
- Department of Medicine, Division of Endocrinology and Nephrology, University of Leipzig, Leipzig, Germany
| | - Sylvia Henger
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Markus Loeffler
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany
| | - Annette Peters
- German Center for Diabetes Research (DZD), München, Neuherberg, Germany; Institute of Epidemiology, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; Institute for Medical Information Processing, Biometry and Epidemiology, Medical Faculty, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Markus Scholz
- Institute for Medical Informatics, Statistics and Epidemiology, Medical Faculty, University of Leipzig, Leipzig, Germany; LIFE Research Center for Civilization Diseases, Medical Faculty, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
3
|
Liu H, Magaye R, Kaye DM, Wang BH. Heart failure with preserved ejection fraction: The role of inflammation. Eur J Pharmacol 2024; 980:176858. [PMID: 39074526 DOI: 10.1016/j.ejphar.2024.176858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 07/15/2024] [Accepted: 07/24/2024] [Indexed: 07/31/2024]
Abstract
Heart failure (HF) is a debilitating clinical syndrome affecting 64.3 million patients worldwide. More than 50% of HF cases are attributed to HF with preserved ejection fraction (HFpEF), an entity growing in prevalence and mortality. Although recent breakthroughs reveal the prognostic benefits of sodium-glucose co-transporter 2 inhibitors (SGLT2i) in HFpEF, there is still a lack of effective pharmacological therapy available. This highlights a major gap in medical knowledge that must be addressed. Current evidence attributes HFpEF pathogenesis to an interplay between cardiometabolic comorbidities, inflammation, and renin-angiotensin-aldosterone-system (RAAS) activation, leading to cardiac remodelling and diastolic dysfunction. However, conventional RAAS blockade has demonstrated limited benefits in HFpEF, which emphasises that alternative therapeutic targets should be explored. Presently, there is limited literature examining the use of anti-inflammatory HFpEF therapies despite growing evidence supporting its importance in disease progression. Hence, this review aims to explore current perspectives on HFpEF pathogenesis, including the importance of inflammation-driven cardiac remodelling and the therapeutic potential of anti-inflammatory therapies.
Collapse
Affiliation(s)
- Hongyi Liu
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia; Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - Ruth Magaye
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - David M Kaye
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia.
| | - Bing H Wang
- Monash Alfred Baker Centre for Cardiovascular Research, School of Translational Medicine, Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, VIC, 3004, Australia; Heart Failure Research Group, Baker Heart and Diabetes Institute, Melbourne, Australia; Biomarker Discovery Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia.
| |
Collapse
|
4
|
Hartley B, Bassiouni W, Roczkowsky A, Fahlman R, Schulz R, Julien O. N-Terminomic Identification of Intracellular MMP-2 Substrates in Cardiac Tissue. J Proteome Res 2024; 23:4188-4202. [PMID: 38647137 PMCID: PMC11460328 DOI: 10.1021/acs.jproteome.3c00755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/25/2024]
Abstract
Proteases are enzymes that induce irreversible post-translational modifications by hydrolyzing amide bonds in proteins. One of these proteases is matrix metalloproteinase-2 (MMP-2), which has been shown to modulate extracellular matrix remodeling and intracellular proteolysis during myocardial injury. However, the substrates of MMP-2 in heart tissue are limited, and lesser known are the cleavage sites. Here, we used degradomics to investigate the substrates of intracellular MMP-2 in rat ventricular extracts. First, we designed a novel, constitutively active MMP-2 fusion protein (MMP-2-Fc) that we expressed and purified from mammalian cells. Using this protease, we proteolyzed ventricular extracts and used subtiligase-mediated N-terminomic labeling which identified 95 putative MMP-2-Fc proteolytic cleavage sites using mass spectrometry. The intracellular MMP-2 cleavage sites identified in heart tissue extracts were enriched for proteins primarily involved in metabolism, as well as the breakdown of fatty acids and amino acids. We further characterized the cleavage of three of these MMP-2-Fc substrates based on the gene ontology analysis. We first characterized the cleavage of sarco/endoplasmic reticulum calcium ATPase (SERCA2a), a known MMP-2 substrate in myocardial injury. We then characterized the cleavage of malate dehydrogenase (MDHM) and phosphoglycerate kinase 1 (PGK1), representing new cardiac tissue substrates. Our findings provide insights into the intracellular substrates of MMP-2 in cardiac cells, suggesting that MMP-2 activation plays a role in cardiac metabolism.
Collapse
Affiliation(s)
- Bridgette Hartley
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Wesam Bassiouni
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Andrej Roczkowsky
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
| | - Richard Fahlman
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| | - Richard Schulz
- Department
of Pharmacology, University of Alberta, Edmonton T6G 2S2, Canada
- Department
of Pediatrics, University of Alberta, Edmonton T6G 2S2, Canada
| | - Olivier Julien
- Department
of Biochemistry, University of Alberta, Edmonton T6G 2H7, Canada
| |
Collapse
|
5
|
Baudier RL, Orlandi PF, Yang W, Chen HY, Bansal N, Blackston JW, Chen J, Deo R, Dobre M, He H, He J, Ricardo AC, Shafi T, Srivastava A, Xie D, Susztak K, Feldman HI, Anderson AH. Matrix Metalloproteinase-2 and CKD Progression: The Chronic Renal Insufficiency Cohort (CRIC) Study. Kidney Med 2024; 6:100850. [PMID: 39131916 PMCID: PMC11315214 DOI: 10.1016/j.xkme.2024.100850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/13/2024] Open
Abstract
Rationale & Objective Matrix metalloproteinase 2 (MMP-2) plays an important role in the development of fibrosis, the final common pathway of chronic kidney disease (CKD). This study aimed to assess the relationship between repeated measures of MMP-2 and CKD progression in a large, diverse prospective cohort. Study Design In a prospective cohort of Chronic Renal Insufficiency Cohort (CRIC) participants (N = 3,827), MMP-2 was measured at baseline. In a case-cohort design, MMP-2 was additionally measured at year 2 in a randomly selected subcohort and cases of estimated glomerular filtration rate (eGFR) halving or kidney replacement therapy (KRT) (N = 1,439). Setting & Participants CRIC is a multicenter prospective cohort of adults with CKD. Exposure MMP-2 measured in plasma at baseline and at year 2. Outcomes A composite kidney endpoint (KRT/eGFR halving). Analytical Approach Weighted Cox proportional hazards models for case-cohort participants. Results Participants were followed for a median of 4.6 years from year 2 and 6.9 years from the baseline. Persistently elevated MMP-2 (≥300 ng/mL at both baseline and year 2) increased the hazard of the composite kidney endpoint (HR, 1.61; 95% CI, 1.07-2.42; P = 0.09) after adjusting for covariates. The relationship of persistently elevated MMP-2 was modified by levels of inflammation, with a 2.6 times higher rate of the composite kidney endpoint in those with high-sensitivity C-reactive protein < 2.5 g/dL at study entry. Heterogeneity of effect was found with proteinuria, with a baseline MMP-2 level of ≥300 ng/mL associated with an increased risk of the composite kidney endpoint (HR, 1.30; 95% CI, 1.09-1.54) only with proteinuria ≥ 442 mg/g. Limitations The observational study design limits causal interpretation. Conclusions Elevated MMP-2 is associated with CKD progression, particularly among those with low inflammation and those with proteinuria. Future investigations are warranted to confirm the reduction in risk of CKD progression among these subgroups of patients with CKD.
Collapse
Affiliation(s)
- Robin L. Baudier
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
- Biostatistics and Design Program, Oregon Health & Science University, Portland, OR
| | | | - Wei Yang
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Hsiang-Yu Chen
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Nisha Bansal
- Renal Division, University of Washington, Seattle, WA
| | | | - Jing Chen
- Department of Medicine, Tulane University, New Orleans, LA
| | - Rajat Deo
- Cardiovascular Institute, University of Pennsylvania, Philadelphia, PA
| | - Mirela Dobre
- Department of Medicine, Case Western Reserve University, Cleveland, OH
| | - Hua He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
| | - Ana C. Ricardo
- Department of Medicine, University of Illinois College of Medicine, Chicago, IL
| | - Tariq Shafi
- Division of Nephrology, Houston Methodist Hospital, Houston, TX
| | - Anand Srivastava
- Department of Medicine, University of Illinois College of Medicine, Chicago, IL
| | - Dawei Xie
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Katalin Susztak
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, PA
| | - Harold I. Feldman
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Amanda H. Anderson
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
- Department of Epidemiology, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
6
|
Kopańko M, Zabłudowska M, Pawlak D, Sieklucka B, Krupa A, Sokołowska K, Ziemińska M, Pawlak K. The Possible Effect of β-Blocker Use on the Circulating MMP-2/TIMP-2 System in Patients with Chronic Kidney Disease on Conservative Treatment. J Clin Med 2024; 13:1847. [PMID: 38610612 PMCID: PMC11012263 DOI: 10.3390/jcm13071847] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Revised: 03/16/2024] [Accepted: 03/21/2024] [Indexed: 04/14/2024] Open
Abstract
Background: The purpose of the study was to determine whether the use of β-adrenoceptor antagonists (β-blockers) can affect metalloproteinase 2 (MMP-2) and its tissue inhibitor (TIMP-2) in patients with chronic kidney disease (CKD) on conservative treatment. Methods: The circulating MMP-2/TIMP-2 system, proinflammatory cytokines (tumor necrosis factor-alpha (TNF-α) and interleukin-6 (IL-6), and the marker of oxidative stress-Cu/Zn superoxide dismutase (Cu/Zn SOD)-were measured in 23 CKD patients treated with β-blockers [β-blockers (+)] and in 27 CKD patients not receiving the above medication [β-blockers (-)]. Results: The levels of MMP-2, TIMP-2, and IL-6 were significantly lower in the β-blockers (+) than in the β-blockers (-) group, whereas Cu/Zn SOD concentrations were not affected by β-blocker use. There was a strong, independent association between MMP-2 and TIMP-2 in both analyzed patient groups. In the β-blockers (+) group, MMP-2 levels were indirectly related to the signs of inflammation, whereas in the β-blockers (-) group, the alterations in the MMP-2/TIMP-2 system were associated with the oxidative stress marker and CKD etiology. Conclusions: This study is the first to suggest that the use of β-blockers was associated with the reduction in IL-6 and the MMP-2/TIMP-2 system in CKD, providing a pharmacological rationale for the use of β-blockers to reduce inflammation and abnormal vascular remodeling in CKD.
Collapse
Affiliation(s)
- Magdalena Kopańko
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (M.K.); (M.Z.); (B.S.); (K.S.); (M.Z.)
| | - Magdalena Zabłudowska
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (M.K.); (M.Z.); (B.S.); (K.S.); (M.Z.)
| | - Dariusz Pawlak
- Department of Pharmacodynamics, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland;
| | - Beata Sieklucka
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (M.K.); (M.Z.); (B.S.); (K.S.); (M.Z.)
| | - Anna Krupa
- Department of Internal Medicine and Metabolic Diseases, Medical University of Bialystok, M. Sklodowskiej-Curie 24A, 15-276 Bialystok, Poland;
| | - Katarzyna Sokołowska
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (M.K.); (M.Z.); (B.S.); (K.S.); (M.Z.)
| | - Marta Ziemińska
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (M.K.); (M.Z.); (B.S.); (K.S.); (M.Z.)
| | - Krystyna Pawlak
- Department of Monitored Pharmacotherapy, Medical University of Bialystok, Mickiewicza 2C, 15-222 Bialystok, Poland; (M.K.); (M.Z.); (B.S.); (K.S.); (M.Z.)
| |
Collapse
|
7
|
Negrin LL, Carlin GL, Ristl R, Hajdu S. Serum levels of matrix metalloproteinases 1, 2, and 7, and their tissue inhibitors 1, 2, 3, and 4 in polytraumatized patients: Time trajectories, correlations, and their ability to predict mortality. PLoS One 2024; 19:e0300258. [PMID: 38457458 PMCID: PMC10923431 DOI: 10.1371/journal.pone.0300258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/23/2024] [Indexed: 03/10/2024] Open
Abstract
There has been limited research on assessing metalloproteinases (MMPs) 1, 2, and 7, as well as their tissue inhibitors (TIMPs) 1, 2, 3, and 4 in the context of polytrauma. These proteins play crucial roles in various physiological and pathological processes and could be a reliable tool in polytrauma care. We aimed to determine their clinical relevance. We assessed 24 blunt polytrauma survivors and 12 fatalities (mean age, 44.2 years, mean ISS, 45) who were directly admitted to our Level I trauma center and spent at least one night in the intensive care unit. We measured serum levels of the selected proteins on admission (day 0) and days 1, 3, 5, 7, and 10. The serum levels of the seven proteins varied considerably among individuals, resulting in similar median trend curves for TIMP1 and TIMP4 and for MMP1, MMP2, TIMP2, and TIMP3. We also found a significant interrelationship between the MMP2, TIMP2, and TIMP3 levels at the same measurement points. Furthermore, we calculated significant cross-correlations between MMP7 and MMP1, TIMP1 and MMP7, TIMP3 and MMP1, TIMP3 and MMP2, and TIMP4 and TIMP3 and an almost significant correlation between MMP7 and TIMP1 for a two-day-lag. The autocorrelation coefficient reached statistical significance for MMP1 and TIMP3. Finally, lower TIMP1 serum levels were associated with in-hospital mortality upon admission. The causal effects and interrelationships between selected proteins might provide new insights into the interactions of MMPs and TIMPs. Identifying the underlying causes might help develop personalized therapies for patients with multiple injuries. Administering recombinant TIMP1 or increasing endogenous production could improve outcomes for those with multiple injuries. However, before justifying further investigations into basic research and clinical relevance, our findings must be validated in a multicenter study using independent cohorts to account for clinical and biological variability.
Collapse
Affiliation(s)
- Lukas L. Negrin
- University Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| | - Greta L. Carlin
- University Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
- University Department of Obstetrics and Gynecology, Medical University of Vienna, Vienna, Austria
| | - Robin Ristl
- Center for Medical Statistics, Informatics and Intelligent Systems, Medical University of Vienna, Vienna, Austria
| | - Stefan Hajdu
- University Department of Orthopedics and Trauma Surgery, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
8
|
Jamshidi S, Rostami A, Shojaei S, Taherkhani A, Taherkhani H. Exploring natural anthraquinones as potential MMP2 inhibitors: A computational study. Biosystems 2024; 235:105103. [PMID: 38123060 DOI: 10.1016/j.biosystems.2023.105103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 11/23/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
OBJECTIVE Matrix metalloproteinase-2 (MMP2) plays a significant role in cleaving extracellular matrix components, leading to many cancer cells' progression and invasion behavior. Therefore, MMP2 inhibition may hold promise for cancer treatment. Anthraquinones have shown antineoplastic effects, some of which have been used in clinical practice as anticancer drugs. This study used a computational drug discovery approach to assess the possible inhibitory effects of selected anthraquinones on MMP2. The results were then compared with that of Captopril, which was considered a standard drug. METHODS This study used the AutoDock 4.0 tool to evaluate the binding affinity of 21 anthraquinones to the MMP2 catalytic domain. The most favorable scores based on the Gibbs free binding energy scores were given to the highest-ranked ligands. The Discovery Studio Visualizer tool illustrated interactions between MMP2 residues and top-ranked anthraquinones. RESULTS A total of 12 anthraquinones were identified with ΔGbinding scores less than - 10 kcal/mol. Pulmatin (Chrysophanol-8-glucoside) was the most potent MMP2 inhibitor, with a ΔGbinding score of - 12.91 kcal/mol. This anthraquinone was able to restrict MMP2 activity within a picomolar range. CONCLUSION MMP2 inhibition by anthraquinones, notably Pulmatin, may be a useful therapeutic approach for cancer treatment.
Collapse
Affiliation(s)
- Shokoofeh Jamshidi
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Ali Rostami
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Setareh Shojaei
- Department of Oral and Maxillofacial Pathology, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Amir Taherkhani
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran.
| | - Heshmatollah Taherkhani
- Department of Paramedical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
9
|
Saremi L, Esmaeili S, Ghaffari ME, Shahbazi S, Lotfipanah S, Kadkhodazadeh M. Evaluation of matrix metalloproteinase-1, -2, -3, -7, and -13 gene polymorphisms in patients with chronic periodontitis and healthy controls. Clin Oral Investig 2023; 27:7417-7423. [PMID: 37848583 DOI: 10.1007/s00784-023-05331-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
OBJECTIVES The current study aimed to investigate the association of matrix metalloproteinase- (MMP-) 1, -2, -3, -7, and -13 gene polymorphisms with chronic periodontitis (CP) in an Iranian population. MATERIALS AND METHODS In this case-control study, 87 subjects with CP and 89 periodontally healthy subjects were allocated to case and control groups, respectively. Subjects' venous blood samples (5 cc) were collected, and DNA extraction was performed. A spectrophotometer was utilized to assess the concentration of extracted DNAs. The desired gene polymorphisms were examined using restriction fragment length polymorphism polymerase chain reaction (RFLP-PCR) followed by electrophoresis. Statistical analyses were done using the Pearson Chi-Square test, odds ratio, and t-Test using SPSS Version 28. RESULTS The MMP-1 (-1607 1G/2G) rs1799750, MMP-3 (-1171 5A/6A) rs3025058, and MMP-7 (-181 A/G) rs11568818 gene polymorphisms significantly differed between case and control groups (PV = 0.019, 0.007, and 0.028, respectively). In contrast, the gene polymorphisms of MMP-2 (-1306 C/T) rs243865 and MMP-13 (-77 A/G) rs2252070 did not make a significant difference. Regarding allele frequencies, the presence of the 2G allele in the MMP-1 (-1607) rs1799750 genotype increased the CP susceptibility significantly, while subjects with the 6A allele in their MMP-3 (-1171) rs3025058 genotype showed significantly lower susceptibility to CP (PV = 0.008 and < 0.001, respectively). CONCLUSION In the studied population, gene polymorphisms in the DNA sequences of MMP-1 (-1607 1G/2G) rs1799750, MMP-3 (-1171 5A/6A) rs3025058, and MMP-7 (-181 A/G) rs11568818 may have impacts on CP incidence. CLINICAL RELEVANCE Clinicians should be cautious about the association between MMP-1, MMP-3, and MMP-7 gene polymorphisms and the incidence of chronic periodontitis during periodontal treatment planning.
Collapse
Affiliation(s)
- Leila Saremi
- Dental Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Saharnaz Esmaeili
- Division of Oral and Maxillofacial Surgery, School of Dentistry, University of California, Los Angeles, Los Angeles, CA, USA
- Dentofacial Deformities Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Ebrahim Ghaffari
- Department of Epidemiology and Biostatistics, Faculty of Health, Qom University of Medical Sciences, Qom, Iran
| | - Soheil Shahbazi
- Dentofacial Deformities Research Center, Research Institute of Dental Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Lotfipanah
- Department of Biology Education, Farhangian University, Tehran, Iran
| | - Mahdi Kadkhodazadeh
- Dental Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
10
|
Yin Y, Zhang H, Sun L, Han Q, Zheng M, Chen H, Fei S, Tan R, Ju X, Wang Z, Gu M. Association between fibrosis-related gene polymorphism and long-term allograft outcome in renal transplant recipients. BMC Med Genomics 2023; 16:255. [PMID: 37867197 PMCID: PMC10591404 DOI: 10.1186/s12920-023-01686-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 10/03/2023] [Indexed: 10/24/2023] Open
Abstract
BACKGROUND Renal allograft fibrosis is one of characteristic causes of long-term renal function loss. The purpose of our study is to investigate the association between fibrosis-related genes single nucleotide polymorphism (SNPs) and kidney function in 5 years after kidney transplantation. METHODS A total of 143 recipients were eligible for screening with 5-year follow-up information and SNP sequencing information from blood samples were included in this study. Minor Allele Frequency (MAF) and Hardy-Weinberg Equilibrium (HWE) analysis was conducted to identify tagger single-nucleotide polymorphisms (SNPs) and haplotypes. SNPs associated with the fifth year chronic kidney disease (CKD) staging were screened by SPSS and the "SNPassoc" package in RStudio and used for subsequent prediction model construction. RESULTS A total of 275 renal transplant-related SNPs identified after target sequencing analysis. 64 Tagger SNPs were selected, and two SNPs (rs13969 and rs243849) were statistically significant for stage of CKD in 5 years. Finally, a model based on Gender, Age, rs1396, and rs243849 was constructed by multivariate linear regression analysis. Additionally, this model has a good performance in predicting uremia five years after kidney transplantation. CONCLUSION Two SNPs (rs13969 and rs243849) were identified to be significantly associated with long-term renal allograft function. Based on this, a prediction model for long-term allograft function was established containing Gender, Age, rs1396, and rs243849. However, an independent cohort should be enrolled to validate the predicting performance.
Collapse
Affiliation(s)
- Yu Yin
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Han Zhang
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Li Sun
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qianguang Han
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ming Zheng
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hao Chen
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Shuang Fei
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Ruoyun Tan
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiaobing Ju
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Zijie Wang
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| | - Min Gu
- Department of Urology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
- Department of Urology, The Second Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
11
|
Saravi SSS, Bonetti NR, Vukolic A, Vdovenko D, Lee P, Liberale L, Basso C, Rizzo S, Akhmedov A, Lüscher TF, Camici GG, Beer JH. Long-term dietary n3 fatty acid prevents aging-related cardiac diastolic and vascular dysfunction. Vascul Pharmacol 2023; 150:107175. [PMID: 37105373 DOI: 10.1016/j.vph.2023.107175] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 04/15/2023] [Accepted: 04/24/2023] [Indexed: 04/29/2023]
Abstract
AIMS The prevalence of left ventricular (LV) diastolic and vascular dysfunction increases with age, eventually leading to heart failure with preserved ejection fraction (HFpEF). A preventive strategy is an unmet medical need. We and others reported previously on the beneficial effects of omega-3 fatty acid alpha linolenic acid (ALA) on cardiovascular disorders in animal models and translational studies. We now investigate whether long-term dietary ALA could prevent LV diastolic dysfunction and vascular aging in a murine model. METHODS AND RESULTS Wild-type C57BL/6 J mice were fed a chow or ALA diet for 12 months, starting at 6 months of age. Here, we show that aged (~18 months) mice recapitulate major hallmarks of HFpEF, including LV diastolic dysfunction with preserved ejection fraction, impaired vascular function, cardiac fibrosis, arterial stiffening and inflammation, as well as elevated B-type natriuretic peptide (BNP). Long-term ALA supplementation upregulated the mitochondrial tricarboxylic acid enzyme Idh2 and the antioxidant enzymes SOD1 and Gpx1. It also has been associated with reduced inflammation and ECM remodeling, accompanied by a significant downregulation of fibrosis biomarkers MMP-2 and TGF-β in both cardiac and vascular tissues obtained from aged mice. Our data exhibited the preventive effects of dietary ALA against LV diastolic dysfunction, impaired vasorelaxation, cardiac fibrosis, inflammation and arterial stiffening in aged mice. CONCLUSIONS We provide evidence and a simplified mechanistic insight on how long-term ALA supplementation is a successful strategy to prevent the development of age-related diastolic and vascular dysfunction.
Collapse
Affiliation(s)
- Seyed Soheil Saeedi Saravi
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, 8952 Schlieren, Switzerland
| | - Nicole R Bonetti
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, 5404 Baden, Switzerland
| | - Ana Vukolic
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Daria Vdovenko
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Pratintip Lee
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, 5404 Baden, Switzerland
| | - Luca Liberale
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, Genoa, Italy
| | - Cristina Basso
- Cardiovascular Pathology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Stefania Rizzo
- Cardiovascular Pathology Unit, Department of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, 35128 Padova, Italy
| | - Alexander Akhmedov
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland; Royal Brompton and Harefield Hospitals, Imperial and Kings College, London, UK
| | - Giovanni G Camici
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland; University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland; Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Jürg H Beer
- Center for Molecular Cardiology, University of Zurich, 8952 Schlieren, Switzerland; Department of Internal Medicine, Cantonal Hospital Baden, 5404 Baden, Switzerland.
| |
Collapse
|
12
|
Welcome MO, Dogo D, Nikos E Mastorakis. Cellular mechanisms and molecular pathways linking bitter taste receptor signalling to cardiac inflammation, oxidative stress, arrhythmia and contractile dysfunction in heart diseases. Inflammopharmacology 2023; 31:89-117. [PMID: 36471190 PMCID: PMC9734786 DOI: 10.1007/s10787-022-01086-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/11/2022] [Indexed: 12/12/2022]
Abstract
Heart diseases and related complications constitute a leading cause of death and socioeconomic threat worldwide. Despite intense efforts and research on the pathogenetic mechanisms of these diseases, the underlying cellular and molecular mechanisms are yet to be completely understood. Several lines of evidence indicate a critical role of inflammatory and oxidative stress responses in the development and progression of heart diseases. Nevertheless, the molecular machinery that drives cardiac inflammation and oxidative stress is not completely known. Recent data suggest an important role of cardiac bitter taste receptors (TAS2Rs) in the pathogenetic mechanism of heart diseases. Independent groups of researchers have demonstrated a central role of TAS2Rs in mediating inflammatory, oxidative stress responses, autophagy, impulse generation/propagation and contractile activities in the heart, suggesting that dysfunctional TAS2R signalling may predispose to cardiac inflammatory and oxidative stress disorders, characterised by contractile dysfunction and arrhythmia. Moreover, cardiac TAS2Rs act as gateway surveillance units that monitor and detect toxigenic or pathogenic molecules, including microbial components, and initiate responses that ultimately culminate in protection of the host against the aggression. Unfortunately, however, the molecular mechanisms that link TAS2R sensing of the cardiac milieu to inflammatory and oxidative stress responses are not clearly known. Therefore, we sought to review the possible role of TAS2R signalling in the pathophysiology of cardiac inflammation, oxidative stress, arrhythmia and contractile dysfunction in heart diseases. Potential therapeutic significance of targeting TAS2R or its downstream signalling molecules in cardiac inflammation, oxidative stress, arrhythmia and contractile dysfunction is also discussed.
Collapse
Affiliation(s)
- Menizibeya O Welcome
- Department of Physiology, Faculty of Basic Medical Sciences, College of Health Sciences, Nile University of Nigeria, Plot 681 Cadastral Zone, C-00 Research and Institution Area, Jabi Airport Road Bypass, FCT, Abuja, Nigeria.
| | - Dilli Dogo
- Department of Surgery, Faculty of Clinical Sciences, College of Health Sciences, Nile University of Nigeria, Abuja, Nigeria
| | - Nikos E Mastorakis
- Technical University of Sofia, Klement Ohridksi 8, Sofia, 1000, Bulgaria
| |
Collapse
|
13
|
Gonçalves PR, Nascimento LD, Gerlach RF, Rodrigues KE, Prado AF. Matrix Metalloproteinase 2 as a Pharmacological Target in Heart Failure. Pharmaceuticals (Basel) 2022; 15:ph15080920. [PMID: 35893744 PMCID: PMC9331741 DOI: 10.3390/ph15080920] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/11/2022] [Accepted: 07/11/2022] [Indexed: 12/18/2022] Open
Abstract
Heart failure (HF) is an acute or chronic clinical syndrome that results in a decrease in cardiac output and an increase in intracardiac pressure at rest or upon exertion. The pathophysiology of HF is heterogeneous and results from an initial harmful event in the heart that promotes neurohormonal changes such as autonomic dysfunction and activation of the renin-angiotensin-aldosterone system, endothelial dysfunction, and inflammation. Cardiac remodeling occurs, which is associated with degradation and disorganized synthesis of extracellular matrix (ECM) components that are controlled by ECM metalloproteinases (MMPs). MMP-2 is part of this group of proteases, which are classified as gelatinases and are constituents of the heart. MMP-2 is considered a biomarker of patients with HF with reduced ejection fraction (HFrEF) or preserved ejection fraction (HFpEF). The role of MMP-2 in the development of cardiac injury and dysfunction has clearly been demonstrated in animal models of cardiac ischemia, transgenic models that overexpress MMP-2, and knockout models for this protease. New research to minimize cardiac structural and functional alterations using non-selective and selective inhibitors for MMP-2 demonstrates that this protease could be used as a possible pharmacological target in the treatment of HF.
Collapse
Affiliation(s)
- Pricila Rodrigues Gonçalves
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
| | - Lisandra Duarte Nascimento
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
| | - Raquel Fernanda Gerlach
- Department of Basic and Oral Biology, Faculty of Dentistry of Ribeirao Preto, University of Sao Paulo (FORP/USP), Ribeirao Preto 14040-904, SP, Brazil;
| | - Keuri Eleutério Rodrigues
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
| | - Alejandro Ferraz Prado
- Cardiovascular System Pharmacology and Toxicology Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (P.R.G.); (L.D.N.); (K.E.R.)
- Correspondence:
| |
Collapse
|
14
|
Sanhueza-Olivares F, Troncoso MF, Pino-de la Fuente F, Martinez-Bilbao J, Riquelme JA, Norambuena-Soto I, Villa M, Lavandero S, Castro PF, Chiong M. A potential role of autophagy-mediated vascular senescence in the pathophysiology of HFpEF. Front Endocrinol (Lausanne) 2022; 13:1057349. [PMID: 36465616 PMCID: PMC9713703 DOI: 10.3389/fendo.2022.1057349] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 10/26/2022] [Indexed: 11/18/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is one of the most complex and most prevalent cardiometabolic diseases in aging population. Age, obesity, diabetes, and hypertension are the main comorbidities of HFpEF. Microvascular dysfunction and vascular remodeling play a major role in its development. Among the many mechanisms involved in this process, vascular stiffening has been described as one the most prevalent during HFpEF, leading to ventricular-vascular uncoupling and mismatches in aged HFpEF patients. Aged blood vessels display an increased number of senescent endothelial cells (ECs) and vascular smooth muscle cells (VSMCs). This is consistent with the fact that EC and cardiomyocyte cell senescence has been reported during HFpEF. Autophagy plays a major role in VSMCs physiology, regulating phenotypic switch between contractile and synthetic phenotypes. It has also been described that autophagy can regulate arterial stiffening and EC and VSMC senescence. Many studies now support the notion that targeting autophagy would help with the treatment of many cardiovascular and metabolic diseases. In this review, we discuss the mechanisms involved in autophagy-mediated vascular senescence and whether this could be a driver in the development and progression of HFpEF.
Collapse
Affiliation(s)
- Fernanda Sanhueza-Olivares
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Mayarling F. Troncoso
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Francisco Pino-de la Fuente
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Javiera Martinez-Bilbao
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Jaime A. Riquelme
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Ignacio Norambuena-Soto
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Monica Villa
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Pablo F. Castro
- Advanced Center for Chronic Diseases, Faculty of Medicine, Pontifical University Catholic of Chile, Santiago, Chile
| | - Mario Chiong
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences, University of Chile, Santiago, Chile
- *Correspondence: Mario Chiong,
| |
Collapse
|
15
|
Kim DK, Lee YH, Kim JS, Kim YG, Lee SY, Ahn SY, Lee DY, Jeong KH, Lee SH, Hwang HS, Moon JY. Circulating Vascular Adhesion Protein-1 Level Predicts the Risk of Cardiovascular Events and Mortality in Hemodialysis Patients. Front Cardiovasc Med 2021; 8:701079. [PMID: 34557529 PMCID: PMC8452851 DOI: 10.3389/fcvm.2021.701079] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Accepted: 08/13/2021] [Indexed: 12/24/2022] Open
Abstract
Background: Vascular adhesion protein-1 (VAP-1) is an oxidative enzyme of primary amines that facilitates the transmigration of inflammatory cells. Its oxidative and inflammatory effects are prominently increased in pathological conditions, such as metabolic, atherosclerotic, and cardiac diseases. However, the clinical significance of circulating VAP-1 levels in hemodialysis (HD) patients is unclear. Methods: A total of 434 HD patients were enrolled in a prospective multicenter cohort study between June 2016 and April 2019. Plasma VAP-1 levels were measured at the time of data entry, and the primary endpoint was defined as a composite of cardiovascular (CV) and cardiac events. Results: Circulating VAP-1 levels were positively correlated with plasma levels of cardiac remodeling markers, including brain natriuretic peptide, galectin-3, and matrix metalloproteinase-2. Multivariable logistic regression analysis revealed that patients with higher circulating VAP-1 levels were more likely to have left ventricular diastolic dysfunction [odds ratio, 1.40; 95% confidence interval [CI], 1.04–1.88]. The cumulative event rate of the composite of CV events was significantly greater in VAP-1 tertile 3 than in VAP-1 tertiles 1 and 2 (P = 0.009). Patients in tertile 3 were also associated with an increased cumulative event rate of cardiac events (P = 0.015), with a 2.06-fold higher risk each for CV (95% CI, 1.10–3.85) and cardiac (95% CI, 1.03–4.12) events after adjusting for multiple variables. Conclusions: Plasma VAP-1 levels were positively associated with left ventricular diastolic dysfunction and the risk of incident CV and cardiac events in HD patients. Our results indicate that VAP-1 may aid clinicians in identifying HD patients at a high risk of CV events.
Collapse
Affiliation(s)
- Dae Kyu Kim
- Department of Medicine, Graduate School, Kyung Hee University, Seoul, South Korea
| | - Yu Ho Lee
- Division of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Jin Sug Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea
| | - Yang Gyun Kim
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea
| | - So-Young Lee
- Division of Nephrology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam, South Korea
| | - Shin Young Ahn
- Division of Nephrology, Department of Internal Medicine, Korea University College of Medicine, Seoul, South Korea
| | - Dong-Young Lee
- Division of Nephrology, Department of Internal Medicine, Veterans Health Service Medical Center, Seoul, South Korea
| | - Kyung Hwan Jeong
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea
| | - Sang-Ho Lee
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea
| | - Hyeon Seok Hwang
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea
| | - Ju-Young Moon
- Division of Nephrology, Department of Internal Medicine, Kyung Hee University, Seoul, South Korea
| |
Collapse
|
16
|
Kaur N, Guan Y, Raja R, Ruiz-Velasco A, Liu W. Mechanisms and Therapeutic Prospects of Diabetic Cardiomyopathy Through the Inflammatory Response. Front Physiol 2021; 12:694864. [PMID: 34234695 PMCID: PMC8257042 DOI: 10.3389/fphys.2021.694864] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 05/10/2021] [Indexed: 12/14/2022] Open
Abstract
The incidence of heart failure (HF) continues to increase rapidly in patients with diabetes. It is marked by myocardial remodeling, including fibrosis, hypertrophy, and cell death, leading to diastolic dysfunction with or without systolic dysfunction. Diabetic cardiomyopathy (DCM) is a distinct myocardial disease in the absence of coronary artery disease. DCM is partially induced by chronic systemic inflammation, underpinned by a hostile environment due to hyperglycemia, hyperlipidemia, hyperinsulinemia, and insulin resistance. The detrimental role of leukocytes, cytokines, and chemokines is evident in the diabetic heart, yet the precise role of inflammation as a cause or consequence of DCM remains incompletely understood. Here, we provide a concise review of the inflammatory signaling mechanisms contributing to the clinical complications of diabetes-associated HF. Overall, the impact of inflammation on the onset and development of DCM suggests the potential benefits of targeting inflammatory cascades to prevent DCM. This review is tailored to outline the known effects of the current anti-diabetic drugs, anti-inflammatory therapies, and natural compounds on inflammation, which mitigate HF progression in diabetic populations.
Collapse
Affiliation(s)
| | | | | | | | - Wei Liu
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine, and Health, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
17
|
Diaz MD, Tran E, Spang M, Wang R, Gaetani R, Luo CG, Braden R, Hill RC, Hansen KC, DeMaria AN, Christman KL. Injectable Myocardial Matrix Hydrogel Mitigates Negative Left Ventricular Remodeling in a Chronic Myocardial Infarction Model. JACC Basic Transl Sci 2021; 6:350-361. [PMID: 33997521 PMCID: PMC8093531 DOI: 10.1016/j.jacbts.2021.01.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 01/04/2021] [Accepted: 01/04/2021] [Indexed: 12/12/2022]
Abstract
Myocardial matrix hydrogel preserves LV volumes and apical wall thickening in a chronic MI model. Myocardial matrix hydrogel trends toward reduced fibrosis. In vivo differential gene expression analysis shows the matrix modulates cardiac muscle contraction, metabolism, fibrosis, and the inflammatory/immune response in a chronic MI model.
A first-in-man clinical study on a myocardial-derived decellularized extracellular matrix hydrogel suggested the potential for efficacy in chronic myocardial infarction (MI) patients. However, little is understood about the mechanism of action in chronic MI. In this study, the authors investigated the efficacy and mechanism by which the myocardial matrix hydrogel can mitigate negative left ventricular (LV) remodeling in a rat chronic MI model. Assessment of cardiac function via magnetic resonance imaging demonstrated preservation of LV volumes and apical wall thickening. Differential gene expression analyses showed the matrix is able to prevent further negative LV remodeling in the chronic MI model through modulation of the immune response, down-regulation of pathways involved in heart failure progression and fibrosis, and up-regulation of genes important for cardiac muscle contraction.
Collapse
Key Words
- CMR, cardiac magnetic resonance
- ECM, extracellular matrix
- EDV, end-diastolic volume
- EF, ejection fraction
- ESV, end-systolic volume
- HF, heart failure
- IHC, immunohistochemistry
- KEGG, Kyoto Encyclopedia of Genes and Genomes
- LV, left ventricular
- MI, myocardial infarction
- MS, mass spectrometry
- QconCAT, quantitative concatamer
- biomaterials
- chronic inflammation
- chronic myocardial infarction
- gene expression
Collapse
Affiliation(s)
- Miranda D Diaz
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| | - Elaine Tran
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| | - Martin Spang
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| | - Raymond Wang
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| | - Roberto Gaetani
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA.,Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Colin G Luo
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| | - Rebecca Braden
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| | - Ryan C Hill
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado, Aurora, Colorado, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, Anschutz Medical Campus, University of Colorado, Aurora, Colorado, USA
| | - Anthony N DeMaria
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Karen L Christman
- Department of Bioengineering, University of California, San Diego, La Jolla, California, USA.,Sanford Consortium for Regenerative Medicine, La Jolla, California, USA
| |
Collapse
|
18
|
Zhang YX, Tang RN, Wang LT, Liu BC. Role of crosstalk between endothelial cells and smooth muscle cells in vascular calcification in chronic kidney disease. Cell Prolif 2021; 54:e12980. [PMID: 33502070 PMCID: PMC7941222 DOI: 10.1111/cpr.12980] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 09/29/2020] [Accepted: 12/22/2020] [Indexed: 02/06/2023] Open
Abstract
Chronic kidney disease (CKD) is a severe health problem worldwide, and vascular calcification (VC) contributes substantially to the cardiovascular morbidity and high mortality of CKD. CKD is often accompanied by a variety of pathophysiological states, such as inflammation, oxidative stress, hyperglycaemia, hyperparathyroidism and haemodynamic derangement, that can cause injuries to smooth muscle cells (SMCs) and endothelial cells (ECs) to promote VC. Similar to SMCs, whose role has been widely explored in VC, ECs may contribute to VC via osteochondral transdifferentiation, apoptosis, etc. In addition, given their location in the innermost layer of the blood vessel lumen and preferential reception of various pro‐calcification stimuli, ECs can pass messages to vascular wall cells and communicate with them. Crosstalk between ECs and SMCs via cytokines through a paracrine mechanism, extracellular vesicles, miRNAs and myoendothelial gap junctions also plays a role in VC. In this review, we emphasize the role of intercellular crosstalk between ECs and SMCs in VC associated with CKD.
Collapse
Affiliation(s)
- Yu-Xia Zhang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.,Institute of Nephrology, Zhongda Hospital, Nanjing Lishui People' Hospital, Nanjing, China
| | - Ri-Ning Tang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.,Institute of Nephrology, Zhongda Hospital, Nanjing Lishui People' Hospital, Nanjing, China
| | - Li-Ting Wang
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.,Institute of Nephrology, Zhongda Hospital, Nanjing Lishui People' Hospital, Nanjing, China
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.,Institute of Nephrology, Zhongda Hospital, Nanjing Lishui People' Hospital, Nanjing, China
| |
Collapse
|
19
|
Ben Braiek A, Chahed H, Dumont F, Abdelhak F, Hichem D, Gamra H, Baudin B. Identification of biomarker panels as predictors of severity in coronary artery disease. J Cell Mol Med 2020; 25:1518-1530. [PMID: 33381894 PMCID: PMC7875935 DOI: 10.1111/jcmm.16244] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/23/2020] [Accepted: 11/26/2020] [Indexed: 02/06/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are implicated in atherosclerotic plaque rupture and recondition. Specific tissue inhibitors (TIMPs) control MMP functions. Both MMPs and TIMPs are potential biomarkers of plaque instability. Elevated Apo‐CII and CIII and Apo‐E levels are recognized as cardiovascular disease risk factors. We aimed to establish the best blood biomarker panel to evaluate the coronary artery disease (CAD) severity. Plasma levels of MMP‐3 and MMP‐9, TIMP‐1 and TIMP‐2, Apo‐CII, Apo‐CIII and Apo‐E were measured in 472 patients with CAD evaluated by coronary angiography and electrocardiography, and in 285 healthy controls. MMP‐3 and MMP‐9 plasma levels in CAD patients were significantly increased (P < 0.001) compared to controls (3.54‐ and 3.81‐fold, respectively). Furthermore, these increments are modulated by CAD severity as well as for Apo‐CII and Apo‐CIII levels (P < 0.001). TIMPs levels were decreased in CAD versus controls (P < 0.001) and in inverse correlation to MMPs. Standard ROC curve approach showed the importance of panels of biomarkers, including MMP‐3, MMP‐9, TIMP‐1, TIMP‐2, Apo‐CII and Apo‐CIII, for disease aggravation diagnosis. A high area under curve (AUC) value (0.995) was reached for the association of MMP‐9, TIMP‐2 and Apo‐CIII. The unbalance between MMPs and TIMPs in vascular wall and dyslipidaemia creates favourable conditions for plaque disruption. Our study suggests that the combination of MMP‐9, TIMP‐2 and Apo‐CIII values (‘CAD aggravation panel’) characterizes the severity of CAD, that is electrophysiological state, number of involved vessels, stent disposal and type of stent.
Collapse
Affiliation(s)
- Assia Ben Braiek
- Service de Biochimie, DMU BioGem, Hôpital Saint-Antoine, Paris, Sorbonne Université, Paris, France.,Molecular Biology Departments, Faculty of Pharmacy, Monastir University, Monastir, Tunisia.,UMS IPSIT - UFR Pharmacie, Université Paris-Saclay, Chatenay-Malabry, France
| | - Hinda Chahed
- Molecular Biology Departments, Faculty of Pharmacy, Monastir University, Monastir, Tunisia
| | - Florent Dumont
- UMS IPSIT - UFR Pharmacie, Université Paris-Saclay, Chatenay-Malabry, France
| | - Fodha Abdelhak
- Department of Cardiology, Fattouma Bourguiba Hospital, Monastir, Tunisia.,Department of Cardiology, Farhat Hached Hospital, Sousse, Tunisia
| | - Denguir Hichem
- Department of Cardiology, Fattouma Bourguiba Hospital, Monastir, Tunisia.,The Regional Hospital Mohamed Ben Sassi, Gabes, Tunisia
| | - Habib Gamra
- Department of Cardiology, Fattouma Bourguiba Hospital, Monastir, Tunisia
| | - Bruno Baudin
- Service de Biochimie, DMU BioGem, Hôpital Saint-Antoine, Paris, Sorbonne Université, Paris, France.,INSERM UMR 1193 - UFR Pharmacie, Université Paris-Saclay, Châtenay-Malabry, France
| |
Collapse
|
20
|
Cabral-Pacheco GA, Garza-Veloz I, Castruita-De la Rosa C, Ramirez-Acuña JM, Perez-Romero BA, Guerrero-Rodriguez JF, Martinez-Avila N, Martinez-Fierro ML. The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases. Int J Mol Sci 2020; 21:E9739. [PMID: 33419373 PMCID: PMC7767220 DOI: 10.3390/ijms21249739] [Citation(s) in RCA: 733] [Impact Index Per Article: 146.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/10/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-dependent extracellular matrix (ECM) remodeling endopeptidases that have the capacity to degrade almost every component of the ECM. The degradation of the ECM is of great importance, since it is related to embryonic development and angiogenesis. It is also involved in cell repair and the remodeling of tissues. When the expression of MMPs is altered, it can generate the abnormal degradation of the ECM. This is the initial cause of the development of chronic degenerative diseases and vascular complications generated by diabetes. In addition, this process has an association with neurodegeneration and cancer progression. Within the ECM, the tissue inhibitors of MMPs (TIMPs) inhibit the proteolytic activity of MMPs. TIMPs are important regulators of ECM turnover, tissue remodeling, and cellular behavior. Therefore, TIMPs (similar to MMPs) modulate angiogenesis, cell proliferation, and apoptosis. An interruption in the balance between MMPs and TIMPs has been implicated in the pathophysiology and progression of several diseases. This review focuses on the participation of both MMPs (e.g., MMP-2 and MMP-9) and TIMPs (e.g., TIMP-1 and TIMP-3) in physiological processes and on how their abnormal regulation is associated with human diseases. The inclusion of current strategies and mechanisms of MMP inhibition in the development of new therapies targeting MMPs was also considered.
Collapse
Affiliation(s)
| | - Idalia Garza-Veloz
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y Ciencias de la Salud, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (G.AC.-P.); (C.C.-D.l.R.); (J.MR.-A.); (B.AP.-R.); (J.FG.-R.); (N.M.-A.)
| | | | | | | | | | | | - Margarita L Martinez-Fierro
- Molecular Medicine Laboratory, Unidad Académica de Medicina Humana y Ciencias de la Salud, Carretera Zacatecas-Guadalajara Km.6. Ejido la Escondida, Zacatecas 98160, Mexico; (G.AC.-P.); (C.C.-D.l.R.); (J.MR.-A.); (B.AP.-R.); (J.FG.-R.); (N.M.-A.)
| |
Collapse
|
21
|
Fujimoto Y, Yokozeki T, Yokoyama A, Tabata Y. Basic fibroblast growth factor enhances proliferation and hepatocyte growth factor expression of feline mesenchymal stem cells. Regen Ther 2020; 15:10-17. [PMID: 32490062 PMCID: PMC7256438 DOI: 10.1016/j.reth.2020.03.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 02/26/2020] [Accepted: 03/18/2020] [Indexed: 12/20/2022] Open
Abstract
Introduction The objective of this study is to evaluate the effect of basic fibroblast growth factor (bFGF) on the proliferation and secretion activity of feline adipose-derived mesenchymal stem cells (MSC). Methods Feline MSC isolated from the subcutaneous adipose tissue of cats were cultured with or without bFGF. Results The bFGF addition enhanced the proliferation of feline MSC to a significant great extent compared with that without bFGF, although the cell proliferation tended to increase with the bFGF concentration. In addition, adipogenic and osteogenic staining assay demonstrated that the bFGF addition allowed MSC to maintain the differentiation ability even after the proliferation. Moreover, no change in the surface markers of MSC was observed between the cultures with or without bFGF. A quantitative RT-PCR assay revealed that the HGF and TSG-6 expression significantly increased by the bFGF addition. The highest mRNA expression of MMP-2 was observed for cells cultured in 1000 ng/ml bFGF concentration. Conclusions The culture with bFGF is a promising way to enhance the proliferation, and HGF secretion ability of MSC as well as maintain their differentiation ability and immunophenotype nature. Feline adipose-derived mesenchymal stem cells (MSC) was cultured with or without the basic fibroblast growth factor (bFGF). The bFGF enhanced the proliferation and increased the mRNA expression of HGF, TSG-6, and MMP-2. The bFGF addition was not influenced to the differentiation ability and cell surface marker of MSC.
Collapse
Key Words
- Basic fibroblast growth factor
- CKD, chronic kidney disease
- ECM, extracellular matrix
- FBS, fetal bovine serum
- FGF, basic fibroblast growth factor
- Feline
- GAPDH, gliyceraldehyde-3-phosphate dehydrogenase
- HGF, hepatocyte growth factor
- Hepatocyte growth factor
- MMP-2, matrix metalloproteinase-2
- MSC, mesenchymal stem cells
- Mesenchymal stem cell
- P1, passage 1
- Proliferation
- SVF, stromal vascular fraction
- TSG-6, tumor necrosis factor-stimulated gene 6
- Tumor necrosis factor-stimulated gene 6
Collapse
Affiliation(s)
- Youhei Fujimoto
- Department of Advanced Pathobiology, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, Osaka, Japan.,Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | | | | | - Yasuhiko Tabata
- Laboratory of Biomaterials, Department of Regeneration Science and Engineering, Institute for Frontier Life and Medical Sciences, Kyoto University, Kyoto, Japan
| |
Collapse
|