1
|
Bai X, Huang X, Yi J, Yan X, Hu T, Wang L, Wang Z, Gao H. Cardiac Fibroblasts Enhance MMP2 Activity to Suppress Gap Junction Function in Cardiomyocytes. Appl Biochem Biotechnol 2024; 196:8493-8512. [PMID: 38878160 DOI: 10.1007/s12010-024-04986-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/05/2024] [Indexed: 01/04/2025]
Abstract
Although it is crucial to promptly restore blood perfusion to revive the ischemic myocardium, reperfusion itself can paradoxically contribute to the electrical instability and arrhythmias of the myocardium. Several studies have revealed that cardiac fibroblasts can impact cardiac electrophysiology through various mechanisms including the deposition of extracellular matrix, release of chemical mediators, and direct electrical coupling with myocytes. Previously, we have shown that hypoxia/reoxygenation (H/R)-treated rat fibroblasts conditional medium (H/R-FCM) could decrease the spontaneous beating frequency of rat neonatal cardiomyocytes and downregulate the expression of gap junction proteins. However, the specific mechanism by which H/R-FCM affects the gap junctions requires further investigation. H/R-FCM was obtained by culturing confluent rat cardiac fibroblasts (RCF) for 4 h under hypoxic conditions. Gap junction function, hemichannel activity, and expression of Cx43 were examined upon treatment with H/R-FCM. Gelatin zymography was performed to detect matrix metalloproteinase (MMP) activity in the conditioned medium. The effect of H/R-FCM and MMP2 inhibitors on cardiac electrophysiology and arrhythmias was investigated with an isolated rat ischemia/reperfusion (I/R) model. H/R-FCM treatment impaired gap junction function, downregulated Cx43 expression, and increased hemichannel activity in rat cardiomyocytes (H9c2). The adverse effect of H/R-FCM on gap junction, which was confirmed by the cardiomyocyte H/R model, was involved in the activation of MMP2. MMP2 inhibition could partially attenuate the detrimental effects of I/R on myocardial electrophysiological indices and arrhythmia susceptibility. Our study indicates that inhibition of MMP2 may be a promising therapeutic target for the treatment of reperfusion arrhythmia.
Collapse
Affiliation(s)
- Xue Bai
- School of Anesthesiology, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Xiang Huang
- School of Anesthesiology, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Jing Yi
- School of Anesthesiology, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Xu Yan
- School of Anesthesiology, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Tingju Hu
- School of Anesthesiology, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Lu Wang
- School of Anesthesiology, Guizhou Medical University, Guiyang, 550004, Guizhou, China
- Guiyang Maternal and Child Health Hospital, Guiyang, 550004, Guizhou, China
| | - Zijun Wang
- School of Anesthesiology, Guizhou Medical University, Guiyang, 550004, Guizhou, China
- Department of Anesthesiology, Guiyang Second People's Hospital, Guiyang, 550081, Guizhou, China
| | - Hong Gao
- School of Anesthesiology, Guizhou Medical University, Guiyang, 550004, Guizhou, China.
- Guizhou Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Guiyang, 550025, Guizhou, China.
| |
Collapse
|
2
|
An Z, Sun Y, Yang X, Zhou J, Yu Y, Zhang B, Xu Z, Zhu Y, Wang G. Enhanced expression of miR-20a driven by nanog exacerbated the degradation of extracellular matrix in thoracic aortic dissection. Noncoding RNA Res 2024; 9:1040-1049. [PMID: 39022686 PMCID: PMC11254500 DOI: 10.1016/j.ncrna.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 05/09/2024] [Accepted: 05/19/2024] [Indexed: 07/20/2024] Open
Abstract
Thoracic aortic dissection (TAD) is a life-threatening vascular disease manifested as intramural bleeding in the medial layers of the thoracic aorta. The key histopathologic feature of TAD is medial degeneration, characterized by depletion of vascular smooth muscle cells (VSMCs) and degradation of extracellular matrix (ECM). MicroRNA, as essential epigenetic regulators, can inhibit the protein expression of target genes without modifying the sequences. This study aimed to elucidate the role and underlying mechanism of miR-20a, a member of the miR-17-92 cluster, in regulating ECM degradation during the pathogenesis of TAD. The expression of the miR-17-92 cluster was significantly increased in synthetic VSMCs derived from TAD lesions compared to contractile VSMCs isolated from normal thoracic aortas. Notably, the expression of miR-20a was increased in VSMCs in response to serum exposure and various stimuli. In TAD lesions, the expression of miR-20a was significantly negatively correlated with that of elastin. Elevated expression of miR-20a was also observed in thoracic aortas of TAD mice induced by β-aminopropionitrile fumarate and angiotensin II. Overexpression of miR-20a via mimic transfection enhanced the growth and invasive capabilities of VSMCs, with no significant impact on their migratory activity or the expression of phenotypic markers (α-SMA, SM22, and OPN). Silencing of miR-20a with inhibitor transfection mitigated the hyperactivation of MMP2 in VSMCs stimulated by PDGF-bb, as evidenced by reduced levels of active-MMP2 and increased levels of pro-MMP2. Subsequently, TIMP2 was identified as a novel target gene of miR-20a. The role of miR-20a in promoting the activation of MMP2 was mediated by the suppression of TIMP2 expression in VSMCs. In addition, the elevated expression of miR-20a was found to be directly driven by Nanog in VSMCs. Collectively, these findings indicate that miR-20a plays a crucial role in maintaining the homeostasis of the thoracic aortic wall during TAD pathogenesis and may represent a potential therapeutic target for TAD.
Collapse
Affiliation(s)
- Zhao An
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yangyong Sun
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
- Department of Cardiothoracic Surgery, Affiliated People's Hospital of Jiangsu University, Zhenjiang, China
| | - Xiaodong Yang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jingwen Zhou
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yongchao Yu
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Boyao Zhang
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Zhiyun Xu
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Yuming Zhu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guokun Wang
- Department of Cardiovascular Surgery, Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| |
Collapse
|
3
|
Zhang M, Zhang R, Hao J, Zhao X, Ma Z, Peng Y, Bao B, Xin J, Yin X, Bi H, Guo D. Quercetin Alleviates Scleral Remodeling Through Inhibiting the PERK-EIF2α Axis in Experiment Myopia. Invest Ophthalmol Vis Sci 2024; 65:11. [PMID: 39504054 PMCID: PMC11549929 DOI: 10.1167/iovs.65.13.11] [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: 07/11/2024] [Accepted: 10/17/2024] [Indexed: 11/10/2024] Open
Abstract
Purpose This study aims to investigate the effect of quercetin (QUE) on scleral remodeling by inhibiting the PERK-EIF2α signaling pathway and to evaluate its potential role in slowing myopia. Methods Lens-induced myopia (LIM) guinea pigs were obtained and treated with QUE. After 4 and 6 weeks of treatments, ocular biological measurements were conducted. Hematoxylin and eosin (H&E) staining was used to observe the changes in scleral morphology and thickness, and Masson staining was used to examine scleral collagen fiber arrangement. Quantitative PCR (qPCR) and Western bolt were utilized to detect the mRNA and protein expression of PERK, EIF2α, MMP-2, TIMP-2, and collagen I in the scleral tissues. Calcium ion flow in each group was measured using noninvasive micro-test technology, and reactive oxygen species levels were detected by flow cytometry. Results Compared with the LIM group, the ocular measurements showed that the refractive errors and axial length of the eyes were significantly reduced in the LIM + QUE group (P < 0.01). H&E and Masson staining showed that sclera in the LIM + QUE group was thickened, collagen was dense, and the fiber gap was reduced. In the LIM + QUE group, the expression levels of PERK, EIF2α, and MMP-2 were decreased, whereas the expression levels of TIMP-2 and collagen I were increased. Calcium release and reactive oxygen species (ROS) in the LIM + QUE group were decreased. Conclusions Quercetin ameliorates scleral remodeling in myopic guinea pigs by inhibiting the PERK-EIF2α signaling pathway, thereby alleviating the progression of myopia. These findings provide new experimental evidence for the potential application of quercetin in myopia prevention and treatment.
Collapse
Affiliation(s)
- Miao Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ruixue Zhang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jiawen Hao
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiaoyue Zhao
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhongyu Ma
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Peng
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bo Bao
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jizhao Xin
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuewei Yin
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dadong Guo
- Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
- Shandong Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Jinan, China
- Shandong Academy of Eye Disease Prevention and Therapy, Jinan, China
| |
Collapse
|
4
|
Făgărășan A, Săsăran MO, Gozar L, Toma D, Șuteu C, Ghiragosian-Rusu S, Al-Akel FC, Szabo B, Huțanu A. Circulating Matrix Metalloproteinases for Prediction of Aortic Dilatation in Children with Bicuspid Aortic Valve: A Single-Center, Observational Study. Int J Mol Sci 2024; 25:10538. [PMID: 39408865 PMCID: PMC11476682 DOI: 10.3390/ijms251910538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 09/19/2024] [Accepted: 09/25/2024] [Indexed: 10/20/2024] Open
Abstract
Circulating biomarkers have been proposed for early identification of aortic dilatation progression associated with bicuspid aortic valve (BAV), but matrix metalloproteinases (MMPs) are distinguished as signatures of increased extracellular matrix degradation, a landmark of aneurysm formation. The current study aims to identify the role of MMP-1, MMP-2, MMP-9, and the MMP inhibitor, TIMP-1, in identifying aortic dilation in children with BAV. We conducted a study on 73 children divided into two study groups, depending on the presence of aortic dilatation (group 1-43 BAV controls and group 2-30 children with BAV and aortic dilatation). Each patient underwent a cardiac ultrasound and, in each case, serum MMP-1, MMP-2, MMP-9, and TIMP-1 were quantified using xMAP technology. Comparison of the MMPs between the two study groups revealed significantly higher values only in the case of TIMP-1, among BAV controls. Moreover, the same TIMP-1 inversely correlated with aortic annulus absolute size and z score, as well as with ascending aorta z score. No particular correlation between the aortic phenotype and the presence of aortic dilatation was found. Future longitudinal research starting at pediatric ages could show the significance of MMPs screening in BAV individuals as predictors of aortic aneurysm formation.
Collapse
Affiliation(s)
- Amalia Făgărășan
- Department of Pediatrics III, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (A.F.); (L.G.); (D.T.); (C.Ș.); (S.G.-R.)
- Department of Pediatric Cardiology, Emergency Institute for Cardiovascular Diseases and Transplantation of Târgu Mureș, Gheorghe Marinescu Street No 50, 540136 Târgu Mureș, Romania; (F.C.A.-A.); (B.S.)
| | - Maria Oana Săsăran
- Department of Pediatrics III, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540142 Târgu Mureș, Romania
| | - Liliana Gozar
- Department of Pediatrics III, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (A.F.); (L.G.); (D.T.); (C.Ș.); (S.G.-R.)
- Department of Pediatric Cardiology, Emergency Institute for Cardiovascular Diseases and Transplantation of Târgu Mureș, Gheorghe Marinescu Street No 50, 540136 Târgu Mureș, Romania; (F.C.A.-A.); (B.S.)
| | - Daniela Toma
- Department of Pediatrics III, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (A.F.); (L.G.); (D.T.); (C.Ș.); (S.G.-R.)
- Department of Pediatric Cardiology, Emergency Institute for Cardiovascular Diseases and Transplantation of Târgu Mureș, Gheorghe Marinescu Street No 50, 540136 Târgu Mureș, Romania; (F.C.A.-A.); (B.S.)
| | - Carmen Șuteu
- Department of Pediatrics III, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (A.F.); (L.G.); (D.T.); (C.Ș.); (S.G.-R.)
- Department of Pediatric Cardiology, Emergency Institute for Cardiovascular Diseases and Transplantation of Târgu Mureș, Gheorghe Marinescu Street No 50, 540136 Târgu Mureș, Romania; (F.C.A.-A.); (B.S.)
| | - Simina Ghiragosian-Rusu
- Department of Pediatrics III, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania; (A.F.); (L.G.); (D.T.); (C.Ș.); (S.G.-R.)
- Department of Pediatric Cardiology, Emergency Institute for Cardiovascular Diseases and Transplantation of Târgu Mureș, Gheorghe Marinescu Street No 50, 540136 Târgu Mureș, Romania; (F.C.A.-A.); (B.S.)
| | - Flavia Cristina Al-Akel
- Department of Pediatric Cardiology, Emergency Institute for Cardiovascular Diseases and Transplantation of Târgu Mureș, Gheorghe Marinescu Street No 50, 540136 Târgu Mureș, Romania; (F.C.A.-A.); (B.S.)
- Pathophysiology Department, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania
| | - Boglarka Szabo
- Department of Pediatric Cardiology, Emergency Institute for Cardiovascular Diseases and Transplantation of Târgu Mureș, Gheorghe Marinescu Street No 50, 540136 Târgu Mureș, Romania; (F.C.A.-A.); (B.S.)
| | - Adina Huțanu
- Department of Laboratory Medicine, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania;
| |
Collapse
|
5
|
Hughes DM, Won T, Talor MV, Kalinoski HM, Jurčová I, Szárszoi O, Stříž I, Čurnová L, Bracamonte-Baran W, Melenovský V, Čiháková D. The protective role of GATA6 + pericardial macrophages in pericardial inflammation. iScience 2024; 27:110244. [PMID: 39040070 PMCID: PMC11260870 DOI: 10.1016/j.isci.2024.110244] [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: 10/22/2023] [Revised: 03/18/2024] [Accepted: 06/07/2024] [Indexed: 07/24/2024] Open
Abstract
Prior research has suggested that GATA6+ pericardial macrophages may traffic to the myocardium to prevent interstitial fibrosis after myocardial infarction (MI), while subsequent literature claims that they do not. We demonstrate that GATA6+ pericardial macrophages are critical for preventing IL-33 induced pericarditis and attenuate trafficking of inflammatory monocytes and granulocytes to the pericardial cavity after MI. However, absence of GATA6+ macrophages did not affect myocardial inflammation due to MI or coxsackievirus-B3 induced myocarditis, or late-stage cardiac fibrosis and cardiac function post MI. GATA6+ macrophages are significantly less transcriptionally active following stimulation in vitro compared to bone marrow-derived macrophages and do not induce upregulation of inflammatory markers in fibroblasts. This suggests that GATA6+ pericardial macrophages attenuate inflammation through their interactions with surrounding cells. We therefore conclude that GATA6+ pericardial macrophages are critical in modulating pericardial inflammation, but do not play a significant role in controlling myocardial inflammation or fibrosis.
Collapse
Affiliation(s)
- David M. Hughes
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, USA
| | - Taejoon Won
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Monica V. Talor
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hannah M. Kalinoski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Ivana Jurčová
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Ondrej Szárszoi
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Ilja Stříž
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Lenka Čurnová
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | | | - Vojtěch Melenovský
- Institute for Clinical and Experimental Medicine (IKEM), Prague, Czech Republic
| | - Daniela Čiháková
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
6
|
Humeres C, Shinde AV, Tuleta I, Hernandez SC, Hanna A, Huang S, Venugopal H, Aguilan JT, Conway SJ, Sidoli S, Frangogiannis NG. Fibroblast Smad7 Induction Protects the Remodeling Pressure-Overloaded Heart. Circ Res 2024; 135:453-469. [PMID: 38899461 PMCID: PMC11257802 DOI: 10.1161/circresaha.123.323360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 06/06/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND Cardiac fibroblast activation contributes to adverse remodeling, fibrosis, and dysfunction in the pressure-overloaded heart. Although early fibroblast TGF-β (transforming growth factor-β)/Smad (small mother against decapentaplegic)-3 activation protects the pressure-overloaded heart by preserving the matrix, sustained TGF-β activation is deleterious, accentuating fibrosis and dysfunction. Thus, endogenous mechanisms that negatively regulate the TGF-β response in fibroblasts may be required to protect from progressive fibrosis and adverse remodeling. We hypothesized that Smad7, an inhibitory Smad that restrains TGF-β signaling, may be induced in the pressure-overloaded myocardium and may regulate fibrosis, remodeling, and dysfunction. METHODS The effects of myofibroblast-specific Smad7 loss were studied in a mouse model of transverse aortic constriction, using echocardiography, histological analysis, and molecular analysis. Proteomic studies in S7KO (Smad7 knockout) and overexpressing cells were used to identify fibroblast-derived mediators modulated by Smad7. In vitro experiments using cultured cardiac fibroblasts, fibroblasts populating collagen lattices, and isolated macrophages were used to dissect the molecular signals responsible for the effects of Smad7. RESULTS Following pressure overload, Smad7 was upregulated in cardiac myofibroblasts. TGF-β and angiotensin II stimulated fibroblast Smad7 upregulation via Smad3, whereas GDF15 (growth differentiation factor 15) induced Smad7 through GFRAL (glial cell line-derived neurotrophic factor family receptor α-like). MFS7KO (myofibroblast-specific S7KO) mice had increased mortality, accentuated systolic dysfunction and dilative remodeling, and accelerated diastolic dysfunction in response to transverse aortic constriction. Increased dysfunction in MFS7KO hearts was associated with accentuated fibrosis and increased MMP (matrix metalloproteinase)-2 activity and collagen denaturation. Secretomic analysis showed that Smad7 loss accentuates secretion of structural collagens and matricellular proteins and markedly increases MMP2 secretion. In contrast, Smad7 overexpression reduced MMP2 levels. In fibroblasts populating collagen lattices, the effects of Smad7 on fibroblast-induced collagen denaturation and pad contraction were partly mediated via MMP2 downregulation. Surprisingly, MFS7KO mice also exhibited significant macrophage expansion caused by paracrine actions of Smad7 null fibroblasts that stimulate macrophage proliferation and fibrogenic activation. Macrophage activation involved the combined effects of the fibroblast-derived matricellular proteins CD5L (CD5 antigen-like), SPARC (secreted protein acidic and rich in cysteine), CTGF (connective tissue growth factor), ECM1 (extracellular matrix protein 1), and TGFBI (TGFB induced). CONCLUSIONS The antifibrotic effects of Smad7 in the pressure-overloaded heart protect from dysfunction and involve not only reduction in collagen deposition but also suppression of MMP2-mediated matrix denaturation and paracrine effects that suppress macrophage activation through inhibition of matricellular proteins.
Collapse
Affiliation(s)
- Claudio Humeres
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Indiana University School of Medicine, Indianapolis IN
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis IN
| | - Arti V Shinde
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Indiana University School of Medicine, Indianapolis IN
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis IN
| | - Izabela Tuleta
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Indiana University School of Medicine, Indianapolis IN
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis IN
| | - Silvia C Hernandez
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Indiana University School of Medicine, Indianapolis IN
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis IN
| | - Anis Hanna
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Indiana University School of Medicine, Indianapolis IN
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis IN
| | - Shuaibo Huang
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Indiana University School of Medicine, Indianapolis IN
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis IN
| | - Harikrishnan Venugopal
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Indiana University School of Medicine, Indianapolis IN
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis IN
| | - Jennifer T Aguilan
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx NY
| | - Simon J Conway
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis IN
| | - Simone Sidoli
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx NY
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Indiana University School of Medicine, Indianapolis IN
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis IN
| |
Collapse
|
7
|
Szabados T, Molnár A, Kenyeres É, Gömöri K, Pipis J, Pósa B, Makkos A, Ágg B, Giricz Z, Ferdinandy P, Görbe A, Bencsik P. Identification of New, Translatable ProtectomiRs against Myocardial Ischemia/Reperfusion Injury and Oxidative Stress: The Role of MMP/Biglycan Signaling Pathways. Antioxidants (Basel) 2024; 13:674. [PMID: 38929113 PMCID: PMC11201193 DOI: 10.3390/antiox13060674] [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: 04/09/2024] [Revised: 05/22/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
INTRODUCTION Ischemic conditionings (ICon) were intensively investigated and several protective signaling pathways were identified. Previously, we have shown the role of matrix metalloproteinases (MMP) in myocardial ischemia/reperfusion injury (MIRI) and the cardioprotective role of biglycan (BGN), a small leucine-rich proteoglycan in vitro. Here, we hypothesized that cardiac MMP and BGN signaling are involved in the protective effects of ICon. METHODS A reverse target-microRNA prediction was performed by using the miRNAtarget™ 2.0 software to identify human microRNAs with a possible regulatory effect on MMP and BGN, such as on related genes. To validate the identified 1289 miRNAs in the predicted network, we compared them to two cardioprotective miRNA omics datasets derived from pig and rat models of MIRI in the presence of ICons. RESULTS Among the experimentally measured miRNAs, we found 100% sequence identity to human predicted regulatory miRNAs in the case of 37 porcine and 24 rat miRNAs. Upon further analysis, 42 miRNAs were identified as MIRI-associated miRNAs, from which 24 miRNAs were counter-regulated due to ICons. CONCLUSIONS Our findings highlight 24 miRNAs that potentially regulate cardioprotective therapeutic targets associated with MMPs and BGN in a highly translatable porcine model of acute myocardial infarction.
Collapse
Affiliation(s)
- Tamara Szabados
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
| | - Arnold Molnár
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
| | - Éva Kenyeres
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
| | - Judit Pipis
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
| | - Bence Pósa
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
| | - András Makkos
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary;
| | - Bence Ágg
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary;
| | - Zoltán Giricz
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary;
| | - Péter Ferdinandy
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary;
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
- Cardiometabolic and HUN-REN-SU System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Nagyvárad tér 4, H-1089 Budapest, Hungary;
| | - Péter Bencsik
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Albert Szent-Györgyi Medical School, University of Szeged, Dóm tér 12, H-6720 Szeged, Hungary; (T.S.); (A.M.); (É.K.); (K.G.); (J.P.); (B.P.); (A.G.)
- Pharmahungary Group, Hajnóczy u. 6, H-6722 Szeged, Hungary; (B.Á.); (Z.G.); (P.F.)
| |
Collapse
|
8
|
Surówka A, Żołnierczuk M, Prowans P, Grabowska M, Kupnicka P, Markowska M, Olejnik-Wojciechowska J, Szlosser Z, Wilk A, Szumilas K, Kędzierska-Kapuza K. The Effects of Chronic Immunosuppressive Treatment on Morphological Changes in Cardiac Tissue and the Balance between Matrix Metalloproteinases (MMP-2 and MMP-9) and Their Inhibitors in the Rat Heart. Int J Mol Sci 2024; 25:4468. [PMID: 38674053 PMCID: PMC11049927 DOI: 10.3390/ijms25084468] [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: 04/01/2024] [Revised: 04/12/2024] [Accepted: 04/16/2024] [Indexed: 04/28/2024] Open
Abstract
Using different three-drug immunosuppressive treatment regimens in a rat model, we aimed to determine the effects of long-term therapy on metalloproteinase-2 and metalloproteinase-9 activity and the expression of their inhibitors, as well as to assess the morphology of the animals' cardiac tissue. Our results suggest that chronic use of immunosuppressive drugs disrupts the balance between the activity of MMPs and TIMPs. Depending on the type of drug regimen used, this leads to abnormalities in the cardiac structure, collagen fiber accumulation, or cardiomyocyte hypertrophy. The information obtained in the present study allows us to conclude that the chronic treatment of rats with the most common clinical immunosuppressive regimens may contribute to abnormalities in the myocardial structure and function. The results presented in this study may serve as a prelude to more in-depth analyses and additional research into the optimal selection of an immunosuppressive treatment with the lowest possible risk of cardiovascular complications for patients receiving organ transplants.
Collapse
Affiliation(s)
- Anna Surówka
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Michał Żołnierczuk
- Department of Vascular Surgery, General Surgery and Angiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Piotr Prowans
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Marta Grabowska
- Department of Histology and Developmental Biology, Faculty of Health Sciences, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Patrycja Kupnicka
- Department of Biochemistry and Medical Chemistry, Pomeranian Medical University, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
| | - Marta Markowska
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
- Department of Plastic and Reconstructive Surgery, 109 Military Hospital, 71-422 Szczecin, Poland
| | | | - Zbigniew Szlosser
- Department of Plastic, Endocrine and General Surgery, Pomeranian Medical University, 72-010 Szczecin, Poland
| | - Aleksandra Wilk
- Department of Histology and Embryology, Pomeranian Medical University, 70-111 Szczecin, Poland;
| | - Kamila Szumilas
- Department of Physiology, Pomeranian Medical University, 70-111 Szczecin, Poland
| | - Karolina Kędzierska-Kapuza
- Department of Gastroenterological Surgery and Transplantology, Centre of Postgraduate Medical Education in Warsaw, 137 Woloska St., 02-507 Warsaw, Poland
| |
Collapse
|
9
|
Mukherjee A, Das B. The role of inflammatory mediators and matrix metalloproteinases (MMPs) in the progression of osteoarthritis. BIOMATERIALS AND BIOSYSTEMS 2024; 13:100090. [PMID: 38440290 PMCID: PMC10910010 DOI: 10.1016/j.bbiosy.2024.100090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 12/04/2023] [Accepted: 02/20/2024] [Indexed: 03/06/2024] Open
Abstract
Osteoarthritis (OA) is a chronic musculoskeletal disorder characterized by an imbalance between (synthesis) and catabolism (degradation) in altered homeostasis of articular cartilage mediated primarily by the innate immune system. OA degenerates the joints resulting in synovial hyperplasia, degradation of articular cartilage with damage of the structural and functional integrity of the cartilage extracellular matrix, subchondral sclerosis, osteophyte formation, and is characterized by chronic pain, stiffness, and loss of function. Inflammation triggered by factors like biomechanical stress is involved in the development of osteoarthritis. In OA apart from catabolic effects, anti-inflammatory anabolic processes also occur continually. There is also an underlying chronic inflammation present, not only in cartilage tissue but also within the synovium, which perpetuates tissue destruction of the OA joint. The consideration of inflammation in OA considers synovitis and/or other cellular and molecular events in the synovium during the progression of OA. In this review, we have presented the progression of joint degradation that results in OA. The critical role of inflammation in the pathogenesis of OA is discussed in detail along with the dysregulation within the cytokine networks composed of inflammatory and anti-inflammatory cytokines that drive catabolic pathways, inhibit matrix synthesis, and promote cellular apoptosis. OA pathogenesis, fluctuation of synovitis, and its clinical impact on disease progression are presented here along with the role of synovial macrophages in promoting inflammatory and destructive responses in OA. The role of interplay between different cytokines, structure, and function of their receptors in the inter-cellular signaling pathway is further explored. The effect of cytokines in the increased synthesis and release of matrix-decomposing proteolytic enzymes, such as matrix metalloproteinase (MMPs) and a disintegrin-like and metalloproteinase with thrombospondin motif (ADAMTS), is elaborated emphasizing the potential impact of MMPs on the chondrocytes, synovial cells, articular and periarticular tissues, and other immune system cells migrating to the site of inflammation. We also shed light on the pathogenesis of OA via oxidative damage particularly due to nitric oxide (NO) via its angiogenic response to inflammation. We concluded by presenting the current knowledge about the tissue inhibitors of metalloproteinases (TIMPs). Synthetic MMP inhibitors include zinc binding group (ZBG), non-ZBG, and mechanism-based inhibitors, all of which have the potential to be therapeutically beneficial in the treatment of osteoarthritis. Improving our understanding of the signaling pathways and molecular mechanisms that regulate the MMP gene expression, may open up new avenues for the creation of therapies that can stop the joint damage associated with OA.
Collapse
Affiliation(s)
- Anwesha Mukherjee
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| | - Bodhisatwa Das
- Department of Biomedical Engineering, Indian Institute of Technology Ropar, India
| |
Collapse
|
10
|
Dattani A, Brady EM, Kanagala P, Stoma S, Parke KS, Marsh AM, Singh A, Arnold JR, Moss AJ, Zhao L, Cvijic ME, Fronheiser M, Du S, Costet P, Schafer P, Carayannopoulos L, Chang CP, Gordon D, Ramirez-Valle F, Jerosch-Herold M, Nelson CP, Squire IB, Ng LL, Gulsin GS, McCann GP. Is atrial fibrillation in HFpEF a distinct phenotype? Insights from multiparametric MRI and circulating biomarkers. BMC Cardiovasc Disord 2024; 24:94. [PMID: 38326736 PMCID: PMC10848361 DOI: 10.1186/s12872-024-03734-0] [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: 06/29/2023] [Accepted: 01/17/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Heart failure with preserved ejection fraction (HFpEF) and atrial fibrillation (AF) frequently co-exist. There is a limited understanding on whether this coexistence is associated with distinct alterations in myocardial remodelling and mechanics. We aimed to determine if patients with atrial fibrillation (AF) and heart failure with preserved ejection fraction (HFpEF) represent a distinct phenotype. METHODS In this secondary analysis of adults with HFpEF (NCT03050593), participants were comprehensively phenotyped with stress cardiac MRI, echocardiography and plasma fibroinflammatory biomarkers, and were followed for the composite endpoint (HF hospitalisation or death) at a median of 8.5 years. Those with AF were compared to sinus rhythm (SR) and unsupervised cluster analysis was performed to explore possible phenotypes. RESULTS 136 subjects were included (SR = 75, AF = 61). The AF group was older (76 ± 8 vs. 70 ± 10 years) with less diabetes (36% vs. 61%) compared to the SR group and had higher left atrial (LA) volumes (61 ± 30 vs. 39 ± 15 mL/m2, p < 0.001), lower LA ejection fraction (EF) (31 ± 15 vs. 51 ± 12%, p < 0.001), worse left ventricular (LV) systolic function (LVEF 63 ± 8 vs. 68 ± 8%, p = 0.002; global longitudinal strain 13.6 ± 2.9 vs. 14.7 ± 2.4%, p = 0.003) but higher LV peak early diastolic strain rates (0.73 ± 0.28 vs. 0.53 ± 0.17 1/s, p < 0.001). The AF group had higher levels of syndecan-1, matrix metalloproteinase-2, proBNP, angiopoietin-2 and pentraxin-3, but lower level of interleukin-8. No difference in clinical outcomes was observed between the groups. Three distinct clusters were identified with the poorest outcomes (Log-rank p = 0.029) in cluster 2 (hypertensive and fibroinflammatory) which had equal representation of SR and AF. CONCLUSIONS Presence of AF in HFpEF is associated with cardiac structural and functional changes together with altered expression of several fibro-inflammatory biomarkers. Distinct phenotypes exist in HFpEF which may have differing clinical outcomes.
Collapse
Affiliation(s)
- Abhishek Dattani
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK.
| | - Emer M Brady
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | | | - Svetlana Stoma
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Kelly S Parke
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Anna-Marie Marsh
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Anvesha Singh
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Jayanth R Arnold
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Alastair J Moss
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Lei Zhao
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | - Shuyan Du
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | | | | | | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Iain B Squire
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Leong L Ng
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Gaurav S Gulsin
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Gerry P McCann
- Department of Cardiovascular Sciences, University of Leicester and the National Institute for Health Research Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| |
Collapse
|
11
|
Buckley LF, Agha AM, Dorbala P, Claggett BL, Yu B, Hussain A, Nambi V, Chen LY, Matsushita K, Hoogeveen RC, Ballantyne CM, Shah AM. MMP-2 Associates With Incident Heart Failure and Atrial Fibrillation: The ARIC Study. Circ Heart Fail 2023; 16:e010849. [PMID: 37753653 PMCID: PMC10842537 DOI: 10.1161/circheartfailure.123.010849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 08/16/2023] [Indexed: 09/28/2023]
Abstract
BACKGROUND MMP (matrix metalloproteinase)-2 participates in extracellular matrix regulation and may be involved in heart failure (HF), atrial fibrillation (AF), and coronary heart disease. METHODS Among the 4693 ARIC study (Atherosclerosis Risk in Communities) participants (mean age, 75±5 years; 42% women) without prevalent HF, multivariable Cox proportional hazard models were used to estimate associations of plasma MMP-2 levels with incident HF, HF with preserved ejection fraction (≥50%), HF with reduced ejection fraction (<50%), AF, and coronary heart disease. Mediation of the association between MMP-2 and HF was assessed by censoring participants who developed AF or coronary heart disease before HF. Multivariable linear regression models were used to assess associations of MMP-2 with measures of left ventricular and left atrial structure and function. RESULTS Compared with the 3 lower quartiles, the highest MMP-2 quartile associated with greater risk of incident HF overall (adjusted hazard ratio, 1.48 [95% CI, 1.21-1.81]), incident HF with preserved ejection fraction (1.44 [95% CI, 1.07-1.94]), incident heart failure with reduced ejection fraction (1.48 [95% CI, 1.08-2.02]), and incident AF (1.44 [95% CI, 1.18-1.77]) but not incident coronary heart disease (0.97 [95% CI, 0.71-1.34]). Censoring AF attenuated the MMP-2 association with HF with preserved ejection fraction. Higher plasma MMP-2 levels were associated with larger left ventricular end-diastolic volume index, greater left ventricular mass index, higher E/e' ratio, larger left atrial volume index, and worse left atrial reservoir and contractile strains (all P<0.001). CONCLUSIONS Higher plasma MMP-2 levels associate with diastolic dysfunction, left atrial dysfunction, and a higher risk of incident HF and AF. AF is a mediator of MMP-2-associated HF with preserved ejection fraction risk.
Collapse
Affiliation(s)
- Leo F Buckley
- Department of Pharmacy Services (L.F.B.), Brigham and Women's Hospital, Boston, MA
| | - Ali M Agha
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Pranav Dorbala
- Division of Cardiovascular Medicine (P.D., B.L.C.), Brigham and Women's Hospital, Boston, MA
| | - Brian L Claggett
- Division of Cardiovascular Medicine (P.D., B.L.C.), Brigham and Women's Hospital, Boston, MA
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, University of Texas Health Science Center at Houston (B.Y.)
| | - Aliza Hussain
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Vijay Nambi
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
- Michael E. DeBakey Veterans Affairs Hospital, Houston, TX (V.N.)
| | - Lin Yee Chen
- Division of Cardiovascular Medicine, University of Minnesota, Minneapolis (L.Y.C.)
| | - Kunihiro Matsushita
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD (K.M.)
| | - Ron C Hoogeveen
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Christie M Ballantyne
- Division of Cardiovascular Medicine, Baylor College of Medicine, Houston, TX (A.A., A.H., V.N., R.C.H., C.M.B.)
| | - Amil M Shah
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas (A.M.S.)
| |
Collapse
|
12
|
Bräuninger H, Krüger S, Bacmeister L, Nyström A, Eyerich K, Westermann D, Lindner D. Matrix metalloproteinases in coronary artery disease and myocardial infarction. Basic Res Cardiol 2023; 118:18. [PMID: 37160529 PMCID: PMC10169894 DOI: 10.1007/s00395-023-00987-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/11/2023]
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of death worldwide. Most cardiovascular deaths are caused by ischaemic heart diseases such as myocardial infarction (MI). Hereby atherosclerosis in the coronary arteries often precedes disease manifestation. Since tissue remodelling plays an important role in the development and progression of atherosclerosis as well as in outcome after MI, regulation of matrix metalloproteinases (MMPs) as the major ECM-degrading enzymes with diverse other functions is crucial. Here, we provide an overview of the expression profiles of MMPs in coronary artery and left ventricular tissue using publicly available data from whole tissue to single-cell resolution. To approach an association between MMP expression and the development and outcome of CVDs, we further review studies investigating polymorphisms in MMP genes since polymorphisms are known to have an impact on gene expression. This review therefore aims to shed light on the role of MMPs in atherosclerosis and MI by summarizing current knowledge from publically available datasets, human studies, and analyses of polymorphisms up to preclinical and clinical trials of pharmacological MMP inhibition.
Collapse
Affiliation(s)
- Hanna Bräuninger
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Side Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Saskia Krüger
- Clinic for Cardiology, University Heart and Vascular Centre Hamburg, Hamburg, Germany
| | - Lucas Bacmeister
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Alexander Nyström
- Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Kilian Eyerich
- Department of Dermatology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dirk Westermann
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany
| | - Diana Lindner
- Department of Cardiology and Angiology, University Heart Center Freiburg-Bad Krozingen, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Str. 55, 79106, Freiburg, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Side Hamburg/Kiel/Lübeck, Hamburg, Germany.
| |
Collapse
|
13
|
Chalise U, Becirovic‐Agic M, Lindsey ML. The cardiac wound healing response to myocardial infarction. WIREs Mech Dis 2023; 15:e1584. [PMID: 36634913 PMCID: PMC10077990 DOI: 10.1002/wsbm.1584] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 03/31/2022] [Accepted: 05/18/2022] [Indexed: 01/14/2023]
Abstract
Myocardial infarction (MI) is defined as evidence of myocardial necrosis consistent with prolonged ischemia. In response to MI, the myocardium undergoes a series of wound healing events that initiate inflammation and shift to anti-inflammation before transitioning to tissue repair that culminates in scar formation to replace the region of the necrotic myocardium. The overall response to MI is determined by two major steps, the first of which is the secretion of proteases by infiltrating leukocytes to breakdown extracellular matrix (ECM) components, a necessary step to remove necrotic cardiomyocytes. The second step is the generation of new ECM that comprises the scar; and this step is governed by the cardiac fibroblasts as the major source of new ECM synthesis. The leukocyte component resides in the middle of the two-step process, contributing to both sides as the leukocytes transition from pro-inflammatory to anti-inflammatory and reparative cell phenotypes. The balance between the two steps determines the final quantity and quality of scar formed, which in turn contributes to chronic outcomes following MI, including the progression to heart failure. This review will summarize our current knowledge regarding the cardiac wound healing response to MI, primarily focused on experimental models of MI in mice. This article is categorized under: Cardiovascular Diseases > Molecular and Cellular Physiology Immune System Diseases > Molecular and Cellular Physiology.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| | - Mediha Becirovic‐Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| | - Merry L. Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular ResearchUniversity of Nebraska Medical CenterOmahaNebraskaUSA
- Research ServiceNebraska‐Western Iowa Health Care SystemOmahaNebraskaUSA
| |
Collapse
|
14
|
Han X, Yang Y, Zhang M, Li L, Xue Y, Jia Q, Wang X, Guan S. Liquiritin Protects Against Cardiac Fibrosis After Myocardial Infarction by Inhibiting CCL5 Expression and the NF-κB Signaling Pathway. Drug Des Devel Ther 2022; 16:4111-4125. [PMID: 36483459 PMCID: PMC9724582 DOI: 10.2147/dddt.s386805] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 11/28/2022] [Indexed: 12/05/2022] Open
Abstract
Purpose Despite significant advances in interventional treatment, myocardial infarction (MI) and subsequent cardiac fibrosis remain major causes of high mortality worldwide. Liquiritin (LQ) is a flavonoid extract from licorice that possesses a variety of pharmacological properties. However, to our knowledge, the effects of LQ on myocardial fibrosis after MI have not been reported in detail. The aim of our research was to explore the potential role and mechanism of LQ in MI-induced myocardial damage. Methods The MI models were established by ligating the left anterior descending branch of the coronary artery. Next, rats were orally administered LQ once a day for 14 days. Biochemical assays, histopathological observations, ELISA, and Western blotting analyses were then conducted. Results LQ improved the heart appearance and ECG, decreased cardiac weight index and reduced levels of cardiac-specific markers such as CK, CK-MB, LDH, cTnI and BNP. Meanwhile, LQ reduced myocardial infarct size and improved hemodynamic parameters such as LVEDP, LVSP and ±dp/dtmax. Moreover, H&E staining showed that LQ attenuated the pathological damage caused by MI. Masson staining showed that LQ alleviated myocardial cell disorder and fibrosis while reducing collagen deposition. LQ also decreased the levels of oxidative stress and inflammation. Western blotting demonstrated that LQ significantly down-regulated the expressions of Collagen I, Collagen III, TGF-β1, MMP-9, α-SMA, CCL5, and p-NF-κB. Conclusion LQ protected against myocardial fibrosis following MI by improving cardiac function, and attenuating oxidative damage and inflammatory response, which may be associated with inhibition of CCL5 expression and the NF-κB pathway.
Collapse
Affiliation(s)
- Xue Han
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - Yakun Yang
- School of Pharmacy, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - Muqing Zhang
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China,Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - Li Li
- School of Pharmacy, Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Yucong Xue
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China
| | - Qingzhong Jia
- School of Pharmacy, Hebei Medical University, Shijiazhuang, People’s Republic of China
| | - Xiangting Wang
- College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China,Hebei Key Laboratory of Integrative Medicine on Liver-Kidney Patterns, Shijiazhuang, People’s Republic of China,Correspondence: Xiangting Wang, College of Integrative Medicine, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China, Email
| | - Shengjiang Guan
- Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China,School of Basic Medicine, Hebei University of Chinese Medicine, Shijiazhuang, People’s Republic of China,Shengjiang Guan, Affiliated Hospital, Hebei University of Chinese Medicine, Shijiazhuang, Hebei, People’s Republic of China, Email
| |
Collapse
|
15
|
Ivanova VV, Milto IV, Serebryakova ON, Sukhodolo IV. Detection of Matrix Metalloproteinases in the Heart of Preterm Rats. BIOL BULL+ 2022; 49:671-676. [DOI: 10.1134/s1062359022060073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 03/14/2022] [Accepted: 06/08/2022] [Indexed: 01/05/2025]
|
16
|
Protective effect of berberine in diabetic nephropathy: A systematic review and meta-analysis revealing the mechanism of action. Pharmacol Res 2022; 185:106481. [DOI: 10.1016/j.phrs.2022.106481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 09/25/2022] [Accepted: 09/29/2022] [Indexed: 12/09/2022]
|
17
|
Cheng Z, Zhang X, Zhang Y, Li L, Chen P. Role of MMP-2 and CD147 in kidney fibrosis. Open Life Sci 2022; 17:1182-1190. [PMID: 36185410 PMCID: PMC9482425 DOI: 10.1515/biol-2022-0482] [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: 03/02/2022] [Revised: 07/18/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
Matrix metalloproteinase-2 (MMP-2) and cluster of differentiation 147 (CD147) both play important roles in the development of kidney fibrosis, and CD147 can induce the production and activation of MMP-2. In the early stage of kidney fibrosis, MMP-2 promotes extracellular matrix (ECM) production and accelerates the development of kidney fibrosis, while in the advanced stage, MMP-2 activity decreases, leading to reduced ECM degradation and making it difficult to alleviate kidney fibrosis. The reason for the decrease in MMP-2 activity in the advanced stage is still unclear. On the one hand, it may be related to hypoxia and endocytosis, which lead to changes in the expression of MMP-2-related active regulatory molecules; on the other hand, it may be related to insufficient CD147 function. At present, the specific process by which CD147 is involved in the regulation of MMP-2 activity is not completely clear, and further in-depth studies are needed to clarify the roles of both factors in the pathophysiology of kidney fibrosis.
Collapse
Affiliation(s)
- Zhengyuan Cheng
- Department of Internal Medicine, Ma'anshan People's Hospital Affiliated to Medical School of Southeast University, Hubei Road 45, Huashan District, Ma'anshan 243099, Anhui Province, China
| | - Xiaojuan Zhang
- Department of Nephrology, Jinling Hospital Affiliated to Nanjing University, Zhongshan East Road 305, Xuanwu District, Nanjing 210008, Jiangsu Province, China
| | - Yu Zhang
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing 210009, Jiangsu Province, China
| | - Li Li
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing 210009, Jiangsu Province, China
| | - Pingsheng Chen
- Department of Pathology and Pathophysiology, Medical School, Southeast University, Dingjiaqiao 87, Gulou District, Nanjing 210009, Jiangsu Province, China
| |
Collapse
|
18
|
Silencing RNA for MMPs May Be Utilized for Cardioprotection. Cardiovasc Ther 2022; 2022:9729018. [PMID: 36082193 PMCID: PMC9433229 DOI: 10.1155/2022/9729018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 07/25/2022] [Accepted: 08/03/2022] [Indexed: 11/17/2022] Open
Abstract
Ischemia/reperfusion (I/R) injury is accompanied by an increase of matrix metalloproteinase 2 (MMP-2) activity, which degrades heart contractile proteins. The aim of the study was to investigate the effect of MMP-2 small interfering RNA (MMP-2 siRNA) administration on I/R heart. Isolated rat hearts perfused by the Langendorff method were subjected to I/R in the presence or absence of MMP-2 siRNA. The hemodynamic parameters of heart function were monitored. Lactate dehydrogenase (LDH) activity was measured in coronary effluents. Activity and concentration of MMPs in the hearts were measured. Concentration of troponin I (TnI) in coronary effluents was examined as a target for MMP-2 degradation. Recovery of heart mechanical function was reduced after I/R; however, administration of MMP-2 siRNA resulted in restoration of proper mechanical function (p < 0.001). LDH activity was decreased after the use of MMP-2 siRNA (p = 0.02), providing evidence for reduced cardiac damage. Both MMP-2 and MMP-9 syntheses as well as their activity were inhibited in the I/R hearts after siRNA administration (p < 0.05). MMP-2 siRNA administration inhibited TnI release into the coronary effluents (p < 0.001). The use of MMP-2 siRNA contributed to the improvement of heart mechanical function and reduction of contractile proteins degradation during I/R; therefore, MMP-2 siRNA may be considered a cardioprotective agent.
Collapse
|
19
|
Novel Roles of MT1-MMP and MMP-2: Beyond the Extracellular Milieu. Int J Mol Sci 2022; 23:ijms23179513. [PMID: 36076910 PMCID: PMC9455801 DOI: 10.3390/ijms23179513] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/17/2022] [Accepted: 08/21/2022] [Indexed: 12/14/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are critical enzymes involved in a variety of cellular processes. MMPs are well known for their ability to degrade the extracellular matrix (ECM) and their extracellular role in cell migration. Recently, more research has been conducted on investigating novel subcellular localizations of MMPs and their intracellular roles at their respective locations. In this review article, we focus on the subcellular localization and novel intracellular roles of two closely related MMPs: membrane-type-1 matrix metalloproteinase (MT1-MMP) and matrix metalloproteinase-2 (MMP-2). Although MT1-MMP is commonly known to localize on the cell surface, the protease also localizes to the cytoplasm, caveolae, Golgi, cytoskeleton, centrosome, and nucleus. At these subcellular locations, MT1-MMP functions in cell migration, macrophage metabolism, invadopodia development, spindle formation and gene expression, respectively. Similar to MT1-MMP, MMP-2 localizes to the caveolae, mitochondria, cytoskeleton, nucleus and nucleolus and functions in calcium regulation, contractile dysfunction, gene expression and ribosomal RNA transcription. Our particular interest lies in the roles MMP-2 and MT1-MMP serve within the nucleus, as they may provide critical insights into cancer epigenetics and tumor migration and invasion. We suggest that targeting nuclear MT1-MMP or MMP-2 to reduce or halt cell proliferation and migration may lead to the development of new therapies for cancer and other diseases.
Collapse
|
20
|
de Almeida LGN, Thode H, Eslambolchi Y, Chopra S, Young D, Gill S, Devel L, Dufour A. Matrix Metalloproteinases: From Molecular Mechanisms to Physiology, Pathophysiology, and Pharmacology. Pharmacol Rev 2022; 74:712-768. [PMID: 35738680 DOI: 10.1124/pharmrev.121.000349] [Citation(s) in RCA: 143] [Impact Index Per Article: 47.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The first matrix metalloproteinase (MMP) was discovered in 1962 from the tail of a tadpole by its ability to degrade collagen. As their name suggests, matrix metalloproteinases are proteases capable of remodeling the extracellular matrix. More recently, MMPs have been demonstrated to play numerous additional biologic roles in cell signaling, immune regulation, and transcriptional control, all of which are unrelated to the degradation of the extracellular matrix. In this review, we will present milestones and major discoveries of MMP research, including various clinical trials for the use of MMP inhibitors. We will discuss the reasons behind the failures of most MMP inhibitors for the treatment of cancer and inflammatory diseases. There are still misconceptions about the pathophysiological roles of MMPs and the best strategies to inhibit their detrimental functions. This review aims to discuss MMPs in preclinical models and human pathologies. We will discuss new biochemical tools to track their proteolytic activity in vivo and ex vivo, in addition to future pharmacological alternatives to inhibit their detrimental functions in diseases. SIGNIFICANCE STATEMENT: Matrix metalloproteinases (MMPs) have been implicated in most inflammatory, autoimmune, cancers, and pathogen-mediated diseases. Initially overlooked, MMP contributions can be both beneficial and detrimental in disease progression and resolution. Thousands of MMP substrates have been suggested, and a few hundred have been validated. After more than 60 years of MMP research, there remain intriguing enigmas to solve regarding their biological functions in diseases.
Collapse
Affiliation(s)
- Luiz G N de Almeida
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Hayley Thode
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Yekta Eslambolchi
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sameeksha Chopra
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Daniel Young
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Sean Gill
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Laurent Devel
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| | - Antoine Dufour
- Departments of Physiology and Pharmacology and Biochemistry and Molecular Biology, University of Calgary, Calgary, Canada (L.G.N.d.A., Y.E., S.C., D.Y., A.D.); Department of Physiology and Pharmacology, University of Western Ontario, London, Canada (S.G., H.T.); and Université Paris-Saclay, CEA, INRAE, Medicaments et Technologies pour la Santé, Gif-sur-Yvette, France (L.D.)
| |
Collapse
|
21
|
Ambardekar AV, Weiser-Evans MCM, McKinsey TA. Arterial wall rejuvenation: the potential of targeting MMP2 to treat vascular aging. Cardiovasc Res 2022; 118:2229-2230. [PMID: 35512358 DOI: 10.1093/cvr/cvac076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Indexed: 11/12/2022] Open
Affiliation(s)
- Amrut V Ambardekar
- Department of Medicine, Divisions of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045-0508.,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-0508
| | - Mary C M Weiser-Evans
- Department of Medicine, Division of Renal Diseases and Hypertension, University of Colorado Anschutz Medical Campus, Aurora, CO 80045-0508.,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-0508
| | - Timothy A McKinsey
- Department of Medicine, Divisions of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045-0508.,Consortium for Fibrosis Research & Translation, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045-0508
| |
Collapse
|
22
|
Hassoun R, Budde H, Zhazykbayeva S, Herwig M, Sieme M, Delalat S, Mostafi N, Gömöri K, Tangos M, Jarkas M, Pabel S, Bruckmüller S, Skrygan M, Lódi M, Jaquet K, Sequeira V, Gambichler T, Remedios CD, Kovács Á, Mannherz HG, Mügge A, Sossalla S, Hamdani N. Do they come together? Protein quality control, stress-activated signaling, and "sarcostat" in hypertrophic cardiomyopathy progression. Int J Cardiol 2022; 347:44-45. [PMID: 34767897 DOI: 10.1016/j.ijcard.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/04/2021] [Indexed: 11/29/2022]
Affiliation(s)
- Roua Hassoun
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Heidi Budde
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Saltanat Zhazykbayeva
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Melissa Herwig
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Marcel Sieme
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Simin Delalat
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Nusratul Mostafi
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Kamilla Gömöri
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Melina Tangos
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Muhammad Jarkas
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany
| | - Steffen Pabel
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany.
| | - Stefanie Bruckmüller
- Department of Dermatology, Skin Cancer Center, Ruhr University Bochum, Bochum, Germany.
| | - Marina Skrygan
- Department of Dermatology, Skin Cancer Center, Ruhr University Bochum, Bochum, Germany.
| | - Mária Lódi
- Department of Neuroanatomy and Molecular Brain Research, Ruhr University Bochum, Medical Faculty, Bochum, Germany.
| | - Kornelia Jaquet
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Vasco Sequeira
- Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Germany.
| | - Thilo Gambichler
- Department of Dermatology, Skin Cancer Center, Ruhr University Bochum, Bochum, Germany.
| | - Cris Dos Remedios
- Molecular Biophysics, Faculty of Medicine and Health, Victor Chang Cardiac Research Institute, Darlinghurst, Australia.
| | - Árpád Kovács
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Hans Georg Mannherz
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Anatomy and Molecular Embryology, Ruhr University Bochum, Bochum, Germany.
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| | - Samuel Sossalla
- Department of Internal Medicine II, University Medical Center Regensburg, Regensburg, Germany; Clinic for Cardiology & Pneumology, Georg-August University Goettingen, and DZHK (German Centre for Cardiovascular Research), partner site Goettingen, Germany.
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany; Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, Bochum, Germany.
| |
Collapse
|
23
|
Chalise U, Becirovic-Agic M, Lindsey ML. Neutrophil crosstalk during cardiac wound healing after myocardial infarction. CURRENT OPINION IN PHYSIOLOGY 2021; 24:100485. [PMID: 35664861 PMCID: PMC9159545 DOI: 10.1016/j.cophys.2022.100485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Myocardial infarction (MI) initiates an intense inflammatory response that induces neutrophil infiltration into the infarct region. Neutrophils commence the pro-inflammatory response that includes upregulation of cytokines and chemokines (e.g., interleukin-1 beta) and degranulation of pre-formed proteases (e.g., matrix metalloproteinases -8 and -9) that degrade existing extracellular matrix to clear necrotic tissue. An increase or complete depletion of neutrophils both paradoxically impair MI resolution, indicating a complex role of neutrophils in cardiac wound healing. Following pro-inflammation, the neutrophil shifts to a reparative phenotype that promotes inflammation resolution and aids in scar formation. Across the shifts in phenotype, the neutrophil communicates with other cells to coordinate repair and scar formation. This review summarizes our current understanding of neutrophil crosstalk with cardiomyocytes and macrophages during MI wound healing.
Collapse
Affiliation(s)
- Upendra Chalise
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198; and Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105
| | - Mediha Becirovic-Agic
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198; and Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105
| | - Merry L. Lindsey
- Department of Cellular and Integrative Physiology, Center for Heart and Vascular Research, University of Nebraska Medical Center, Omaha, NE 68198; and Research Service, Nebraska-Western Iowa Health Care System, Omaha, NE 68105
| |
Collapse
|
24
|
Zhu Y. Metalloproteases in gonad formation and ovulation. Gen Comp Endocrinol 2021; 314:113924. [PMID: 34606745 PMCID: PMC8576836 DOI: 10.1016/j.ygcen.2021.113924] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 01/13/2023]
Abstract
Changes in expression or activation of various metalloproteases including matrix metalloproteases (Mmp), a disintegrin and metalloprotease (Adam) and a disintegrin and metalloprotease with thrombospondin motif (Adamts), and their endogenous inhibitors (tissue inhibitors of metalloproteases, Timp), have been shown to be critical for ovulation in various species from studies in past decades. Some of these metalloproteases such as Adamts1, Adamts9, Mmp2, and Mmp9 have also been shown to be regulated by luteinizing hormone (LH) and/or progestin, which are essential triggers for ovulation in all vertebrate species. Most of these metalloproteases also express broadly in various tissues and cells including germ cells and somatic gonad cells. Thus, metalloproteases likely play roles in gonad formation processes comprising primordial germ cell (PGC) migration, development of germ and somatic cells, and sex determination. However, our knowledge on the functions and mechanisms of metalloproteases in these processes in vertebrates is still lacking. This review will summarize our current knowledge on the metalloproteases in ovulation and gonad formation with emphasis on PGC migration and germ cell development.
Collapse
Affiliation(s)
- Yong Zhu
- Department of Biology, East Carolina University, Greenville, NC 27858, USA.
| |
Collapse
|
25
|
Wang X, Fan H, Wang Y, Yin X, Liu G, Gao C, Li X, Liang B. Elevated Peripheral T Helper Cells Are Associated With Atrial Fibrillation in Patients With Rheumatoid Arthritis. Front Immunol 2021; 12:744254. [PMID: 34721413 PMCID: PMC8554094 DOI: 10.3389/fimmu.2021.744254] [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: 07/20/2021] [Accepted: 09/29/2021] [Indexed: 11/23/2022] Open
Abstract
Patients with rheumatoid arthritis (RA) have a significantly high risk of atrial fibrillation (AF). This study aimed to compare the absolute and relative changes in peripheral T cells in patients with RA who were also affected with and without AF. To help make an early diagnosis and prevent the initiation and progression of AF, the changes in the lymphocyte subsets were assessed in RA patients with and without AF. A propensity score matching (PSM) system (1:3) was used to perform a matched case-control study with 40 RA-AF cases and 120 RA controls. Changes in the erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), anti-citrullinated peptide antibody (ACPA), and rheumatoid factor (RF) were examined. The percentage and absolute number of T, B, natural killer (NK), T helper (Th)1, Th2, Th17, and T-regulatory (Treg) cells in the peripheral blood of patients with and without RA-AF were determined using flow cytometry. Univariate and multivariate analyses were performed to determine the association between peripheral lymphocytes and RA-AF. Demographic data, ESR, CRP, ACPA, and the percentage, as well as the absolute value of B, NK, Th2, and Treg cells, showed no significant differences between the propensity score-matched groups of RA and RA-AF. Meanwhile, the absolute number and percentage of Th1 cells, the absolute number of Th17 cells, the ratio of Th1/Treg, Th17/Treg, and RF were significantly higher in patients with RA-AF than those in the control groups (P < 0.05). Univariate and multivariate logistic regression analyses also revealed that the percentage of Th1 cells, the absolute number of Th17 cells, and the ratio of Th1/Treg were associated with a significantly higher risk of AF. This PSM study demonstrated that the incidence of AF was higher in RA patients with Th cell immunological derangements.
Collapse
Affiliation(s)
- Xin Wang
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Hongxuan Fan
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Yongle Wang
- Department of Neurology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xufang Yin
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Guangying Liu
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Chong Gao
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States
| | - Xiaofeng Li
- Department of Rheumatology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Bin Liang
- Department of Cardiology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
26
|
Yu W, Ke X, Li M, Ye P, Peng J, Li H. Downregulation of RAB7 and Caveolin-1 increases MMP-2 activity in renal tubular epithelial cells under hypoxic conditions. Open Med (Wars) 2021; 16:1428-1437. [PMID: 34676302 PMCID: PMC8483063 DOI: 10.1515/med-2021-0341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 07/21/2021] [Accepted: 08/12/2021] [Indexed: 12/24/2022] Open
Abstract
Tubulointerstitial fibrosis leads to tubular basement membrane thickening and accumulation of interstitial extracellular matrix (ECM). Matrix metallopeptidase-2 (MMP-2) is involved in the breakdown of ECM. Chronic hypoxia often occurs in the kidney tissues of patients with chronic kidney disease. Our previous study reported the effect of autophagy and endocytosis on MMP-2 activity in hypoxia-treated HK-2 cells. In this study, the relationship between the Ras-related protein Rab-7a (RAB7) and MMP-2 activity was further investigated. RAB7 overexpression decreased MMP-2 activity. In contrast, the results for RAB7 knockdown displayed the opposite pattern. Short hairpin RNA technology was used to knockdown Caveolin-1 (Cav-1) or Beclin-1 (Bec-1) in HK-2 cells. The two genes displayed differential effects on MMP-2 activity. Cav-1 and RAB7 interference increased MMP-2 activity. This study suggested that autophagy and endocytosis, RAB7, Cav-1, and Bec-1 may serve as potential mediators for altered MMP-2 activity.
Collapse
Affiliation(s)
- Wenmin Yu
- The School of Basic Medical Science, Jiujiang University, 320 XunYang East Road, Jiujiang 332000, Jiangxi, People's Republic of China
| | - Xiumei Ke
- The School of Basic Medical Science, Jiujiang University, 320 XunYang East Road, Jiujiang 332000, Jiangxi, People's Republic of China
| | - Meiren Li
- Jiujiang University Clinic College/Hospital, Jiujiang University, Jiangxi, People's Republic of China
| | - Ping Ye
- Jiujiang University Clinic College/Hospital, Jiujiang University, Jiangxi, People's Republic of China
| | - Jing Peng
- Jiujiang University Clinic College/Hospital, Jiujiang University, Jiangxi, People's Republic of China
| | - Huimin Li
- The School of Basic Medical Science, Jiujiang University, 320 XunYang East Road, Jiujiang 332000, Jiangxi, People's Republic of China
| |
Collapse
|
27
|
McNair ED, Bezaire J, Moser M, Mondal P, Conacher J, Franczak A, Sawicki G, Reid D, Khani-Hanjani A. The Association of Matrix Metalloproteinases With Acute Kidney Injury Following CPB-Supported Cardiac Surgery. Can J Kidney Health Dis 2021; 8:20543581211019640. [PMID: 34350005 PMCID: PMC8287351 DOI: 10.1177/20543581211019640] [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] [Received: 09/02/2020] [Accepted: 04/19/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Cardiac surgery-associated acute kidney injury (AKI) is an adverse outcome that increases morbidity and mortality in patients undergoing cardiac surgical procedures. To date, the use of serum creatinine levels as an early indicator of AKI has limitations because of its slow rise and poor predictive accuracy for renal injury. This delay in diagnosis may lead to prolonged initiation in treatment and increased risk for adverse outcomes. OBJECTIVE This pilot study explores serum and urine matrix metalloproteinases (MMPs)-2 and MMP-9 and their association, and potentially earlier detection of AKI in patients following cardiopulmonary bypass (CPB)-supported cardiac surgery. We hypothesize that increased activity of serum and urine levels MMP-2 and/ or MMP-9 are associated with AKI. Furthermore, MMP-2 and/ or MMP-9 may provide earlier identification of AKI as compared with serum levels of creatinine. METHODS During the study period, there were 150 CPB-supported surgeries, 21 of which developed AKI according to the Kidney Disease Improving Global Outcomes criteria. We then selected a sample of 21 matched cases from those patients who went through the surgery without developing AKI. Primary outcomes were the measurement via gel zymography of the serum and urine activity of MMP-2 and MMP-9 drawn at the following intervals: pre-CPB; 10-minute post-CPB; and 4-hour post-CPB time points. Secondary variables were the measurement of serum creatinine, intensive care unit (ICU) fluid balance, and length of ICU stay. RESULTS At the 10-minute and 4-hour post-CPB time points, the serum MMP-2 activity of AKI patients were significantly higher as compared with non-AKI patients (P < .001 and P = .004), respectively. Similarly, at the 10-minute and 4-hour post-CPB time points, the serum MMP-9 activity of AKI patients was significantly higher as compared with non-AKI patients (P = .001 and P = .014), respectively. The activity of urine MMP-2 and MMP-9 of AKI patients was significantly higher as compared with non-AKI patients at all 3 time points (P = .004, P < .001, P < .001), respectively. CONCLUSION Although the pilot study may have limitations, it has demonstrated that the serum and urine levels of activity of MMP-2 and MMP-9 are associated with the clinical endpoint of AKI and appear to have earlier rising levels as compared with those of serum creatinine. Furthermore, in depth, exploration is underway with a larger sample size to attempt validation of the analytical performance and reproducibility of the assay for MMP-2 and MMP-9 to aid in earlier diagnosis of AKI following CPB-supported cardiac surgery.
Collapse
Affiliation(s)
- Erick D. McNair
- Department of Pathology and Laboratory
Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada
- Department of Surgery/Division of
Cardiac Surgery, College of Medicine, University of Saskatchewan, Saskatoon,
Canada
| | - Jennifer Bezaire
- Department of Pathology and Laboratory
Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Michael Moser
- Department of Medicine, College of
Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Prosanta Mondal
- Department of Community Health and
Epidemiology, College of Medicine, University of Saskatchewan, Saskatoon,
Canada
| | - Josie Conacher
- Department of Pathology and Laboratory
Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Aleksandra Franczak
- Department of Medicine, College of
Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Greg Sawicki
- Department of Pharmacology, College of
Medicine, University of Saskatchewan, Saskatoon, Canada
| | - David Reid
- Department of Medicine, College of
Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Abass Khani-Hanjani
- Department of Surgery/Division of
Cardiac Surgery, College of Medicine, University of Saskatchewan, Saskatoon,
Canada
| |
Collapse
|
28
|
Adewale AO, Ahn YH. Titin N2A Domain and Its Interactions at the Sarcomere. Int J Mol Sci 2021; 22:ijms22147563. [PMID: 34299183 PMCID: PMC8305307 DOI: 10.3390/ijms22147563] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/09/2021] [Accepted: 07/12/2021] [Indexed: 12/16/2022] Open
Abstract
Titin is a giant protein in the sarcomere that plays an essential role in muscle contraction with actin and myosin filaments. However, its utility goes beyond mechanical functions, extending to versatile and complex roles in sarcomere organization and maintenance, passive force, mechanosensing, and signaling. Titin’s multiple functions are in part attributed to its large size and modular structures that interact with a myriad of protein partners. Among titin’s domains, the N2A element is one of titin’s unique segments that contributes to titin’s functions in compliance, contraction, structural stability, and signaling via protein–protein interactions with actin filament, chaperones, stress-sensing proteins, and proteases. Considering the significance of N2A, this review highlights structural conformations of N2A, its predisposition for protein–protein interactions, and its multiple interacting protein partners that allow the modulation of titin’s biological effects. Lastly, the nature of N2A for interactions with chaperones and proteases is included, presenting it as an important node that impacts titin’s structural and functional integrity.
Collapse
|
29
|
Cruz JDO, Silva AO, Ribeiro JM, Luizon MR, Ceron CS. Epigenetic Regulation of the N-Terminal Truncated Isoform of Matrix Metalloproteinase-2 (NTT-MMP-2) and Its Presence in Renal and Cardiac Diseases. Front Genet 2021; 12:637148. [PMID: 33732288 PMCID: PMC7959838 DOI: 10.3389/fgene.2021.637148] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/26/2021] [Indexed: 12/19/2022] Open
Abstract
Several clinical and experimental studies have documented a compelling and critical role for the full-length matrix metalloproteinase-2 (FL-MMP-2) in ischemic renal injury, progressive renal fibrosis, and diabetic nephropathy. A novel N-terminal truncated isoform of MMP-2 (NTT-MMP-2) was recently discovered, which is induced by hypoxia and oxidative stress by the activation of a latent promoter located in the first intron of the MMP2 gene. This NTT-MMP-2 isoform is enzymatically active but remains intracellular in or near the mitochondria. In this perspective article, we first present the findings about the discovery of the NTT-MMP-2 isoform, and its functional and structural differences as compared with the FL-MMP-2 isoform. Based on publicly available epigenomics data from the Encyclopedia of DNA Elements (ENCODE) project, we provide insights into the epigenetic regulation of the latent promoter located in the first intron of the MMP2 gene, which support the activation of the NTT-MMP-2 isoform. We then focus on its functional assessment by covering the alterations found in the kidney of transgenic mice expressing the NTT-MMP-2 isoform. Next, we highlight recent findings regarding the presence of the NTT-MMP-2 isoform in renal dysfunction, in kidney and cardiac diseases, including damage observed in aging, acute ischemia-reperfusion injury (IRI), chronic kidney disease, diabetic nephropathy, and human renal transplants with delayed graft function. Finally, we briefly discuss how our insights may guide further experimental and clinical studies that are needed to elucidate the underlying mechanisms and the role of the NTT-MMP-2 isoform in renal dysfunction, which may help to establish it as a potential therapeutic target in kidney diseases.
Collapse
Affiliation(s)
- Juliana de Oliveira Cruz
- Graduate Program in Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Alessandra O Silva
- Department of Food and Drugs, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Jessyca M Ribeiro
- Department of Food and Drugs, Faculty of Pharmaceutical Sciences, Federal University of Alfenas, Alfenas, Brazil
| | - Marcelo R Luizon
- Graduate Program in Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil.,Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Carla S Ceron
- Department of Biological Sciences, Institute of Exact and Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Brazil
| |
Collapse
|
30
|
Kirkham AA, Pituskin E, Thompson RB, Mackey JR, Koshman SL, Jassal D, Pitz M, Haykowsky MJ, Pagano JJ, Chow K, Tsui AK, Ezekowitz JA, Oudit GY, Paterson DI. Cardiac and Cardiometabolic Phenotyping of Trastuzumab-Mediated Cardiotoxicity: a Secondary Analysis of the MANTICORE trial. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2021; 8:130-139. [PMID: 33605416 DOI: 10.1093/ehjcvp/pvab016] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 10/26/2020] [Accepted: 02/17/2021] [Indexed: 01/02/2023]
Abstract
AIMS An improved understanding of the pathophysiology of trastuzumab-mediated cardiotoxicity is required to improve outcomes of patients with HER2-positive breast cancer. We aimed to characterize the cardiac and cardiometabolic phenotype of trastuzumab-mediated toxicity and potential interactions with cardiac pharmacotherapy. METHODS AND RESULTS This study was an analysis of serial magnetic resonance imaging (MRI) and circulating biomarker data acquired from patients with HER2-positive early stage breast cancer participating in a randomized-controlled clinical trial for the pharmaco-prevention of trastuzumab-associated cardiotoxicity. Circulating biomarkers (B-type natriuretic peptide, troponin I, MMP-2 and -9, GDF-15, neuregulin-1 and IGF-1) and MRI of cardiac structure and function and abdominal fat distribution were acquired prior to trastuzumab, post-cycle 4 and post-cycle 17. Ninety-four participants (51±8 years) completed the study with 30 on placebo, 33 on perindopril, and 31 on bisoprolol. Post-cycle 4, global longitudinal strain (GLS) deteriorated from baseline in both placebo (+2.0±2.7%, p = 0.002) and perindopril (+0.9±2.5%, p = 0.04), but not with bisoprolol (-0.2±2.1%, p = 0.55). In all groups combined, extracellular volume fraction and GDF-15 increased post-cycle 4 (+1.3±4.4%, p = 0.004; +130±150%, p ≤ 0.001, respectively). However, no significant change in troponin I was detected throughout trastuzumab. In all groups combined, visceral and intermuscular fat volume increased post-cycle 4 (+7±17%, p = 0.02, +8±23%, p = 0.02, respectively), while muscle volume and IGF-1 decreased from post-cycle 4 to 17 (-2±10%, p = 0.008, -18±28%, p < 0.001, respectively). CONCLUSION Trastuzumab results in impaired cardiac function and early myocardial inflammation. Trastuzumab is also associated with deleterious changes to the cardiometabolic phenotype which may contribute to the increased cardiovascular risk in this population.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Kelvin Chow
- Cardiovascular MR R&D, Siemens Medical Solutions USA, Chicago, Illinois
| | | | | | | | | |
Collapse
|
31
|
Mollace V, Rosano GMC, Anker SD, Coats AJS, Seferovic P, Mollace R, Tavernese A, Gliozzi M, Musolino V, Carresi C, Maiuolo J, Macrì R, Bosco F, Chiocchi M, Romeo F, Metra M, Volterrani M. Pathophysiological Basis for Nutraceutical Supplementation in Heart Failure: A Comprehensive Review. Nutrients 2021; 13:257. [PMID: 33477388 PMCID: PMC7829856 DOI: 10.3390/nu13010257] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/25/2020] [Accepted: 01/13/2021] [Indexed: 02/06/2023] Open
Abstract
There is evidence demonstrating that heart failure (HF) occurs in 1-2% of the global population and is often accompanied by comorbidities which contribute to increasing the prevalence of the disease, the rate of hospitalization and the mortality. Although recent advances in both pharmacological and non-pharmacological approaches have led to a significant improvement in clinical outcomes in patients affected by HF, residual unmet needs remain, mostly related to the occurrence of poorly defined strategies in the early stages of myocardial dysfunction. Nutritional support in patients developing HF and nutraceutical supplementation have recently been shown to possibly contribute to protection of the failing myocardium, although their place in the treatment of HF requires further assessment, in order to find better therapeutic solutions. In this context, the Optimal Nutraceutical Supplementation in Heart Failure (ONUS-HF) working group aimed to assess the optimal nutraceutical approach to HF in the early phases of the disease, in order to counteract selected pathways that are imbalanced in the failing myocardium. In particular, we reviewed several of the most relevant pathophysiological and molecular changes occurring during the early stages of myocardial dysfunction. These include mitochondrial and sarcoplasmic reticulum stress, insufficient nitric oxide (NO) release, impaired cardiac stem cell mobilization and an imbalanced regulation of metalloproteinases. Moreover, we reviewed the potential of the nutraceutical supplementation of several natural products, such as coenzyme Q10 (CoQ10), a grape seed extract, Olea Europea L.-related antioxidants, a sodium-glucose cotransporter (SGLT2) inhibitor-rich apple extract and a bergamot polyphenolic fraction, in addition to their support in cardiomyocyte protection, in HF. Such an approach should contribute to optimising the use of nutraceuticals in HF, and the effect needs to be confirmed by means of more targeted clinical trials exploring the efficacy and safety of these compounds.
Collapse
Affiliation(s)
- Vincenzo Mollace
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Giuseppe M. C. Rosano
- Cardiology Clinical Academic Group, St George’s Hospitals NHS Trust University of London, London SW17 0QT, UK;
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (A.J.S.C.); (M.V.)
| | - Stefan D. Anker
- Department of Cardiology, Charité–Universitätsmedizin Berlin, 10117 Berlin, Germany;
| | - Andrew J. S. Coats
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (A.J.S.C.); (M.V.)
| | - Petar Seferovic
- Faculty of Medicine, Belgrade University, 11000 Belgrade, Serbia;
| | - Rocco Mollace
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
- Department of Experimental and Applied Medicine, Institute of Cardiology, University of Brescia, 25121 Brescia, Italy;
| | - Annamaria Tavernese
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
- Department of Experimental and Applied Medicine, Institute of Cardiology, University of Brescia, 25121 Brescia, Italy;
| | - Micaela Gliozzi
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Vincenzo Musolino
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Cristina Carresi
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Jessica Maiuolo
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Roberta Macrì
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Francesca Bosco
- Department of Health Sciences, Institute of Research for Food Safety & Health, University “Magna Graecia” of Catanzaro, 88100 Catanzaro, Italy; (R.M.); (A.T.); (M.G.); (V.M.); (C.C.); (J.M.); (R.M.); (F.B.)
| | - Marcello Chiocchi
- Department of Diagnostic Imaging and Interventional Radiology, Policlinico Tor Vergata, 00199 Rome, Italy;
| | - Francesco Romeo
- Department of Experimental Medicine, University of Rome “Tor Vergata”, 00199 Rome, Italy;
| | - Marco Metra
- Department of Experimental and Applied Medicine, Institute of Cardiology, University of Brescia, 25121 Brescia, Italy;
| | - Maurizio Volterrani
- Department of Cardiology, IRCCS San Raffaele Pisana, 00166 Rome, Italy; (A.J.S.C.); (M.V.)
| |
Collapse
|
32
|
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: 718] [Impact Index Per Article: 143.6] [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
|
33
|
Owolabi US, Amraotkar AR, Coulter AR, Singam NSV, Aladili BN, Singh A, Trainor PJ, Mitra R, DeFilippis AP. Change in matrix metalloproteinase 2, 3, and 9 levels at the time of and after acute atherothrombotic myocardial infarction. J Thromb Thrombolysis 2020; 49:235-244. [PMID: 31808123 DOI: 10.1007/s11239-019-02004-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Elevated measures of matrix metalloproteinases (MMPs) are associated with acute myocardial infarction (MI), but it is not known how long these changes persist post-MI or if these measures differ between atherothrombotic versus non-atherothrombotic MI. MMPs-2, 3, and 9 were measured in 80 subjects with acute MI (atherothrombotic and non-atherothrombotic MI) or stable coronary artery disease (CAD). Measurements were made at, the time of acute MI, and > 3-month following acute MI (quiescent phase). Outcome measures were compared between groups and between time of acute MI and quiescent post-MI follow-up using Wilcoxon's and repeated measures analysis of variance. Forty-nine subjects met the criteria for acute MI with clearly defined atherothrombotic (n = 22) and non-atherothrombotic (n = 12) subsets. Fifteen subjects met criteria for stable CAD. MMP-3 was higher in acute MI versus stable CAD subjects at the time of acute MI: (453 vs. 217 pg/mL, p = 0.010) but not at quiescent phase follow-up (p > 0.05). MMP-9 was higher in acute MI versus stable CAD subjects at the time of acute MI: (412 vs. 168 pg/mL, p = 0.002) but not at the quiescent phase follow-up (p > 0.05). MMP-9 was higher at the time of acute MI versus quiescent phase follow-up in acute MI (412 vs. 213 pg/mL, p = 0.001) and atherothrombotic MI specifically (458 vs. 212 pg/mL, p = 0.001). No difference in MMP-2, 3, or 9 was observed between atherothrombotic versus non-atherothrombotic MI subgroups. MMPs-3 and 9 are significantly elevated in acute MI verses stable CAD subjects at time of acute MI but not different at quiescent phase follow-up. MMP-9 is elevated at the time of acute MI and specifically in acute atherothrombotic MI at time of MI versus quiescent phase follow-up.
Collapse
Affiliation(s)
- Ugochukwu Shola Owolabi
- Diabetes & Obesity Center, University of Louisville, Louisville, KY, USA.,Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, KY, USA
| | - Alok Ravindra Amraotkar
- Diabetes & Obesity Center, University of Louisville, Louisville, KY, USA.,Division of Cardiovascular Medicine, University of Louisville, Louisville, KY, USA
| | - Amanda R Coulter
- Diabetes & Obesity Center, University of Louisville, Louisville, KY, USA.,Division of Cardiovascular Medicine, University of Louisville, Louisville, KY, USA
| | | | - Bahjat N Aladili
- Division of Cardiovascular Medicine, University of Louisville, Louisville, KY, USA
| | - Ayesha Singh
- School of Medicine, University of Louisville, Louisville, KY, USA
| | - Patrick James Trainor
- Division of Cardiovascular Medicine, University of Louisville, Louisville, KY, USA.,Applied Statistics, EASIB Department, New Mexico State University, Las Cruces, NM, USA
| | - Riten Mitra
- Department of Bioinformatics and Biostatistics, University of Louisville, Louisville, KY, USA
| | - Andrew Paul DeFilippis
- Division of Cardiovascular Medicine, University of Louisville, Louisville, KY, USA. .,Ciccarone Center for the Prevention of Heart Disease, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
34
|
Gömöri K, Szabados T, Kenyeres É, Pipis J, Földesi I, Siska A, Dormán G, Ferdinandy P, Görbe A, Bencsik P. Cardioprotective Effect of Novel Matrix Metalloproteinase Inhibitors. Int J Mol Sci 2020; 21:ijms21196990. [PMID: 32977437 PMCID: PMC7582346 DOI: 10.3390/ijms21196990] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Accepted: 09/15/2020] [Indexed: 12/11/2022] Open
Abstract
Background: We recently developed novel matrix metalloproteinase-2 (MMP-2) inhibitor small molecules for cardioprotection against ischemia/reperfusion injury and validated their efficacy in ischemia/reperfusion injury in cardiac myocytes. The aim of the present study was to test our lead compounds for cardioprotection in vivo in a rat model of acute myocardial infarction (AMI) in the presence or absence of hypercholesterolemia, one of the major comorbidities affecting cardioprotection. Methods: Normocholesterolemic adult male Wistar rats were subjected to 30 min of coronary occlusion followed by 120 min of reperfusion to induce AMI. MMP inhibitors (MMPI)-1154 and -1260 at 0.3, 1, and 3 µmol/kg, MMPI-1248 at 1, 3, and 10 µmol/kg were administered at the 25th min of ischemia intravenously. In separate groups, hypercholesterolemia was induced by a 12-week diet (2% cholesterol, 0.25% cholic acid), then the rats were subjected to the same AMI protocol and single doses of the MMPIs that showed the most efficacy in normocholesterolemic animals were tested in the hypercholesterolemic animals. Infarct size/area at risk was assessed at the end of reperfusion in all groups by standard Evans blue and 2,3,5-triphenyltetrazolium chloride (TTC) staining, and myocardial microvascular obstruction (MVO) was determined by thioflavine-S staining. Results: MMPI-1154 at 1 µmol/kg, MMPI-1260 at 3 µmol/kg and ischemic preconditioning (IPC) as the positive control reduced infarct size significantly; however, this effect was not seen in hypercholesterolemic animals. MVO in hypercholesterolemic animals decreased by IPC only. Conclusions: This is the first demonstration that MMPI-1154 and MMPI-1260 showed a dose-dependent infarct size reduction in an in vivo rat AMI model; however, single doses that showed the most efficacy in normocholesterolemic animals were abolished by hypercholesterolemia. The further development of these promising cardioprotective MMPIs should be continued with different dose ranges in the study of hypercholesterolemia and other comorbidities.
Collapse
Affiliation(s)
- Kamilla Gömöri
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (K.G.); (T.S.); (É.K.); (A.G.)
| | - Tamara Szabados
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (K.G.); (T.S.); (É.K.); (A.G.)
| | - Éva Kenyeres
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (K.G.); (T.S.); (É.K.); (A.G.)
| | - Judit Pipis
- Pharmahungary Group, H-6722 Szeged, Hungary; (J.P.); (P.F.)
| | - Imre Földesi
- Department of Laboratory Medicine, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (I.F.); (A.S.)
| | - Andrea Siska
- Department of Laboratory Medicine, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (I.F.); (A.S.)
| | | | - Péter Ferdinandy
- Pharmahungary Group, H-6722 Szeged, Hungary; (J.P.); (P.F.)
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, H-1089 Budapest, Hungary
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (K.G.); (T.S.); (É.K.); (A.G.)
- Pharmahungary Group, H-6722 Szeged, Hungary; (J.P.); (P.F.)
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, H-1089 Budapest, Hungary
| | - Péter Bencsik
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, H-6720 Szeged, Hungary; (K.G.); (T.S.); (É.K.); (A.G.)
- Pharmahungary Group, H-6722 Szeged, Hungary; (J.P.); (P.F.)
- Correspondence: ; Tel.: +36-30-212-3469
| |
Collapse
|
35
|
Muraoka H, Hasegawa K, Sakamaki Y, Minakuchi H, Kawaguchi T, Yasuda I, Kanda T, Tokuyama H, Wakino S, Itoh H. Role of Nampt-Sirt6 Axis in Renal Proximal Tubules in Extracellular Matrix Deposition in Diabetic Nephropathy. Cell Rep 2020; 27:199-212.e5. [PMID: 30943401 DOI: 10.1016/j.celrep.2019.03.024] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 02/16/2019] [Accepted: 03/07/2019] [Indexed: 10/27/2022] Open
Abstract
Nicotinamide adenine dinucleotide (NAD+) metabolism plays a critical role in kidneys. We previously reported that decreased secretion of a NAD+ precursor, nicotinamide mononucleotide (NMN), from proximal tubules (PTs) can trigger diabetic albuminuria. In the present study, we investigated the role of NMN-producing enzyme nicotinamide phosphoribosyltransferase (Nampt) in diabetic nephropathy. The expression of Nampt in PTs was downregulated in streptozotocin (STZ)-treated diabetic mice when they exhibited albuminuria. This albuminuria was ameliorated in PT-specific Nampt-overexpressing transgenic (TG) mice. PT-specific Nampt-conditional knockout (Nampt CKO) mice exhibited TBM thickening and collagen deposition, which were associated with the upregulation of the profibrogenic gene TIMP-1. Nampt CKO mice also exhibited the downregulation of sirtuins, particularly in Sirt6. PT-specific Sirt6-knockout mice exhibited enhanced fibrotic phenotype resembling that of Nampt CKO mice with increased Timp1 expression. In conclusion, the Nampt-Sirt6 axis in PTs serves as a key player in fibrogenic extracellular matrix remodeling in diabetic nephropathy.
Collapse
Affiliation(s)
- Hirokazu Muraoka
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Kazuhiro Hasegawa
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Yusuke Sakamaki
- Department of Internal Medicine, Tokyo Dental College Ichikawa General Hospital, Chiba 272-8583, Japan
| | - Hitoshi Minakuchi
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Takahisa Kawaguchi
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Itaru Yasuda
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Takeshi Kanda
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Hirobumi Tokuyama
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan
| | - Shu Wakino
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan.
| | - Hiroshi Itoh
- Department of Internal Medicine, School of Medicine, Keio University, Tokyo 160-8582, Japan
| |
Collapse
|
36
|
Cassaglia P, Penas F, Betazza C, Fontana Estevez F, Miksztowicz V, Martínez Naya N, Llamosas MC, Noli Truant S, Wilensky L, Volberg V, Cevey ÁC, Touceda V, Cicale E, Berg G, Fernández M, Goren N, Morales C, González GE. Genetic Deletion of Galectin-3 Alters the Temporal Evolution of Macrophage Infiltration and Healing Affecting the Cardiac Remodeling and Function after Myocardial Infarction in Mice. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:1789-1800. [PMID: 32473918 DOI: 10.1016/j.ajpath.2020.05.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 04/22/2020] [Accepted: 05/05/2020] [Indexed: 12/13/2022]
Abstract
We studied the role of galectin-3 (Gal-3) in the expression of alternative activation markers (M2) on macrophage, cytokines, and fibrosis through the temporal evolution of healing, ventricular remodeling, and function after myocardial infarction (MI). C57BL/6J and Gal-3 knockout mice (Lgals3-/-) were subjected to permanent coronary ligation or sham. We studied i) mortality, ii) macrophage infiltration and expression of markers of alternative activation, iii) cytokine, iv) matrix metalloproteinase-2 activity, v) fibrosis, and vi) cardiac function and remodeling. At 1 week post-MI, lack of Gal-3 markedly attenuated F4/80+ macrophage infiltration and significantly increased the expression of Mrc1 and Chil1, markers of M2 macrophages at the MI zone. Levels of IL-10, IL-6, and matrix metalloproteinase-2 were significantly increased, whereas tumor necrosis factor-α, transforming growth factor-β, and fibrosis were remarkably attenuated at the infarct zone. In Gal-3 knockout mice, scar thinning ratio, expansion, and cardiac remodeling and function were severely affected from the onset of MI. At 4 weeks post-MI, the natural evolution of fibrosis in Gal-3 knockout mice was also affected. Our results suggest that Gal-3 is essential for wound healing because it regulates the dynamics of macrophage infiltration, proinflammatory and anti-inflammatory cytokine expression, and fibrosis along the temporal evolution of MI in mice. The deficit of Gal-3 affected the dynamics of wound healing, thus aggravating the evolution of remodeling and function.
Collapse
Affiliation(s)
- Pablo Cassaglia
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Federico Penas
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Celeste Betazza
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina; Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina
| | - Florencia Fontana Estevez
- Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina
| | - Verónica Miksztowicz
- Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina; Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica-INFIBIOC, Laboratorio de Lípidos y Aterosclerosis, Buenos Aires, Argentina
| | - Nadia Martínez Naya
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - María Clara Llamosas
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Sofía Noli Truant
- Facultad de Farmacia y Bioquímica-CONICET, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, Argentina
| | - Luciana Wilensky
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Verónica Volberg
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Ágata C Cevey
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Vanessa Touceda
- Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica-INFIBIOC, Laboratorio de Lípidos y Aterosclerosis, Buenos Aires, Argentina
| | - Eliana Cicale
- Facultad de Ciencias Veterinarias, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Gabriela Berg
- Facultad de Farmacia y Bioquímica, Departamento de Bioquímica Clínica-INFIBIOC, Laboratorio de Lípidos y Aterosclerosis, Buenos Aires, Argentina
| | - Marisa Fernández
- Facultad de Farmacia y Bioquímica-CONICET, Instituto de Estudios de la Inmunidad Humoral (IDEHU), Buenos Aires, Argentina
| | - Nora Goren
- Facultad de Medicina, Instituto de Investigaciones Biomédicas en Retrovirus y SIDA (INBIRS), Buenos Aires, Argentina
| | - Celina Morales
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina
| | - Germán E González
- Facultad de Medicina-CONICET, Departamento de Patología, Instituto de Fisiopatología Cardiovascular, Buenos Aires, Argentina; Facultad de Medicina, Pontificia Universidad Católica Argentina (UCA), Instituto de Investigaciones Biomédicas (UCA-CONICET), Laboratorio de Patología Cardiovascular Experimental e Hipertensi Arterial, Buenos Aires, Argentina.
| |
Collapse
|
37
|
Cardiac fibroblast activation during myocardial infarction wound healing: Fibroblast polarization after MI. Matrix Biol 2020; 91-92:109-116. [PMID: 32446909 DOI: 10.1016/j.matbio.2020.03.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 03/23/2020] [Accepted: 03/25/2020] [Indexed: 12/13/2022]
Abstract
Cardiac wound healing after myocardial infarction (MI) evolves from pro-inflammatory to anti-inflammatory to reparative responses, and the cardiac fibroblast is a central player during the entire transition. The fibroblast mirrors changes seen in the left ventricle infarct by undergoing a continuum of polarization phenotypes that follow pro-inflammatory, anti-inflammatory, and pro-scar producing profiles. The development of each phenotype transition is contingent upon the MI environment into which the fibroblast enters. In this mini-review, we summarize our current knowledge regarding cardiac fibroblast activation during MI and highlight key areas where gaps remain.
Collapse
|
38
|
Roberti SL, Higa R, Sato H, Gomez Ribot D, Capobianco E, Jawerbaum A. Olive oil supplementation prevents extracellular matrix deposition and reduces prooxidant markers and apoptosis in the offspring´s heart of diabetic rats. Reprod Toxicol 2020; 95:137-147. [PMID: 32417168 DOI: 10.1016/j.reprotox.2020.05.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 04/23/2020] [Accepted: 05/05/2020] [Indexed: 12/11/2022]
Abstract
Maternal diabetes induces fetal programming of cardiovascular diseases. Diabetes induced-cardiac fibrosis is a process that may start in utero and may be related to the prooxidant/proinflammatory environment. The aim of this study was to investigate the effect of a maternal diet enriched in olive oil on the levels of components and regulators of the extracellular matrix, on prooxidant markers and on apoptosis rate in the heart of 21-day-old offspring of diabetic rats. Maternal diabetes was induced by neonatal administration of streptozotocin. During pregnancy, diabetic and control rats were fed with diets supplemented or not with 6% olive oil. The heart of the offspring was studied at 21 days of age. We found increased deposition of collagen IV and fibronectin in the offspring´s heart of diabetic rats, which was prevented by the maternal diets enriched in olive oil. Increases in connective tissue growth factor were also prevented by the maternal diets enriched in olive oil. Prooxidant markers as well as apoptosis, which were increased in the heart of the offspring of diabetic rats, were prevented by the maternal olive oil dietary treatment. Our findings identified powerful effects of a maternal diet enriched in olive oil on the prevention of increased extracellular matrix deposition and increased prooxidant markers in the heart of 21-day-old offspring of diabetic rats.
Collapse
Affiliation(s)
- Sabrina L Roberti
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Romina Higa
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Hugo Sato
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Dalmiro Gomez Ribot
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Evangelina Capobianco
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina
| | - Alicia Jawerbaum
- Universidad de Buenos Aires, Facultad de Medicina, Argentina; CONICET-Universidad de Buenos Aires, Laboratory of Reproduction and Metabolism, CEFYBO, Buenos Aires, Argentina.
| |
Collapse
|
39
|
Fu S, Li Y, Wu Y, Yue Y, Yang D. Icariside II improves myocardial fibrosis in spontaneously hypertensive rats by inhibiting collagen synthesis. ACTA ACUST UNITED AC 2019; 72:227-235. [PMID: 31820448 DOI: 10.1111/jphp.13190] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/21/2019] [Indexed: 01/01/2023]
Abstract
OBJECTIVES We aimed to investigate the effects of icariside II (ICS II) on myocardial fibrosis in spontaneously hypertensive rats (SHRs) and to explore the possible mechanisms. METHODS We used SHRs as animal models, and we administered ICS II (4, 8 or 16 mg/kg) orally by gavage for 12 consecutive weeks (Fu et al., Biomed Pharmacother 2018; 100: 64). The left ventricular morphology of the rats was observed using haematoxylin-eosin (HE) staining. The occurrence of myocardial interstitial fibrosis was detected by Masson's trichrome staining. The protein levels of alpha smooth muscle actin (α-SMA), Collagen I, III, matrix metalloproteinases 2 and 9 (MMP-2 and MMP-9, respectively), tissue inhibitor of metalloproteinase 1 (TIMP-1), transforming growth factor-β1 (TGF-β1), phospho-Smad2 (p-Smad2), phospho-Smad3 (p-Smad3) and phospho-p38 (p-p38) were examined by Western blotting. KEY FINDINGS The results suggested that ICS II improved myocardial interstitial and perivascular collagen deposition and decreased Collagen I/III and α-SMA expression. ICS II (8 and 16 mg/kg) downregulated the expression of MMP-2 and MMP9 and upregulated the expression of TIMP1. In addition, the protein levels of p-Smad2/3, TGF-β1 and p-p38 were decreased by ICS II treatment. CONCLUSIONS The results suggest that ICS II can inhibit the expression of Collagen I and Collagen III through the MMP/TIMP-1 and TGF-β1/Smad2,3/p-p38 signalling pathways and that it has therapeutic effects on myocardial fibrosis.
Collapse
Affiliation(s)
- Shu Fu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yeli Li
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuting Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yun Yue
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| | - Danli Yang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
40
|
Matchett EF, Wang S, Crawford BD. Paralogues of Mmp11 and Timp4 Interact during the Development of the Myotendinous Junction in the Zebrafish Embryo. J Dev Biol 2019; 7:jdb7040022. [PMID: 31816958 PMCID: PMC6955687 DOI: 10.3390/jdb7040022] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/25/2019] [Accepted: 11/27/2019] [Indexed: 02/07/2023] Open
Abstract
The extracellular matrix (ECM) of the myotendinous junction (MTJ) undergoes dramatic physical and biochemical remodeling during the first 48 h of development in zebrafish, transforming from a rectangular fibronectin-dominated somite boundary to a chevron-shaped laminin-dominated MTJ. Matrix metalloproteinase 11 (Mmp11, a.k.a. Stromelysin-3) is both necessary and sufficient for the removal of fibronectin at the MTJ, but whether this protease acts directly on fibronectin and how its activity is regulated remain unknown. Using immunofluorescence, we show that both paralogues of Mmp11 accumulate at the MTJ during this time period, but with Mmp11a present early and later replaced by Mmp11b. Moreover, Mmp11a also accumulates intracellularly, associated with the Z-discs of sarcomeres within skeletal muscle cells. Using the epitope-mediated MMP activation (EMMA) assay, we show that despite having a weaker paired basic amino acid motif in its propeptide than Mmp11b, Mmp11a is activated by furin, but may also be activated by other mechanisms intracellularly. One or both paralogues of tissue inhibitors of metalloproteinase-4 (Timp4) are also present at the MTJ throughout this process, and yeast two-hybrid assays reveal distinct and specific interactions between various domains of these proteins. We propose a model in which Mmp11a activity is modulated (but not inhibited) by Timp4 during early MTJ remodeling, followed by a phase in which Mmp11b activity is both inhibited and spatially constrained by Timp4 in order to maintain the structural integrity of the mature MTJ.
Collapse
|
41
|
Intracellular Localization in Zebrafish Muscle and Conserved Sequence Features Suggest Roles for Gelatinase A Moonlighting in Sarcomere Maintenance. Biomedicines 2019; 7:biomedicines7040093. [PMID: 31795436 PMCID: PMC6966518 DOI: 10.3390/biomedicines7040093] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 12/24/2022] Open
Abstract
Gelatinase A (Mmp2 in zebrafish) is a well-characterized effector of extracellular matrix remodeling, extracellular signaling, and along with other matrix metalloproteinases (MMPs) and extracellular proteases, it plays important roles in the establishment and maintenance of tissue architecture. Gelatinase A is also found moonlighting inside mammalian striated muscle cells, where it has been implicated in the pathology of ischemia-reperfusion injury. Gelatinase A has no known physiological function in muscle cells, and its localization within mammalian cells appears to be due to inefficient recognition of its N-terminal secretory signal. Here we show that Mmp2 is abundant within the skeletal muscle cells of zebrafish, where it localizes to the M-line of sarcomeres and degrades muscle myosin. The N-terminal secretory signal of zebrafish Mmp2 is also challenging to identify, and this is a conserved characteristic of gelatinase A orthologues, suggesting a selective pressure acting to prevent the efficient secretion of this protease. Furthermore, there are several strongly conserved phosphorylation sites within the catalytic domain of gelatinase A orthologues, some of which are phosphorylated in vivo, and which are known to regulate the activity of this protease. We conclude that gelatinase A likely participates in uncharacterized physiological functions within the striated muscle, possibly in the maintenance of sarcomere proteostasis, that are likely regulated by kinases and phosphatases present in the sarcomere.
Collapse
|
42
|
Landry N, Kavosh MS, Filomeno KL, Rattan SG, Czubryt MP, Dixon IMC. Ski drives an acute increase in MMP-9 gene expression and release in primary cardiac myofibroblasts. Physiol Rep 2019; 6:e13897. [PMID: 30488595 PMCID: PMC6429976 DOI: 10.14814/phy2.13897] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/24/2018] [Accepted: 09/26/2018] [Indexed: 12/12/2022] Open
Abstract
Many etiologies of heart disease are characterized by expansion and remodeling of the cardiac extracellular matrix (ECM or matrix) which results in cardiac fibrosis. Cardiac fibrosis is mediated in cardiac fibroblasts by TGF‐β1/R‐Smad2/3 signaling. Matrix component proteins are synthesized by activated resident cardiac fibroblasts known as myofibroblasts (MFB). These events are causal to heart failure with diastolic dysfunction and reduced cardiac filling. We have shown that exogenous Ski, a TGF‐β1/Smad repressor, localizes in the cellular nucleus and deactivates cardiac myofibroblasts. This deactivation is associated with reduction of myofibroblast marker protein expression in vitro, including alpha smooth muscle actin (α‐SMA) and extracellular domain‐A (ED‐A) fibronectin. We hypothesize that Ski also acutely regulates MMP expression in cardiac MFB. While acute Ski overexpression in cardiac MFB in vitro was not associated with any change in intracellular MMP‐9 protein expression versus LacZ‐treated controls,exogenous Ski caused elevated MMP‐9 mRNA expression and increased MMP‐9 protein secretion versus controls. Zymographic analysis revealed increased MMP‐9‐specific gelatinase activity in myofibroblasts overexpressing Ski versus controls. Moreover, Ski expression was attended by reduced paxillin and focal adhesion kinase phosphorylation (FAK ‐ Tyr 397) versus controls. As myofibroblasts are hypersecretory and less motile relative to fibroblasts, Ski's reduction of paxillin and FAK expression may reflect the relative deactivation of myofibroblasts. Thus, in addition to its known antifibrotic effects, Ski overexpression elevates expression and extracellular secretion/release of MMP‐9 and thus may facilitate internal cytoskeletal remodeling as well as extracellular ECM components. Further, as acute TGF‐β1 treatment of primary cardiac MFB is known to cause rapid translocation of Ski to the nucleus, our data support an autoregulatory role for Ski in mediating cardiac ECM accumulation.
Collapse
Affiliation(s)
- Natalie Landry
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Morvarid S Kavosh
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Krista L Filomeno
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sunil G Rattan
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Michael P Czubryt
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Ian M C Dixon
- Department of Physiology and Pathophysiology, Institute of Cardiovascular Sciences, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
43
|
Ren X, Lamb GD, Murphy RM. Distribution and activation of matrix metalloproteinase-2 in skeletal muscle fibers. Am J Physiol Cell Physiol 2019; 317:C613-C625. [PMID: 31241984 DOI: 10.1152/ajpcell.00113.2019] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
A substantial intracellular localization of matrix metalloproteinase 2 (MMP2) has been reported in cardiomyocytes, where it plays a role in the degradation of the contractile apparatus following ischemia-reperfusion injury. Whether MMP2 may have a similar function in skeletal muscle is unknown. This study determined that the absolute amount of MMP2 is similar in rat skeletal and cardiac muscle and human muscle (~10-18 nmol/kg muscle wet wt) but is ~50- to 100-fold less than the amount of calpain-1. We compared mechanically skinned muscle fibers, where the extracellular matrix (ECM) is completely removed, with intact fiber segments and found that ~30% of total MMP2 was associated with the ECM, whereas ~70% was inside the muscle fibers. Concordant with whole muscle fractionation, further separation of skinned fiber segments into cytosolic, membranous, and cytoskeletal and nuclear compartments indicated that ~57% of the intracellular MMP2 was freely diffusible, ~6% was associated with the membrane, and ~37% was bound within the fiber. Under native zymography conditions, only 10% of MMP2 became active upon prolonged (17 h) exposure to 20 μM Ca2+, a concentration that would fully activate calpain-1 in seconds to minutes; full activation of MMP2 would require ~1 mM Ca2+. Given the prevalence of intracellular MMP2 in skeletal muscle, it is necessary to investigate its function using physiological conditions, including isolation of any potential functional relevance of MMP2 from that of the abundant protease calpain-1.
Collapse
Affiliation(s)
- Xiaoyu Ren
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| | - Graham D Lamb
- School of Life Sciences, La Trobe University, Melbourne, Victoria, Australia
| | - Robyn M Murphy
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, Victoria, Australia
| |
Collapse
|
44
|
Maldonado M, Salgado-Aguayo A, Herrera I, Cabrera S, Ortíz-Quintero B, Staab-Weijnitz CA, Eickelberg O, Ramírez R, Manicone AM, Selman M, Pardo A. Upregulation and Nuclear Location of MMP28 in Alveolar Epithelium of Idiopathic Pulmonary Fibrosis. Am J Respir Cell Mol Biol 2019; 59:77-86. [PMID: 29373068 DOI: 10.1165/rcmb.2017-0223oc] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a chronic and progressive aging-associated disease of unknown etiology. A growing body of evidence indicates that aberrant activated alveolar epithelial cells induce the expansion and activation of the fibroblast population, leading to the destruction of the lung architecture. Some matrix metalloproteinases (MMPs) are upregulated in IPF, indicating that they may be important in the pathogenesis and/or progression of IPF. In the present study, we examined the expression of MMP28 in this disease and evaluated its functional effects in two alveolar epithelial cell lines and in human primary bronchial epithelial cells. We found that the enzyme is expressed in bronchial (apical and cytoplasmic localization) and alveolar (cytoplasmic and nuclear localization) epithelial cells in two different groups of patients with IPF. In vitro MMP28 epithelial silencing decreased the proliferation rate and delayed wound closing, whereas overexpression showed opposite effects, protecting from apoptosis and enhanced epithelial-mesenchymal transition. Our findings demonstrate that MMP28 is upregulated in epithelial cells from IPF lungs, where it may play a role in increasing the proliferative and migratory phenotype in a catalysis-dependent manner.
Collapse
Affiliation(s)
- Mariel Maldonado
- 1 Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Alfonso Salgado-Aguayo
- 2 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - Iliana Herrera
- 2 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - Sandra Cabrera
- 1 Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Blanca Ortíz-Quintero
- 2 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - Claudia A Staab-Weijnitz
- 3 Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany
| | - Oliver Eickelberg
- 3 Comprehensive Pneumology Center, Helmholtz Zentrum München, Member of the German Center of Lung Research (DZL), Munich, Germany.,4 Division of Pulmonary Sciences and Critical Care Medicine, School of Medicine, University of Colorado-Anschutz Medical Campus, Aurora, Colorado; and
| | - Remedios Ramírez
- 1 Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Anne M Manicone
- 5 Center for Lung Biology, Division of Pulmonary and Critical Care Medicine, Department of Medicine, University of Washington, Seattle, Washington
| | - Moisés Selman
- 2 Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Ciudad de México, México
| | - Annie Pardo
- 1 Facultad de Ciencias, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
45
|
Nagel F, Santer D, Stojkovic S, Kaun C, Schaefer AK, Krššák M, Abraham D, Bencsik P, Ferdinandy P, Kenyeres E, Szabados T, Wojta J, Trescher K, Kiss A, Podesser BK. The impact of age on cardiac function and extracellular matrix component expression in adverse post-infarction remodeling in mice. Exp Gerontol 2019; 119:193-202. [PMID: 30763602 DOI: 10.1016/j.exger.2019.02.008] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 02/06/2019] [Accepted: 02/08/2019] [Indexed: 01/18/2023]
Abstract
The aim of this study was to describe the potential associations of the expression of matricellular components in adverse post-infarction remodeling of the geriatric heart. In male geriatric (OM, age: 18 months) and young (YM, age: 11 weeks) OF1 mice myocardial infarction (MI) was induced by permanent ligation of the left anterior descending coronary artery. Cardiac function was evaluated by MRI. Plasma and myocardial tissue samples were collected 3d, 7d, and 32d post-MI. Age and MI were associated with impaired cardiac function accompanied by left-ventricular (LV) dilatation. mRNA expression of MMP-2 (7d: p < 0.05), TIMP-1 (7d: p < 0.05), TIMP-2 (7d: p < 0.05), Collagen-1 (3d and 7d: p < 0.05) and Collagen-3 (7d: p < 0.05) in LV non-infarcted myocardium was significantly higher in YM than in OM after MI. MMP-9 activity in plasma was increased in OM after MI (3d: p < 0.01). Tenascin-C protein levels assessed by ELISA were decreased in OM as compared to YM after MI in plasma (3d: p < 0.001, 7d: p < 0.05) and LV non-infarcted myocardium (7d: p < 0.01). Dysregulation in ECM components in non-infarcted LV might be associated and contribute to adverse LV remodeling and impaired cardiac function. Thus, targeting ECM might be a potential therapeutic approach to enhance cardiac function in geriatric patients following MI.
Collapse
Affiliation(s)
- Felix Nagel
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria; Department of Cardiac Surgery, University Hospital St. Poelten, Dunant-Platz 1, 3100 St. Poelten, Austria
| | - David Santer
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria; Department of Cardiovascular Surgery, Hospital Hietzing, Wolkersbergenstr. 1, 1130 Wien, Austria
| | - Stefan Stojkovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Wien, Austria
| | - Christoph Kaun
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Wien, Austria
| | - Anne-Kristin Schaefer
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria
| | - Martin Krššák
- Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Wien, Austria; High Field MR Centre, Department of Biomedical Imaging and Image Guided Therapy, Medical University of Vienna, Lazarettg. 14, 1090 Wien, Austria
| | - Dietmar Abraham
- Laboratory for Molecular Cellular Biology, Medical University of Vienna, Schwarzspanierstr. 17, 1090 Wien, Austria
| | - Péter Bencsik
- Pharmahungary Group, Szeged, Hungary; Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dom ter 12, 6721 Szeged, Hungary
| | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, Budapest 1089, Hungary
| | - Eva Kenyeres
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dom ter 12, 6721 Szeged, Hungary
| | - Tamara Szabados
- Cardiovascular Research Group, Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Dom ter 12, 6721 Szeged, Hungary
| | - Johann Wojta
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria; Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Wien, Austria
| | - Karola Trescher
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria; Department of Cardiac Surgery, University Hospital St. Poelten, Dunant-Platz 1, 3100 St. Poelten, Austria
| | - Attila Kiss
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria
| | - Bruno K Podesser
- Ludwig Boltzmann Cluster for Cardiovascular Research at the Center for Biomedical Research, Medical University of Vienna, Waehringer Guertel 18-20, Leitstelle 1Q, 1090 Wien, Austria; Department of Cardiac Surgery, University Hospital St. Poelten, Dunant-Platz 1, 3100 St. Poelten, Austria.
| |
Collapse
|
46
|
Saxena S, Gupta A, Shukla V, Rani V. Functional annotation of differentially expressed fetal cardiac microRNA targets: implication for microRNA-based cardiovascular therapeutics. 3 Biotech 2018; 8:494. [PMID: 30498667 DOI: 10.1007/s13205-018-1520-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 11/17/2018] [Indexed: 01/23/2023] Open
Abstract
Gene expression pattern of a failing heart depicts remarkable similarity with developing fetal heart. Elucidating genetic as well as epigenetic mechanisms regulating the gene expression during cardiac development will improve our understanding of cardiovascular diseases. In the present study, we aimed to validate and characterize differentially expressed known microRNAs (miRNA) obtained from next generation sequencing data of two fetal cardiac developmental stages (days 4th and 14th) from chicken (G. gallus domesticus) using bioinformatic approaches. Potential mRNA targets of individual miRNA were identified and classified according to their biological, cellular, and molecular functions. Functional annotation of putative target genes was performed to predict their association with cardiovascular diseases. We identified a total of 19 differentially expressed miRNAs between 4th and 14th day sample from the data sets obtained by next generation sequencing. A total of nearly 1522 potential targets ranging from 15 to 270 for each miRNA were predicted out of which 1221 were unique, while 301 were overlapping. Gene ontology and KEGG analysis revealed that majority of these target genes regulate critical cellular and molecular processes including transcriptional regulation, protein transport, signal transduction, matrix remodeling, Ras signaling, MAPK signaling, and TGF-beta signaling pathways indicating the complex nature of microRNA-mediated gene regulation during cardiogenesis. We found a significant association between potential target genes and cardiovascular diseases validating a link between fetal cardiac miRNAs and regulation of cardiovascular disease-related genes. These important findings may lay a foundation for further understanding the regulatory mechanisms operative in gene re-programming in the failing heart.
Collapse
|
47
|
Chan BYH, Roczkowsky A, Moser N, Poirier M, Hughes BG, Ilarraza R, Schulz R. Doxorubicin induces de novo expression of N-terminal-truncated matrix metalloproteinase-2 in cardiac myocytes. Can J Physiol Pharmacol 2018; 96:1238-1245. [PMID: 30308129 DOI: 10.1139/cjpp-2018-0275] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Anthracyclines, such as doxorubicin, are commonly prescribed antineoplastic agents that cause irreversible cardiac injury. Doxorubicin cardiotoxicity is initiated by increased oxidative stress in cardiomyocytes. Oxidative stress enhances intracellular matrix metalloproteinase-2 (MMP-2) by direct activation of its full-length isoform and (or) de novo expression of an N-terminal-truncated isoform (NTT-MMP-2). As MMP-2 is localized to the sarcomere, we tested whether doxorubicin activates intracellular MMP-2 in neonatal rat ventricular myocytes (NRVM) and whether it thereby proteolyzes two of its identified sarcomeric targets, α-actinin and troponin I. Doxorubicin increased oxidative stress within 12 h as indicated by reduced aconitase activity. This was associated with a twofold increase in MMP-2 protein levels and threefold higher gelatinolytic activity. MMP inhibitors ARP-100 or ONO-4817 (1 μM) prevented doxorubicin-induced MMP-2 activation. Doxorubicin also increased the levels and activity of MMP-2 secreted into the conditioned media. Doxorubicin upregulated the mRNA expression of both full-length MMP-2 and NTT-MMP-2. α-Actinin levels remained unchanged, whereas doxorubicin downregulated troponin I in an MMP-independent manner. Doxorubicin induces oxidative stress and stimulates a robust increase in MMP-2 expression and activity in NRVM, including NTT-MMP-2. The sarcomeric proteins α-actinin and troponin I are, however, not targeted by MMP-2 under these conditions.
Collapse
Affiliation(s)
- Brandon Y H Chan
- Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada.,Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Andrej Roczkowsky
- Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada.,Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Nils Moser
- Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada.,Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Mathieu Poirier
- Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada.,Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Bryan G Hughes
- Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada.,Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Ramses Ilarraza
- Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada.,Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada
| | - Richard Schulz
- Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada.,Departments of Pediatrics and Pharmacology, 462 HMRC, University of Alberta, Edmonton, AB T6G 2S2, Canada
| |
Collapse
|
48
|
Kiugel M, Kytö V, Saanijoki T, Liljenbäck H, Metsälä O, Ståhle M, Tuomela J, Li XG, Saukko P, Knuuti J, Roivainen A, Saraste A. Evaluation of 68Ga-labeled peptide tracer for detection of gelatinase expression after myocardial infarction in rat. J Nucl Cardiol 2018; 25:1114-1123. [PMID: 27914007 DOI: 10.1007/s12350-016-0744-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 11/11/2016] [Indexed: 12/26/2022]
Abstract
BACKGROUND Matrix metalloproteinases 2 and 9 (MMP-2/9) play a role in extracellular matrix remodeling after an ischemic myocardial injury. We evaluated 68Ga-DOTA-peptide targeting MMP-2/9 for the detection of gelatinase expression after myocardial infarction (MI) in rat. METHODS Rats were injected with 43 ± 7.7 MBq of 68Ga-DOTA-peptide targeting MMP-2/9 at 7 days (n = 7) or 4 weeks (n = 8) after permanent coronary ligation or sham operation (n = 5 at both time points) followed by positron emission tomography (PET). The left ventricle was cut in frozen sections for autoradiography and immunohistochemistry 30 minutes after tracer injection. RESULTS Immunohistochemical staining showed MMP-2 and MMP-9 expressing cells, CD31-positive endothelial cells, and CD68-positive macrophages in the infarcted myocardium. Autoradiography showed increased tracer uptake in the infarcted area both at 7 days and 4 weeks after MI (MI-to-remote area ratio 2.5 ± 0.46 and 3.1 ± 1.0, respectively). Tracer uptake in damaged tissue correlated with the amount of CD68-positive macrophages at 7 days after MI, and CD31-positive endothelial cells at 7 days and 4 weeks after MI. The tracer was rapidly metabolized, radioactivity in the blood exceeded that of the myocardium, and tracer accumulation in the heart was not detectable by in vivo PET. CONCLUSIONS 68Ga-DOTA-peptide targeting MMP-2/9 accumulates in the damaged rat myocardium after an ischemic injury, but tracer instability and slow clearance in vivo make it unsuitable for further evaluation.
Collapse
Affiliation(s)
- Max Kiugel
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | - Ville Kytö
- Heart Center, Turku University Hospital, Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular Medicine, University of Turku, Turku, Finland
| | - Tiina Saanijoki
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | - Heidi Liljenbäck
- Turku PET Centre, University of Turku, 20521, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
| | - Olli Metsälä
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | - Mia Ståhle
- Turku PET Centre, University of Turku, 20521, Turku, Finland
| | - Johanna Tuomela
- Department of Cell Biology and Anatomy, University of Turku, Turku, Finland
| | - Xiang-Guo Li
- Turku PET Centre, University of Turku, 20521, Turku, Finland
- Turku PET Centre, Åbo Akademi University, Turku, Finland
| | - Pekka Saukko
- Department of Pathology and Forensic Medicine, University of Turku, Turku, Finland
| | - Juhani Knuuti
- Turku PET Centre, University of Turku, 20521, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Anne Roivainen
- Turku PET Centre, University of Turku, 20521, Turku, Finland
- Turku Center for Disease Modeling, University of Turku, Turku, Finland
- Turku PET Centre, Turku University Hospital, Turku, Finland
| | - Antti Saraste
- Turku PET Centre, University of Turku, 20521, Turku, Finland.
- Heart Center, Turku University Hospital, Turku, Finland.
- Institute of Clinical Medicine, University of Turku, Turku, Finland.
| |
Collapse
|
49
|
White Wine Consumption Influences Inflammatory Phase of Repair After Myocardial Infarction in Rats. J Cardiovasc Pharmacol 2018; 70:293-299. [PMID: 28731891 DOI: 10.1097/fjc.0000000000000519] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Effects of white wine (WW) consumption on the expression of inflammatory markers/mediators (MMP-2, MMP-9, NF-ĸB p65 and TGF-β1) in myocardial tissue after experimentally induced permanent myocardial ischemia was investigated. Male Sprague-Dawley rats were given either a combination of WW and water or only water, for 28 days. After coronary ligation, animals were left to survive for 24 hours. Three representative areas: infarct/ischemic, peri-infarct/border zone, and control/non-ischemic zones were analyzed for expression of immunoreactivity by measuring the threshold area % of signal density. For MMP-9, significantly smaller expression was found in all 3 zones of wine drinking animals (P < 0.001). There was no difference in MMP-2 immunoreactivity between the 2 groups, except in peri-infarct zones, where the signal was significantly decreased (P < 0.001). The same pattern of expression was found for the NF-κB p65 signal, although no differences between experimental groups were observed for TGF-β1. White wine consumption decreases the expression of the 3 investigated inflammatory markers/mediators in the peri-infarct zone, suggesting its significant modulatory effect. For MMP-9 and MMP-2, expression was similar to the effect of postischemic reperfusion. No effect on TGF-β1 was observed, highlighting its role in being the master-switch, changing from the inflammatory to the proliferative stage of infarct healing.
Collapse
|
50
|
Bencsik P, Kupai K, Görbe A, Kenyeres É, Varga ZV, Pálóczi J, Gáspár R, Kovács L, Weber L, Takács F, Hajdú I, Fabó G, Cseh S, Barna L, Csont T, Csonka C, Dormán G, Ferdinandy P. Development of Matrix Metalloproteinase-2 Inhibitors for Cardioprotection. Front Pharmacol 2018; 9:296. [PMID: 29674965 PMCID: PMC5896266 DOI: 10.3389/fphar.2018.00296] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 03/14/2018] [Indexed: 12/14/2022] Open
Abstract
The objective of our present study is to develop novel inhibitors for MMP-2 for acute cardioprotection. In a series of pilot studies, novel substituted carboxylic acid derivatives were synthesized based on imidazole and thiazole scaffolds and then tested in a screeening cascade for MMP inhibition. We found that the MMP-inhibiting effects of imidazole and thiazole carboxylic acid-based compounds are superior in efficacy in comparison to the conventional hydroxamic acid derivatives of the same molecules. Based on these results, a 568-membered focused library of imidazole and thiazole compounds was generated in silico and then the library members were docked to the 3D model of MMP-2 followed by an in vitro medium throughput screening (MTS) based on a fluorescent assay employing MMP-2 catalytic domain. Altogether 45 compounds showed a docking score of >70, from which 30 compounds were successfully synthesized. Based on the MMP-2 inhibitory tests using gelatin zymography, 7 compounds were then selected and tested in neonatal rat cardiac myocytes subjected to simulated I/R injury. Six compounds showed significant cardio-cytoprotecion and the most effective compound (MMPI-1154) significantly decreased infarct size when applied at 1 μM in an ex vivo model for acute myocardial infarction. This is the first demonstration that imidazole and thiazole carboxylic acid-based compounds are more efficacious MMP-2 inhibitor than their hydroxamic acid derivatives. MMPI-1154 is a promising novel cardio-cytoprotective imidazole-carboxylic acid MMP-2 inhibitor lead candidate for the treatment of acute myocardial infarction.
Collapse
Affiliation(s)
- Péter Bencsik
- Cardiovascular Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Krisztina Kupai
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Éva Kenyeres
- Cardiovascular Research Group, Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - János Pálóczi
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Renáta Gáspár
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | | | | | | | - István Hajdú
- Targetex Biosciences, Dunakeszi, Hungary.,Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary
| | | | | | - László Barna
- Research Centre for Natural Sciences, Institute of Enzymology, Hungarian Academy of Sciences, Budapest, Hungary.,Microscopy Center at IEM HAS, Institute of Experimental Medicine, Hungarian Academy of Sciences, Budapest, Hungary
| | - Tamás Csont
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Csaba Csonka
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | | | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| |
Collapse
|