1
|
Wang H, Wang H, Zheng Q, Wang J, Si J. Unusual norcucurbitacin glycosides from the roots of Siraitia grosvenorii. PHYTOCHEMISTRY 2024; 227:114230. [PMID: 39102929 DOI: 10.1016/j.phytochem.2024.114230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 07/22/2024] [Accepted: 07/25/2024] [Indexed: 08/07/2024]
Abstract
Siraitia grosvenorii Swingle is one of the first approved medicine food homology species in China, and it has been used as a natural sweetener in the food industry and as a traditional medicine to relieve cough and reduce phlegm. However, many S. grosvenorii roots are discarded yearly, which results in a great waste of resources. Twelve undescribed norcucurbitacin-type triterpenoid glycosides, siraitiaosides A-L (1-12), and six known analogs (13-18) were isolated from the roots of S. grosvenorii. The structures of isolated norcucurbitacin glycosides were elucidated by comprehensive data analyses, including HRESIMS, UV, IR, NMR, ECD calculations, and X-ray crystallography analysis. Siraitiaosides A-E (1-5) featured an unusual 19,29-norcucurbitacin framework while siraitiaosides F-L (6-12) featured a rare 29-norcucurbitacin framework. Notably, compound 4 displayed moderate anti-acetylcholinesterase (AChE) activity with an IC50 of 21.0 μM, meanwhile, compounds 16 and 18 exhibited pronounced cytotoxic activities against MCF-7, CNE-1, and HeLa cancer cell lines with IC50 values of 2.1-15.2 μM. In silico studies showed that compound 4 bound closely to AChE with a binding energy of -5.04 kcal/mol, and compound 18 could tightly bind to PI3K, AKT1, ERK2, and MMP9 proteins that related to autophagy, apoptosis, migration/invasion, and growth/proliferation. In summary, the roots of Siraitia grosvenorii have potential medicinal values due to the multiple bioactive components.
Collapse
Affiliation(s)
- Huijuan Wang
- The Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Huaxiang Wang
- The Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Qi Zheng
- The Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China
| | - Junchi Wang
- The Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| | - Jianyong Si
- The Key Laboratory of Bioactive Substances and Resources Utilization of Chinese Herbal Medicine, Ministry of Education, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100193, China.
| |
Collapse
|
2
|
Bassiouni W, Valencia R, Mahmud Z, Seubert JM, Schulz R. Matrix metalloproteinase-2 proteolyzes mitofusin-2 and impairs mitochondrial function during myocardial ischemia-reperfusion injury. Basic Res Cardiol 2023; 118:29. [PMID: 37495895 DOI: 10.1007/s00395-023-00999-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 07/12/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
During myocardial ischemia and reperfusion (IR) injury matrix metalloproteinase-2 (MMP-2) is rapidly activated in response to oxidative stress. MMP-2 is a multifunctional protease that cleaves both extracellular and intracellular proteins. Oxidative stress also impairs mitochondrial function which is regulated by different proteins, including mitofusin-2 (Mfn-2), which is lost in IR injury. Oxidative stress and mitochondrial dysfunction trigger the NLRP3 inflammasome and the innate immune response which invokes the de novo expression of an N-terminal truncated isoform of MMP-2 (NTT-MMP-2) at or near mitochondria. We hypothesized that MMP-2 proteolyzes Mfn-2 during myocardial IR injury, impairing mitochondrial function and enhancing the inflammasome response. Isolated hearts from mice subjected to IR injury (30 min ischemia/40 min reperfusion) showed a significant reduction in left ventricular developed pressure (LVDP) compared to aerobically perfused hearts. IR injury increased MMP-2 activity as observed by gelatin zymography and increased degradation of troponin I, an intracellular MMP-2 target. MMP-2 preferring inhibitors, ARP-100 or ONO-4817, improved post-ischemic recovery of LVDP compared to vehicle perfused IR hearts. In muscle fibers isolated from IR hearts the rates of mitochondrial oxygen consumption and ATP production were impaired compared to those from aerobic hearts, whereas ARP-100 or ONO-4817 attenuated these reductions. IR hearts showed higher levels of NLRP3, cleaved caspase-1 and interleukin-1β in the cytosolic fraction, while the mitochondria-enriched fraction showed reduced levels of Mfn-2, compared to aerobic hearts. ARP-100 or ONO-4817 attenuated these changes. Co-immunoprecipitation showed that MMP-2 is associated with Mfn-2 in aerobic and IR hearts. ARP-100 or ONO-4817 also reduced infarct size and cell death in hearts subjected to 45 min ischemia/120 min reperfusion. Following myocardial IR injury, impaired contractile function and mitochondrial respiration and elevated inflammasome response could be attributed, at least in part, to MMP-2 activation, which targets and cleaves mitochondrial Mfn-2. Inhibition of MMP-2 activity protects against cardiac contractile dysfunction in IR injury in part by preserving Mfn-2 and suppressing inflammation.
Collapse
Affiliation(s)
- Wesam Bassiouni
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Robert Valencia
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Zabed Mahmud
- Department of Pediatrics, Faculty of Medicine and Dentistry, 4-62 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada
| | - John M Seubert
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Richard Schulz
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
- Department of Pediatrics, Faculty of Medicine and Dentistry, 4-62 Heritage Medical Research Centre, University of Alberta, Edmonton, AB, T6G 2S2, Canada.
| |
Collapse
|
3
|
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
|
4
|
Maslov LN, Popov SV, Mukhomedzyanov AV, Naryzhnaya NV, Voronkov NS, Ryabov VV, Boshchenko AA, Khaliulin I, Prasad NR, Fu F, Pei JM, Logvinov SV, Oeltgen PR. Reperfusion Cardiac Injury: Receptors and the Signaling Mechanisms. Curr Cardiol Rev 2022; 18:63-79. [PMID: 35422224 PMCID: PMC9896422 DOI: 10.2174/1573403x18666220413121730] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 01/01/2022] [Accepted: 01/10/2022] [Indexed: 11/22/2022] Open
Abstract
It has been documented that Ca2+ overload and increased production of reactive oxygen species play a significant role in reperfusion injury (RI) of cardiomyocytes. Ischemia/reperfusion induces cell death as a result of necrosis, necroptosis, apoptosis, and possibly autophagy, pyroptosis and ferroptosis. It has also been demonstrated that the NLRP3 inflammasome is involved in RI of the heart. An increase in adrenergic system activity during the restoration of coronary perfusion negatively affected cardiac resistance to RI. Toll-like receptors are involved in RI of the heart. Angiotensin II and endothelin-1 aggravated ischemic/reperfusion injury of the heart. Activation of neutrophils, monocytes, CD4+ T-cells and platelets contributes to cardiac ischemia/reperfusion injury. Our review outlines the role of these factors in reperfusion cardiac injury.
Collapse
Affiliation(s)
- Leonid N. Maslov
- Address correspondence to this author at the Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Kyevskskaya 111A, 634012 Tomsk, Russia; Tel. +7 3822 262174; E-mail:
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Li J, Thangaiyan R, Govindasamy K, Wei J. Anti-inflammatory and anti-apoptotic effect of zingiberene on isoproterenol-induced myocardial infarction in experimental animals. Hum Exp Toxicol 2020; 40:915-927. [PMID: 33242989 DOI: 10.1177/0960327120975131] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
This study aimed to investigate the antihyperlipidemic and anti-inflammatory effect of zingiberene (ZBN) on isoproterenol-(ISO) induced myocardial infarction in rats. ZBN (10 mg/kg b.wt.) was orally administered to rats for 21 days and ISO (85 mg/kg b.wt.) was subcutaneously injected into the rats at 24 h intervals for the last 2 consecutive days. We observed increased serum creatine kinase, creatine kinase-MB, cardiac troponin T, and I levels in ISO-treated MI rats. Conversely, ZBN oral administration significantly prevented in cardiac marker enzyme activities in ISO-mediated rats. We also noticed that ZBN oral administration prevented ISO-induced expression of lipid peroxidative markers, total cholesterol, triglycerides, phospholipids, free fatty acids, very-low-density lipoprotein cholesterol (VLDL-C), low-density lipoprotein cholesterol (LDL-C) to the normal basal level. Furthermore, ZBN restored ISO-mediated antioxidant status, increased level of high-density lipoprotein cholesterol (HDL-C), and tissue phospholipids to the near-normal levels. Besides, ZBN pre-treatment significantly reduced the level of inflammatory markers (TNF-α, IL-6, NF-κB, and IL-1β) in ISO-induced MI in rats. We noticed that ZBN pretreatment inhibited the pro-apoptotic proteins Bax and cytochrome c and increased the Bcl-2 expression in ISO induced rats. The gene expression profiling by qRT-PCR array illustrates that ZBN treatment prevents the ISO mediated activation of cardiac markers, inflammatory, and fibrosis-related genes in the heart tissue. Taken together, pre-treatment with ZBN attenuated ISO-induced MI resolved exhibits the anti-inflammatory and antiapoptotic effect.
Collapse
Affiliation(s)
- Jianwei Li
- Department of Cardiology, Xi'an Fourth Hospital, Xi'an, Shaanxi, China
| | - Radhiga Thangaiyan
- Department of Biochemistry and Biotechnology, 29895Annamalai University, Annamalai Nagar, Tamil Nadu, India
| | - Kanimozhi Govindasamy
- Department of Biochemistry, Dharmapuram Gnanambigai Government Arts College for Women, Mayiladuthurai, Tamil Nadu, India
| | - Jianxia Wei
- Department of Cardiology, The Third Affiliated Hospital of Xi 'an Medical College, Xi'an, Shaanxi, China
| |
Collapse
|
6
|
Pečan P, Hambalkó S, Ha VT, Nagy CT, Pelyhe C, Lainšček D, Kenyeres B, Brenner GB, Görbe A, Kittel Á, Barteková M, Ferdinandy P, Manček-Keber M, Giricz Z. Calcium Ionophore-Induced Extracellular Vesicles Mediate Cytoprotection against Simulated Ischemia/Reperfusion Injury in Cardiomyocyte-Derived Cell Lines by Inducing Heme Oxygenase 1. Int J Mol Sci 2020; 21:ijms21207687. [PMID: 33081396 PMCID: PMC7589052 DOI: 10.3390/ijms21207687] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 10/06/2020] [Accepted: 10/14/2020] [Indexed: 12/25/2022] Open
Abstract
Cardioprotection against ischemia/reperfusion injury is still an unmet clinical need. The transient activation of Toll-like receptors (TLRs) has been implicated in cardioprotection, which may be achieved by treatment with blood-derived extracellular vesicles (EVs). However, since the isolation of EVs from blood takes considerable effort, the aim of our study was to establish a cellular model from which cardioprotective EVs can be isolated in a well-reproducible manner. EV release was induced in HEK293 cells with calcium ionophore A23187. EVs were characterized and cytoprotection was assessed in H9c2 and AC16 cell lines. Cardioprotection afforded by EVs and its mechanism were investigated after 16 h simulated ischemia and 2 h reperfusion. The induction of HEK293 cells by calcium ionophore resulted in the release of heterogenous populations of EVs. In H9c2 and AC16 cells, stressEVs induced the downstream signaling of TLR4 and heme oxygenase 1 (HO-1) expression in H9c2 cells. StressEVs decreased necrosis due to simulated ischemia/reperfusion injury in H9c2 and AC16 cells, which was independent of TLR4 induction, but not that of HO-1. Calcium ionophore-induced EVs exert cytoprotection by inducing HO-1 in a TLR4-independent manner.
Collapse
Affiliation(s)
- Peter Pečan
- National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (P.P.); (V.T.H.); (D.L.)
- Graduate School of Biomedicine, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Szabolcs Hambalkó
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (S.H.); (C.T.N.); (C.P.); (B.K.); (G.B.B.); (A.G.); (P.F.)
| | - Van Thai Ha
- National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (P.P.); (V.T.H.); (D.L.)
- Graduate School of Biomedicine, Faculty of Medicine, University of Ljubljana, SI-1000 Ljubljana, Slovenia
| | - Csilla T. Nagy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (S.H.); (C.T.N.); (C.P.); (B.K.); (G.B.B.); (A.G.); (P.F.)
| | - Csilla Pelyhe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (S.H.); (C.T.N.); (C.P.); (B.K.); (G.B.B.); (A.G.); (P.F.)
| | - Duško Lainšček
- National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (P.P.); (V.T.H.); (D.L.)
- Centre of Excelence EN-FIST, SI-1000 Ljubljana, Slovenia
| | - Bence Kenyeres
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (S.H.); (C.T.N.); (C.P.); (B.K.); (G.B.B.); (A.G.); (P.F.)
| | - Gábor B. Brenner
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (S.H.); (C.T.N.); (C.P.); (B.K.); (G.B.B.); (A.G.); (P.F.)
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (S.H.); (C.T.N.); (C.P.); (B.K.); (G.B.B.); (A.G.); (P.F.)
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Ágnes Kittel
- Institute of Experimental Medicine, ELRN, 1083 Budapest, Hungary;
| | - Monika Barteková
- Centre of Experimental Medicine, Institute for Heart Research, Slovak Academy of Sciences, 84104 Bratislava, Slovakia;
- Institute of Physiology, Faculty of Medicine, Comenius University in Bratislava, 81372 Bratislava, Slovakia
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (S.H.); (C.T.N.); (C.P.); (B.K.); (G.B.B.); (A.G.); (P.F.)
- Pharmahungary Group, 6722 Szeged, Hungary
| | - Mateja Manček-Keber
- National Institute of Chemistry, SI-1000 Ljubljana, Slovenia; (P.P.); (V.T.H.); (D.L.)
- Centre of Excelence EN-FIST, SI-1000 Ljubljana, Slovenia
- Correspondence: (M.M.-K.); (Z.G.); Tel.: +386-1-476-0393 (M.M.-K.); +36-1-210-4416 (Z.G.)
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, 1085 Budapest, Hungary; (S.H.); (C.T.N.); (C.P.); (B.K.); (G.B.B.); (A.G.); (P.F.)
- Pharmahungary Group, 6722 Szeged, Hungary
- Correspondence: (M.M.-K.); (Z.G.); Tel.: +386-1-476-0393 (M.M.-K.); +36-1-210-4416 (Z.G.)
| |
Collapse
|
7
|
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
|
8
|
Hidden Cardiotoxicity of Rofecoxib Can be Revealed in Experimental Models of Ischemia/Reperfusion. Cells 2020; 9:cells9030551. [PMID: 32111102 PMCID: PMC7140447 DOI: 10.3390/cells9030551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/20/2020] [Accepted: 02/24/2020] [Indexed: 12/21/2022] Open
Abstract
Cardiac adverse effects are among the leading causes of the discontinuation of clinical trials and the withdrawal of drugs from the market. The novel concept of ‘hidden cardiotoxicity’ is defined as cardiotoxicity of a drug that manifests in the diseased (e.g., ischemic/reperfused), but not in the healthy heart or as a drug-induced deterioration of cardiac stress adaptation (e.g., ischemic conditioning). Here, we aimed to test if the cardiotoxicity of a selective COX-2 inhibitor rofecoxib that was revealed during its clinical use, i.e., increased occurrence of proarrhythmic and thrombotic events, could have been revealed in early phases of drug development by using preclinical models of ischemia/reperfusion (I/R) injury. Rats that were treated with rofecoxib or vehicle for four weeks were subjected to 30 min. coronary artery occlusion and 120 min. reperfusion with or without cardioprotection that is induced by ischemic preconditioning (IPC). Rofecoxib increased overall the arrhythmias including ventricular fibrillation (VF) during I/R. The proarrhythmic effect of rofecoxib during I/R was not observed in the IPC group. Rofecoxib prolonged the action potential duration (APD) in isolated papillary muscles, which was not seen in the simulated IPC group. Interestingly, while showing hidden cardiotoxicity manifested as a proarrhythmic effect during I/R, rofecoxib decreased the infarct size and increased the survival of adult rat cardiac myocytes that were subjected to simulated I/R injury. This is the first demonstration that rofecoxib increased acute mortality due to its proarrhythmic effect via increased APD during I/R. Rofecoxib did not interfere with the cardiprotective effect of IPC; moreover, IPC was able to protect against rofecoxib-induced hidden cardiotoxicity. These results show that cardiac safety testing with simple preclinical models of I/R injury uncovers hidden cardiotoxicity of rofecoxib and might reveal the hidden cardiotoxicity of other drugs.
Collapse
|
9
|
Makkos A, Szántai Á, Pálóczi J, Pipis J, Kiss B, Poggi P, Ferdinandy P, Chatgilialoglu A, Görbe A. A Comorbidity Model of Myocardial Ischemia/Reperfusion Injury and Hypercholesterolemia in Rat Cardiac Myocyte Cultures. Front Physiol 2020; 10:1564. [PMID: 31992989 PMCID: PMC6962358 DOI: 10.3389/fphys.2019.01564] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Accepted: 12/12/2019] [Indexed: 12/19/2022] Open
Abstract
Introduction The use of comorbidity models is crucial in cardioprotective drug development. Hypercholesterolemia causes endothelial and myocardial dysfunction, as well as aggravates ischemia/reperfusion (I/R)-induced myocardial injury. Endogenous cardioprotective mechanisms against I/R are impaired in hyperlipidemic and hyperglycemic in vivo animal models. Therefore, our aim was to develop a medium throughput comorbidity cell-based test system of myocardial I/R injury, hypercholesterolemia and hyperglycemia that mimics comorbidity conditions. Methods Cardiac myocytes isolated from neonatal or adult rat hearts were cultured in control or in three different hypercholesterolemic media with increasing cholesterol content (hiChol) or hiChol + hyperglycemic medium, respectively. Each group was then subjected to simulated ischemia/reperfusion (SI/R) or corresponding normoxic condition, respectively. Cholesterol uptake was tested by Filipin staining in neonatal cardiac myocytes. Cell viability, total cell count and oxidative stress, i.e., total reactive oxygen species (ROS) and superoxide level were measured by fluorescent assays. Results Neonatal cardiac myocytes took up cholesterol from the different hiChol media at a concentration-dependent manner. In normoxia, viability of hiChol neonatal cardiac myocytes was not significantly changed, however, superoxide levels were increased as compared to vehicle. After SI/R, the viability of hiChol neonatal cardiac myocytes was decreased and total ROS level was increased as compared to vehicle. HiChol combined with hyperglycemia further aggravated cell death and oxidative stress in normoxic as well as in SI/R conditions. Viability of hiChol adult cardiac myocytes was significantly decreased and superoxide level was increased in normoxia and these changes were further aggravated by SI/R. HiChol combined with hyperglycemia further aggravated cell death, however level of oxidative stress increased only in normoxic condition. Conclusion HiChol rat cardiac myocytes showed reduction of cell viability and increased oxidative stress, which were further aggravated by SI/R and with additional hyperglycemia. This is the first demonstration that the combination of the current hypercholesterolemic medium and SI/R in cardiac myocytes mimics the cardiac pathology of the comorbid heart with I/R and hypercholesterolemia.
Collapse
Affiliation(s)
- András Makkos
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Ágnes Szántai
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - János Pálóczi
- Department of Biochemistry, University of Szeged, Szeged, Hungary
| | | | - Bernadett Kiss
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Department of Biochemistry, University of Szeged, Szeged, Hungary
| | | | - Péter Ferdinandy
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Department of Biochemistry, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| | | | - Anikó Görbe
- Cardiometabolic Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Department of Biochemistry, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| |
Collapse
|
10
|
Kim JS, Lee KB, Park JH, Sung SM, Oh K, Kim EG, Chang DI, Hwang YH, Lee EJ, Kim WK, Ju C, Kim BS, Ryu JM. Safety and Efficacy of Otaplimastat in Patients with Acute Ischemic Stroke Requiring tPA (SAFE-TPA): A Multicenter, Randomized, Double-Blind, Placebo-Controlled Phase 2 Study. Ann Neurol 2019; 87:233-245. [PMID: 31721277 PMCID: PMC7003891 DOI: 10.1002/ana.25644] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 10/22/2019] [Accepted: 11/10/2019] [Indexed: 12/30/2022]
Abstract
Objective Otaplimastat is a neuroprotectant that inhibits matrix metalloprotease pathway, and reduces edema and intracerebral hemorrhage induced by recombinant tissue plasminogen activator (rtPA) in animal stroke models. We aimed to assess the safety and efficacy of otaplimastat in patients receiving rtPA. Methods This was a phase 2, 2‐part, multicenter trial in stroke patients (19–80 years old) receiving rtPA. Intravenous otaplimastat was administered <30 minutes after rtPA. Stage 1 was a single‐arm, open‐label safety study in 11 patients. Otaplimastat 80 mg was administered twice daily for 3 days. Stage 2 was a randomized, double‐blind, placebo‐controlled study involving 69 patients, assigned (1:1:1) to otaplimastat 40 mg, otaplimastat 80 mg, or a placebo. The primary endpoint was the occurrence of parenchymal hematoma (PH) on day 1. Secondary endpoints included serious adverse events (SAEs), mortality, and modified Rankin scale (mRS) distribution at 90 days (http://clinicaltrials.gov identifier: NCT02787278). Results No safety issues were encountered in stage 1. The incidence of PH during stage 2 was comparable: 0 of 22 with the placebo, 0 of 22 with otaplimastat 40 mg, and 1 of 21 with the 80 mg dose. No differences in SAEs (13%, 17%, 14%) or death (8.3%, 4.2%, 4.8%) were observed among the 3 groups. Three adverse events (chills, muscle rigidity, hepatotoxicity) were judged to be related to otaplimastat. Interpretation Intravenous otaplimastat adjunctive therapy in patients receiving rtPA is feasible and generally safe. The functional efficacy of otaplimastat needs to be investigated with further large trials. ANN NEUROL 2020;87:233–245
Collapse
Affiliation(s)
- Jong S Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul
| | - Kyung Bok Lee
- Department of Neurology, Soonchunhyang University School of Medicine, Seoul
| | - Jong-Ho Park
- Department of Neurology, Myongji Hospital, Hanyang University College of Medicine, Goyang
| | - Sang Min Sung
- Department of Neurology, Pusan National University Hospital, Busan
| | - Kyungmi Oh
- Department of Neurology, Korea University Guro Hospital, Seoul
| | - Eung-Gyu Kim
- Department of Neurology, Inje University Busan Paik Hospital, Busan
| | - Dae-Il Chang
- Department of Neurology, Kyung Hee University Hospital, Seoul
| | - Yang Ha Hwang
- Department of Neurology, Kyungpook National University School of Medicine and Hospital, Daegu
| | - Eun-Jae Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul
| | - Won-Ki Kim
- Department of Neuroscience, Korea University College of Medicine, Seoul
| | - Chung Ju
- Research Headquarters, Shin Poong Pharmaceutical, Ansan, Korea
| | - Byung Su Kim
- Research Headquarters, Shin Poong Pharmaceutical, Ansan, Korea
| | - Jei-Man Ryu
- Research Headquarters, Shin Poong Pharmaceutical, Ansan, Korea
| | | |
Collapse
|
11
|
Scrimgeour NR, Wrobel A, Pinho MJ, Høydal MA. microRNA-451a prevents activation of matrix metalloproteinases 2 and 9 in human cardiomyocytes during pathological stress stimulation. Am J Physiol Cell Physiol 2019; 318:C94-C102. [PMID: 31618079 DOI: 10.1152/ajpcell.00204.2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Matrix metalloproteinases (MMP) are important for cardiac remodeling. Recently, microRNA (miR)-451a has been found to inhibit the expression of both MMP-2 and MMP-9 in human malignancies, but its role in cardiomyocytes has not been explored. We hypothesized that miR-451a modulates MMP-2 and MMP-9 levels in human cardiomyocytes. The role of miR-451a on regulation of MMP-2 and MMP-9 was evaluated in two separate pathological models using Cor.4U human inducible pluripotent stem cell-derived cardiomyocytes (hiPS-CMs): 1) endothelin-1 (ET-1), and 2) 48-h hypoxia (1% O2). Both models were transfected with synthetic miR-451a mimics or scramble control. Expression of both mRNA and miR was determined by quantitative real-time polymerase chain reaction and protein activity by (MMP-2/9) activity assay. Bioinformatic analyses were performed using Targetscan 7.1 and STRING 10.5. hiPS-CMs stimulated by hypoxia increased both MMP-2 and MMP-9 expression levels compared with normoxia (P < 0.05), whereas ET-1 stimulation only increased the MMP-9 level compared with vehicle controls (P < 0.05). miR-451a mimics prevented the increase of MMP-2 and MMP-9 expression in both models. Protein activity of MMP-2 and MMP-9 was confirmed to be lower following treatment with miR-451a mimic compared with scramble-controls. Six of 28 predicted gene transcripts of miR-451a were linked to MMP-2 and MMP-9; Macrophage migration inhibitory factor (MIF) was the only predicted target of miR-451a that was increased by ET-1 and hypoxia and reduced following miR-451a mimic transfection. miR-451a prevent the increase of MMP-2 and MMP-9 in human cardiomyocytes during pathological stress. The modulation by miR-451a on MMP-2 and MMP-9 is caused by MIF.
Collapse
Affiliation(s)
- Nathan Robert Scrimgeour
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway.,St. Olav's University Hospital, Trondheim, Norway
| | - Aleksandra Wrobel
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway.,St. Olav's University Hospital, Trondheim, Norway
| | - Maria João Pinho
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway.,St. Olav's University Hospital, Trondheim, Norway
| | - Morten Andre Høydal
- Group of Molecular and Cellular Cardiology, Department of Circulation and Medical Imaging, Faculty of Medicine and Health, Norwegian University of Science and Technology, Trondheim, Norway.,St. Olav's University Hospital, Trondheim, Norway
| |
Collapse
|
12
|
Tokuhara CK, Santesso MR, Oliveira GSND, Ventura TMDS, Doyama JT, Zambuzzi WF, Oliveira RCD. Updating the role of matrix metalloproteinases in mineralized tissue and related diseases. J Appl Oral Sci 2019; 27:e20180596. [PMID: 31508793 DOI: 10.1590/1678-7757-2018-0596] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 05/30/2019] [Indexed: 02/22/2023] Open
Abstract
Bone development and healing processes involve a complex cascade of biological events requiring well-orchestrated synergism with bone cells, growth factors, and other trophic signaling molecules and cellular structures. Beyond health processes, MMPs play several key roles in the installation of heart and blood vessel related diseases and cancer, ranging from accelerating metastatic cells to ectopic vascular mineralization by smooth muscle cells in complementary manner. The tissue inhibitors of MMPs (TIMPs) have an important role in controlling proteolysis. Paired with the post-transcriptional efficiency of specific miRNAs, they modulate MMP performance. If druggable, these molecules are suggested to be a platform for development of "smart" medications and further clinical trials. Thus, considering the pleiotropic effect of MMPs on mammals, the purpose of this review is to update the role of those multifaceted proteases in mineralized tissues in health, such as bone, and pathophysiological disorders, such as ectopic vascular calcification and cancer.
Collapse
Affiliation(s)
- Cintia Kazuko Tokuhara
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Bauru, São Paulo, Brasil
| | - Mariana Rodrigues Santesso
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Bauru, São Paulo, Brasil
| | - Gabriela Silva Neubern de Oliveira
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Bauru, São Paulo, Brasil
| | - Talita Mendes da Silva Ventura
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Bauru, São Paulo, Brasil
| | - Julio Toshimi Doyama
- Universidade Estadual Paulista Júlio de Mesquita Filho, Campus Botucatu, Rubião Jr, São Paulo, Brasil
| | - Willian Fernando Zambuzzi
- Universidade Estadual Paulista Júlio de Mesquita Filho, Campus Botucatu, Rubião Jr, São Paulo, Brasil
| | - Rodrigo Cardoso de Oliveira
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Laboratório de Bioquímica, Bauru, São Paulo, Brasil
| |
Collapse
|
13
|
Horváth Á, Botz B, Kiss T, Csekő K, Kiss I, Felinger A, Szabados T, Kenyeres É, Bencsik P, Mócsai A, Ferdinandy P, Helyes Z. Subantimicrobial Dose Doxycycline Worsens Chronic Arthritis-Induced Bone Microarchitectural Alterations in a Mouse Model: Role of Matrix Metalloproteinases? Front Pharmacol 2019; 10:233. [PMID: 30949048 PMCID: PMC6435543 DOI: 10.3389/fphar.2019.00233] [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: 07/01/2018] [Accepted: 02/22/2019] [Indexed: 11/23/2022] Open
Abstract
Background: Rheumatoid arthritis (RA) is a chronic inflammatory joint disease hallmarked by irreversible damage of cartilage and bone. Matrix metalloproteinases (MMPs) involved in connective tissue remodeling play an important role in this process. Numerous MMPs have been examined in humans and animals, but their functions are still not fully understood. Therefore, we investigated the role of MMPs in the K/BxN serum-transfer model of RA with the broad-spectrum MMP inhibitor subantimicrobial dose doxycycline (SDD) using complex in vivo and in vitro methodolgy. Methods: Chronic arthritis was induced by repetitive i.p. injections of K/BxN serum in C57BL/6J mice. SDD was administered daily in acidified drinking water (0.5 mg/mL, 80 mg/kg) during the 30 days experimental period. Mechanonociceptive threshold of the paw was evaluated by aesthesiometry, grasping ability by grid test, arthritis severity by scoring, neutrophil myeloperoxidase activity by luminescence, vascular hyperpermeability and MMP activity by fluorescence in vivo imaging and the latter also by gelatin zymography, bone structure by micro-computed tomography (micro-CT). Plasma concentrations of doxycycline were determined by liquid chromatography-mass spectrometry analysis. Results: K/BxN serum induced significant inflammatory signs, mechanical hyperalgesia, joint function impairment, increased myeloperoxidase activity and vascular hyperpermeability. Significant increase of MMP activity was also observed both in vivo and ex vivo with elevation of the 57–60, 75, and 92 kDa gelatinolytic isoforms in the arthritic ankle joints, but neither MMP activity nor any above described functional parameters were influenced by SDD. Most importantly, SDD significantly reduced bone mineral density in the distal tibia and enhanced the Euler number in the ankle. Arthritis-induced microarchitectural alterations demonstrating increased irregularity and cancellous bone remodeling, such as increased Euler number was significantly elevated by SDD in both regions. Conclusion: We showed increase of various MMP activities in the joints by in vivo fluorescence imaging together with ex vivo zymography, and investigated their functional significance using the broad-spectrum MMP inhibitor SDD in the translational RA model. This is the first demonstration that SDD worsens arthritis-induced bone microarchitectural alterations, but it appears to be independent of MMP inhibition.
Collapse
Affiliation(s)
- Ádám Horváth
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,Molecular Pharmacology Research Group, Centre for Neuroscience, János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Bálint Botz
- Molecular Pharmacology Research Group, Centre for Neuroscience, János Szentágothai Research Centre, University of Pécs, Pécs, Hungary.,Department of Radiology, Clinical Centre, University of Pécs, Pécs, Hungary
| | - Tamás Kiss
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,Molecular Pharmacology Research Group, Centre for Neuroscience, János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Kata Csekő
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,Molecular Pharmacology Research Group, Centre for Neuroscience, János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Ibolya Kiss
- Department of Analytical and Environmental Chemistry, Faculty of Sciences, Institute of Chemistry, University of Pécs, Pécs, Hungary.,Environmental Analytical and Geoanalytical Research Group, János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Attila Felinger
- Department of Analytical and Environmental Chemistry, Faculty of Sciences, Institute of Chemistry, University of Pécs, Pécs, Hungary.,Environmental Analytical and Geoanalytical Research Group, János Szentágothai Research Centre, University of Pécs, Pécs, Hungary
| | - Tamara Szabados
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Éva Kenyeres
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Péter Bencsik
- Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Attila Mócsai
- Department of Physiology, Faculty of Medicine, MTA-SE "Lendület" Inflammation Physiology Research Group of the Hungarian Academy of Sciences, Semmelweis University, Budapest, Hungary
| | - Péter Ferdinandy
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary.,Pharmahungary Group, Szeged, Hungary.,Department of Pharmacology and Pharmacotherapy, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Zsuzsanna Helyes
- Department of Pharmacology and Pharmacotherapy, Medical School, University of Pécs, Pécs, Hungary.,Molecular Pharmacology Research Group, Centre for Neuroscience, János Szentágothai Research Centre, University of Pécs, Pécs, Hungary.,Chronic Pain Research Group, National Brain Research Program, Medical School, University of Pécs, Pécs, Hungary.,PharmInVivo Ltd., Pécs, Hungary
| |
Collapse
|
14
|
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
|
15
|
Hausenloy DJ, Garcia-Dorado D, Bøtker HE, Davidson SM, Downey J, Engel FB, Jennings R, Lecour S, Leor J, Madonna R, Ovize M, Perrino C, Prunier F, Schulz R, Sluijter JPG, Van Laake LW, Vinten-Johansen J, Yellon DM, Ytrehus K, Heusch G, Ferdinandy P. Novel targets and future strategies for acute cardioprotection: Position Paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart. Cardiovasc Res 2018; 113:564-585. [PMID: 28453734 DOI: 10.1093/cvr/cvx049] [Citation(s) in RCA: 254] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 03/15/2017] [Indexed: 02/06/2023] Open
Abstract
Ischaemic heart disease and the heart failure that often results, remain the leading causes of death and disability in Europe and worldwide. As such, in order to prevent heart failure and improve clinical outcomes in patients presenting with an acute ST-segment elevation myocardial infarction and patients undergoing coronary artery bypass graft surgery, novel therapies are required to protect the heart against the detrimental effects of acute ischaemia/reperfusion injury (IRI). During the last three decades, a wide variety of ischaemic conditioning strategies and pharmacological treatments have been tested in the clinic-however, their translation from experimental to clinical studies for improving patient outcomes has been both challenging and disappointing. Therefore, in this Position Paper of the European Society of Cardiology Working Group on Cellular Biology of the Heart, we critically analyse the current state of ischaemic conditioning in both the experimental and clinical settings, provide recommendations for improving its translation into the clinical setting, and highlight novel therapeutic targets and new treatment strategies for reducing acute myocardial IRI.
Collapse
Affiliation(s)
- Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK; The National Institute of Health Research University College London Hospitals Biomedical Research Centre, 149 Tottenham Court Road London, W1T 7DN, UK; Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, 8 College Road, Singapore 169857; National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Dr, Singapore 169609, Singapore; Yong Loo Lin School of Medicine, National University Singapore, Singapore; Barts Heart Centre, St Bartholomew's Hospital, London, UK
| | - David Garcia-Dorado
- Department of Cardiology, Vall d Hebron University Hospital and Research Institute. Universitat Autònoma, Passeig de la Vall d'Hebron, 119-129, 08035 Barcelona, Spain
| | - Hans Erik Bøtker
- Department of Cardiology, Aarhus University Hospital Skejby, Palle Juul-Jensens Boulevard 99, 8200 Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - James Downey
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, 5851 USA Dr. N., MSB 3074, Mobile, AL 36688, USA
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nßrnberg, Schloßplatz 4, 91054 Erlangen, Germany
| | - Robert Jennings
- Department of Cardiology, Duke University, Durham, NC 27708, USA
| | - Sandrine Lecour
- Department of Medicine, Hatter Institute for Cardiovascular Research in Africa and South African Medical Research Council Inter-University Cape Heart Group, Faculty of Health Sciences, University of Cape Town, Chris Barnard Building, Anzio Road, Observatory, 7925, Cape Town, Western Cape, South Africa
| | - Jonathan Leor
- Tamman Cardiovascular Research Institute, Sheba Medical Center, Tel Hashomer, Israel; Neufeld Cardiac Research Institute, Tel-Aviv University, Sheba Medical Center, Tel Hashomer, 5265601, Israel; Sheba Center for Regenerative Medicine, Stem Cell, and Tissue Engineering, Tel Hashomer, 5265601, Israel
| | - Rosalinda Madonna
- Center of Aging Sciences and Translational Medicine - CESI-MeT, "G. d'Annunzio" University, Chieti, Italy; Institute of Cardiology, Department of Neurosciences, Imaging, and Clinical Sciences, "G. d'Annunzio University, Chieti, Italy; Texas Heart Institute and University of Texas Medical School in Houston, Department of Internal Medicine, 6770 Bertner Avenue, Houston, Texas 77030 USA
| | - Michel Ovize
- Explorations Fonctionnelles Cardiovasculaires, Hôpital Louis Pradel, 28 Avenue du Doyen Jean Lépine, 69500 Bron, France; UMR 1060 (CarMeN), Université Claude Bernard Lyon, 43 Boulevard du 11 Novembre 1918, 69100 Villeurbanne, France
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Division of Cardiology, Federico II University Corso Umberto I, 40, 80138 Napoli, Italy
| | - Fabrice Prunier
- Department of Cardiology, University of Angers, University Hospital of Angers, 4 Rue Larrey, 49100 Angers, France
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig, University of Giessen, Ludwigstraße 23, 35390 Gießen, Germany
| | - Joost P G Sluijter
- Cardiology and UMC Utrecht Regenerative Medicine Center, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| | - Linda W Van Laake
- Division Heart and Lungs, University Medical Center Utrecht, Heidelberglaan 100, 3584 CX Utrecht, Netherlands
| | - Jakob Vinten-Johansen
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University, 201 Dowman Dr, Atlanta, GA 30322, USA
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK; The National Institute of Health Research University College London Hospitals Biomedical Research Centre, 149 Tottenham Court Road London, W1T 7DN, UK
| | - Kirsti Ytrehus
- Cardiovascular Research Group, Department of Medical Biology, UiT The Arctic University of Norway, Hansine Hansens veg 18, 9019 Tromsø, Norway
| | - Gerd Heusch
- Institute for Pathophysiology, West-German Heart and Vascular Center, University Hospital Essen, Hufelandstrasse 55, 45147 Essen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Nagyvárad tér 4, 1089 Hungary; Pharmahungary Group, Graphisoft Park, 7 Záhony street, Budapest, H-1031, Hungary
| |
Collapse
|
16
|
Serum MMP-9 Diagnostics, Prognostics, and Activation in Acute Coronary Syndrome and Its Recurrence. J Cardiovasc Transl Res 2018; 11:210-220. [PMID: 29349668 PMCID: PMC5974001 DOI: 10.1007/s12265-018-9789-x] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 01/08/2018] [Indexed: 12/21/2022]
Abstract
Matrix metalloproteinase (MMP)-9 is crucial in atherosclerotic plaque rupture and tissue remodeling after a cardiac event. The balance between MMP-9 and endogenous inhibitor, tissue inhibitors of matrix metalloproteinase 1 (TIMP-1), is important in acute coronary syndrome (ACS). This is an age- and gender-matched case-control study of ACS (N = 669). Patients (45.7%) were resampled after recovery, and all were followed up for 6 years. The molecular forms of MMP-9 were investigated by gelatin zymography. Diagnostically, MMP-9 and the MMP-9/TIMP-1 molar ratio were associated with ACS (OR 5.81, 95% CI 2.65–12.76, and 4.96, 2.37–10.38). The MMP-9 concentrations decreased 49% during recovery (p < 0.001). The largest decrease of these biomarkers between acute and recovery phase (ΔMMP-9) protected the patients from major adverse cardiac events, especially the non-fatal events. The fatal events were associated with in vitro activatable MMP-9 levels (p = 0.028). Serum MMP-9 and the MMP-9/TIMP-1 molar ratio may be valuable in ACS diagnosis and prognosis. High serum MMP-9 activation potential is associated with poor cardiovascular outcome.
Collapse
|
17
|
Schreckenberg R, Bencsik P, Weber M, Abdallah Y, Csonka C, Gömöri K, Kiss K, Pálóczi J, Pipis J, Sárközy M, Ferdinandy P, Schulz R, Schlüter KD. Adverse Effects on β-Adrenergic Receptor Coupling: Ischemic Postconditioning Failed to Preserve Long-Term Cardiac Function. J Am Heart Assoc 2017; 6:e006809. [PMID: 29273639 PMCID: PMC5779008 DOI: 10.1161/jaha.117.006809] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Accepted: 11/02/2017] [Indexed: 02/07/2023]
Abstract
BACKGROUND Ischemic preconditioning (IPC) and ischemic postconditioning (IPoC) are currently among the most efficient strategies protecting the heart against ischemia/reperfusion injury. However, the effect of IPC and IPoC on functional recovery following ischemia/reperfusion is less clear, particularly with regard to the specific receptor-mediated signaling of the postischemic heart. The current article examines the effect of IPC or IPoC on the regulation and coupling of β-adrenergic receptors and their effects on postischemic left ventricular function. METHODS AND RESULTS The β-adrenergic signal transduction was analyzed in 3-month-old Wistar rats for each of the intervention strategies (Sham, ischemia/reperfusion, IPC, IPoC) immediately and 7 days after myocardial infarction. Directly after the infarction a cardioprotective potential was demonstrated for both IPC and IPoC: the infarct size was reduced, apoptosis and production of reactive oxygen species were lowered, and the myocardial tissue was preserved. Seven days after myocardial ischemia, only IPC hearts showed significant functional improvement. Along with a deterioration in fractional shortening, IPoC hearts no longer responded adequately to β-adrenergic stimulation. The stabilization of β-adrenergic receptor kinase-2 via increased phosphorylation of Mdm2 (an E3-ubiquitin ligase) was responsible for desensitization of β-adrenergic receptors and identified as a characteristic specific to IPoC hearts. CONCLUSIONS Immediately after myocardial infarction, rapid and transient activation of β-adrenergic receptor kinase-2 may be an appropriate means to protect the injured heart from excessive stress. In the long term, however, induction and stabilization of β-adrenergic receptor kinase-2, with the resultant loss of positive inotropic function, leads to the functional picture of heart failure.
Collapse
Affiliation(s)
- Rolf Schreckenberg
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Péter Bencsik
- Pharmahungary Group, Szeged, Hungary
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - Martin Weber
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Yaser Abdallah
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Gießen, Germany
| | - Csaba Csonka
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - Kamilla Gömöri
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - Krisztina Kiss
- Pharmahungary Group, Szeged, Hungary
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - János Pálóczi
- Pharmahungary Group, Szeged, Hungary
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | | | - Márta Sárközy
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Hungary
| | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Rainer Schulz
- Physiologisches Institut, Justus-Liebig-Universität Gießen, Gießen, Germany
| | | |
Collapse
|
18
|
Andreadou I, Iliodromitis EK, Lazou A, Görbe A, Giricz Z, Schulz R, Ferdinandy P. Effect of hypercholesterolaemia on myocardial function, ischaemia-reperfusion injury and cardioprotection by preconditioning, postconditioning and remote conditioning. Br J Pharmacol 2017; 174:1555-1569. [PMID: 28060997 DOI: 10.1111/bph.13704] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 12/16/2016] [Accepted: 12/20/2016] [Indexed: 01/15/2023] Open
Abstract
Hypercholesterolaemia is considered to be a principle risk factor for cardiovascular disease, having direct negative effects on the myocardium itself, in addition to the development of atherosclerosis. Since hypercholesterolaemia affects the global cardiac gene expression profile, among many other factors, it results in increased myocardial oxidative stress, mitochondrial dysfunction and inflammation triggered apoptosis, all of which may account for myocardial dysfunction and increased susceptibility of the myocardium to infarction. In addition, numerous experimental and clinical studies have revealed that hyperlcholesterolaemia may interfere with the cardioprotective potential of conditioning mechanisms. Although not fully elucidated, the underlying mechanisms for the lost cardioprotection in hypercholesterolaemic animals have been reported to involve dysregulation of the endothelial NOS-cGMP, reperfusion injury salvage kinase, peroxynitrite-MMP2 signalling pathways, modulation of ATP-sensitive potassium channels and apoptotic pathways. In this review article, we summarize the current knowledge on the effect of hypercholesterolaemia on the non-ischaemic and ischaemic heart as well as on the cardioprotection induced by drugs or ischaemic preconditioning, postconditioning and remote conditioning. Future perspectives concerning the mechanisms and the design of preclinical and clinical trials are highlighted. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, School of Health Sciences, National and Kapodistrian University of Athens, Athens, Greece
| | - Efstathios K Iliodromitis
- Second Department of Cardiology, Medical School, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Antigone Lazou
- School of Biology, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary.,Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Rainer Schulz
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary.,Department of Physiology, Justus-Liebig-University, Giessen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary.,Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged, Hungary
| |
Collapse
|
19
|
Bencsik P, Bartekova M, Görbe A, Kiss K, Pálóczi J, Radosinska J, Szűcs G, Ferdinandy P. MMP Activity Detection in Zymograms. Methods Mol Biol 2017; 1626:53-70. [PMID: 28608200 DOI: 10.1007/978-1-4939-7111-4_6] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
Matrix metalloproteinases (MMP) belong to a distinguished class of zinc-dependent endopeptidases. Zymography is a semi-quantitative tool for determining the activity of different MMP isoenzymes in a variety of biological samples. In substrate gel zymography, protein samples of different origin (tissue, cell lysates, plasma/serum, perfusates, other liquids) are separated in sodium dodecyl sulfate (SDS) polyacrylamide gels containing copolymerized substrate (gelatin, casein, elastin, etc.), and after incubation-enabling substrate cleavage by MMPs, MMP activities are detected after the gel staining as transparent bands against a dark-blue background. In situ zymography is a histological modification of substrate zymography in frozen sections, allowing detection of the localization of the MMP activities within the tissue. Here, we describe detailed experimental protocols of all abovementioned techniques and provide examples for several sample measurements.
Collapse
Affiliation(s)
- Péter Bencsik
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary.
- Pharmahungary Group, Szeged, Hungary.
| | - Monika Bartekova
- Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic
- Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic
| | - Anikó Görbe
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Krisztina Kiss
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | | | - Jana Radosinska
- Institute for Heart Research, Slovak Academy of Sciences, Bratislava, Slovak Republic
- Institute of Physiology, Faculty of Medicine, Comenius University, Bratislava, Slovak Republic
| | - Gergő Szűcs
- Department of Biochemistry, Faculty of Medicine, University of Szeged, Szeged, Hungary
| | - Péter Ferdinandy
- Pharmahungary Group, Szeged, Hungary
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| |
Collapse
|
20
|
Influence of Ischemic Pre- and Post-Conditioning on Cardiac Expression of Calcium-Sensing Receptor. BIONANOSCIENCE 2016. [DOI: 10.1007/s12668-016-0316-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
21
|
Kiss K, Csonka C, Pálóczi J, Pipis J, Görbe A, Kocsis GF, Murlasits Z, Sárközy M, Szűcs G, Holmes CP, Pan Y, Bhandari A, Csont T, Shamloo M, Woodburn KW, Ferdinandy P, Bencsik P. Novel, selective EPO receptor ligands lacking erythropoietic activity reduce infarct size in acute myocardial infarction in rats. Pharmacol Res 2016; 113:62-70. [PMID: 27521836 DOI: 10.1016/j.phrs.2016.08.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 08/09/2016] [Indexed: 10/21/2022]
Abstract
Erythropoietin (EPO) has been shown to protect the heart against acute myocardial infarction in pre-clinical studies, however, EPO failed to reduce infarct size in clinical trials and showed significant safety problems. Here, we investigated cardioprotective effects of two selective non-erythropoietic EPO receptor ligand dimeric peptides (AF41676 and AF43136) lacking erythropoietic activity, EPO, and the prolonged half-life EPO analogue, darbepoetin in acute myocardial infarction (AMI) in rats. In a pilot study, EPO at 100U/mL significantly decreased cell death compared to vehicle (33.8±2.3% vs. 40.3±1.5%, p<0.05) in rat neonatal cardiomyocytes subjected to simulated ischemia/reperfusion. In further studies (studies 1-4), in vivo AMI was induced by 30min coronary occlusion and 120min reperfusion in male Wistar rats. Test compounds and positive controls for model validation (B-type natriuretic peptide, BNP or cyclosporine A, CsA) were administered iv. before the onset of reperfusion. Infarct size (IS) was measured by standard TTC staining. In study 1, 5000U/kg EPO reduced infarct size significantly compared to vehicle (45.3±4.8% vs. 59.8±4.5%, p<0.05). In study 2, darbepoetin showed a U-shaped dose-response curve with maximal infarct size-reducing effect at 5μg/kg compared to the vehicle (44.4±5.7% vs. 65.9±2.7%, p<0.01). In study 3, AF41676 showed a U-shaped dose-response curve, where 3mg/kg was the most effective dose compared to the vehicle (24.1±3.9% vs. 44.3±2.5%, p<0.001). The positive control BNP significantly decreased infarct size in studies 1-3 by approximately 35%. In study 4, AF43136 at 10mg/kg decreased infarct size, similarly to the positive control CsA compared to the appropriate vehicle (39.4±5.9% vs. 58.1±5.4% and 45.9±2.4% vs. 63.8±4.1%, p<0.05, respectively). This is the first demonstration that selective, non-erythropoietic EPO receptor ligand dimeric peptides AF41676 and AF43136 administered before reperfusion are able to reduce infarct size in a rat model of AMI. Therefore, non-erythropoietic EPO receptor peptide ligands may be promising cardioprotective agents.
Collapse
Affiliation(s)
- Krisztina Kiss
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Dom ter 9, Szeged H-6720, Hungary.
| | - Csaba Csonka
- Department of Biochemistry, University of Szeged, Dom ter 9, Szeged H-6720, Hungary.
| | - János Pálóczi
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Dom ter 9, Szeged H-6720, Hungary; Pharmahungary Group, Dom ter 9, Szeged H-6720, Hungary.
| | - Judit Pipis
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Dom ter 9, Szeged H-6720, Hungary; Pharmahungary Group, Dom ter 9, Szeged H-6720, Hungary.
| | - Anikó Görbe
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Dom ter 9, Szeged H-6720, Hungary; Pharmahungary Group, Dom ter 9, Szeged H-6720, Hungary.
| | - Gabriella F Kocsis
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Dom ter 9, Szeged H-6720, Hungary; Pharmahungary Group, Dom ter 9, Szeged H-6720, Hungary.
| | - Zsolt Murlasits
- Pharmahungary Group, Dom ter 9, Szeged H-6720, Hungary; Sports Science Program, Qatar University, Doha 00974, Qatar.
| | - Márta Sárközy
- Department of Biochemistry, University of Szeged, Dom ter 9, Szeged H-6720, Hungary.
| | - Gergő Szűcs
- Department of Biochemistry, University of Szeged, Dom ter 9, Szeged H-6720, Hungary.
| | | | - Yijun Pan
- Affymax, Inc., 4015 Miranda Ave Fl 1, Palo Alto, CA 94304, United States.
| | - Ashok Bhandari
- Affymax, Inc., 4015 Miranda Ave Fl 1, Palo Alto, CA 94304, United States.
| | - Tamás Csont
- Department of Biochemistry, University of Szeged, Dom ter 9, Szeged H-6720, Hungary.
| | - Mehrdad Shamloo
- Stanford University School of Medicine, Department of Neurosurgery, 1050 Arastradero Road, Building A, Palo Alto, CA 94304, United States.
| | - Kathryn W Woodburn
- Affymax, Inc., 4015 Miranda Ave Fl 1, Palo Alto, CA 94304, United States; Avalanche Biotechnologies, 1035 O'Brien Drive, Menlo Park, CA 94025, United States.
| | - Péter Ferdinandy
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Dom ter 9, Szeged H-6720, Hungary; Pharmahungary Group, Dom ter 9, Szeged H-6720, Hungary; Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvarad ter 4, Budapest H-1089, Hungary.
| | - Péter Bencsik
- Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Dom ter 9, Szeged H-6720, Hungary; Pharmahungary Group, Dom ter 9, Szeged H-6720, Hungary.
| |
Collapse
|
22
|
Baghirova S, Hughes BG, Poirier M, Kondo MY, Schulz R. Nuclear matrix metalloproteinase-2 in the cardiomyocyte and the ischemic-reperfused heart. J Mol Cell Cardiol 2016; 94:153-161. [PMID: 27079252 DOI: 10.1016/j.yjmcc.2016.04.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2016] [Accepted: 04/07/2016] [Indexed: 10/22/2022]
Abstract
Matrix metalloproteinases (MMPs) are zinc-dependent proteases involved in intra- and extra-cellular matrix remodeling resulting from oxidative stress injury to the heart. MMP-2 was the first MMP to be localized to the nucleus; however, its biological functions there are unclear. We hypothesized that MMP-2 is present in the nucleus under normal physiological conditions but increases during myocardial ischemia-reperfusion (I/R) injury-induced oxidative stress, proteolyzing nuclear structural proteins. Lamins are intermediate filament proteins that provide structural support to the nucleus and are putative targets of MMP-2. To identify lamin susceptibility to MMP-2 proteolysis, purified lamin A or B was incubated with MMP-2 in vitro. Lamin A, but not lamin B, was proteolysed by MMP-2 into an approximately 50kDa fragment, which was also predicted by in silico cleavage site analysis. Immunofluorescent confocal microscopy and subcellular fractionation showed MMP-2 both in the cytosol and nuclei of neonatal rat ventricular myocytes. Rat hearts were isolated and perfused by the Langendorff method aerobically, or subjected to I/R injury in the presence or absence of o-phenanthroline, an MMP inhibitor. Nuclear fractions extracted from I/R hearts showed increased MMP-2 activity, but not protein level. The level of troponin I, a known sarcomeric target of MMP-2, was rescued in I/R hearts treated with o-phenanthroline, demonstrating the efficacy of MMP inhibition. However, lamin A or B levels remained unchanged in I/R hearts. MMP-2 has a widespread subcellular distribution in cardiomyocytes, including a significant presence in the nucleus. The increase in nuclear MMP-2 activity seen during stunning injury here, indicates yet unknown biological actions, other than lamin proteolysis, which may require more severe ischemia to effect.
Collapse
Affiliation(s)
- Sabina Baghirova
- Department of Pharmacology, Cardiovascular Research Institute, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| | - Bryan G Hughes
- Department of Pharmacology, Cardiovascular Research Institute, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, Cardiovascular Research Institute, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| | - Mathieu Poirier
- Department of Pharmacology, Cardiovascular Research Institute, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, Cardiovascular Research Institute, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| | - Marcia Y Kondo
- Department of Pharmacology, Cardiovascular Research Institute, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, Cardiovascular Research Institute, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| | - Richard Schulz
- Department of Pharmacology, Cardiovascular Research Institute, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada; Department of Pediatrics, Cardiovascular Research Institute, Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
23
|
Amin M, Pushpakumar S, Muradashvili N, Kundu S, Tyagi SC, Sen U. Regulation and involvement of matrix metalloproteinases in vascular diseases. FRONT BIOSCI-LANDMRK 2016; 21:89-118. [PMID: 26709763 PMCID: PMC5462461 DOI: 10.2741/4378] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc dependent endopeptidases whose main function is to degrade and deposit structural proteins within the extracellular matrix (ECM). A dysregulation of MMPs is linked to vascular diseases. MMPs are classified into collagenases, gelatinases, membrane-type, metalloelastase, stromelysins, matrilysins, enamelysins, and unclassified subgroups. The production of MMPs is stimulated by factors such as oxidative stress, growth factors and inflammation which lead to its up- or down-regulation with subsequent ECM remodeling. Normally, excess activation of MMPs is controlled by their endogenous inhibitors, tissue inhibitors of metalloproteinases (TIMPs). An imbalance of MMPs and TIMPs has been implicated in hypertension, atherosclerotic plaque formation and instability, aortic aneurysms and varicose vein wall remodeling. Also, recent evidence suggests epigenetic regulation of some MMPs in angiogenesis and atherosclerosis. Over the years, pharmacological inhibitors of MMPs have been used to modify or prevent the development of the disease with some success. In this review, we discuss recent advances in MMP biology, and their involvement in the manifestation of vascular disease.
Collapse
Affiliation(s)
- Matthew Amin
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Sathnur Pushpakumar
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Sourav Kundu
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Suresh C Tyagi
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202
| | - Utpal Sen
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY-40202,
| |
Collapse
|
24
|
High Throughput Screening Identifies a Novel Compound Protecting Cardiomyocytes from Doxorubicin-Induced Damage. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:178513. [PMID: 26137186 PMCID: PMC4475553 DOI: 10.1155/2015/178513] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 04/27/2015] [Accepted: 05/20/2015] [Indexed: 12/01/2022]
Abstract
Antracyclines are effective antitumor agents. One of the most commonly used antracyclines is doxorubicin, which can be successfully used to treat a diverse spectrum of tumors. Application of these drugs is limited by their cardiotoxic effect, which is determined by a lifetime cumulative dose. We set out to identify by high throughput screening cardioprotective compounds protecting cardiomyocytes from doxorubicin-induced injury. Ten thousand compounds of ChemBridge's DIVERSet compound library were screened to identify compounds that can protect H9C2 rat cardiomyocytes against doxorubicin-induced cell death. The most effective compound proved protective in doxorubicin-treated primary rat cardiomyocytes and was further characterized to demonstrate that it significantly decreased doxorubicin-induced apoptotic and necrotic cell death and inhibited doxorubicin-induced activation of JNK MAP kinase without having considerable radical scavenging effect or interfering with the antitumor effect of doxorubicin. In fact the compound identified as 3-[2-(4-ethylphenyl)-2-oxoethyl]-1,2-dimethyl-1H-3,1-benzimidazol-3-ium bromide was toxic to all tumor cell lines tested even without doxorubicine treatment. This benzimidazole compound may lead, through further optimalization, to the development of a drug candidate protecting the heart from doxorubicin-induced injury.
Collapse
|
25
|
Ferdinandy P, Hausenloy DJ, Heusch G, Baxter GF, Schulz R. Interaction of risk factors, comorbidities, and comedications with ischemia/reperfusion injury and cardioprotection by preconditioning, postconditioning, and remote conditioning. Pharmacol Rev 2014; 66:1142-74. [PMID: 25261534 DOI: 10.1124/pr.113.008300] [Citation(s) in RCA: 461] [Impact Index Per Article: 41.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Pre-, post-, and remote conditioning of the myocardium are well described adaptive responses that markedly enhance the ability of the heart to withstand a prolonged ischemia/reperfusion insult and provide therapeutic paradigms for cardioprotection. Nevertheless, more than 25 years after the discovery of ischemic preconditioning, we still do not have established cardioprotective drugs on the market. Most experimental studies on cardioprotection are still undertaken in animal models, in which ischemia/reperfusion is imposed in the absence of cardiovascular risk factors. However, ischemic heart disease in humans is a complex disorder caused by, or associated with, cardiovascular risk factors and comorbidities, including hypertension, hyperlipidemia, diabetes, insulin resistance, heart failure, altered coronary circulation, and aging. These risk factors induce fundamental alterations in cellular signaling cascades that affect the development of ischemia/reperfusion injury per se and responses to cardioprotective interventions. Moreover, some of the medications used to treat these risk factors, including statins, nitrates, and antidiabetic drugs, may impact cardioprotection by modifying cellular signaling. The aim of this article is to review the recent evidence that cardiovascular risk factors and their medication may modify the response to cardioprotective interventions. We emphasize the critical need to take into account the presence of cardiovascular risk factors and concomitant medications when designing preclinical studies for the identification and validation of cardioprotective drug targets and clinical studies. This will hopefully maximize the success rate of developing rational approaches to effective cardioprotective therapies for the majority of patients with multiple risk factors.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Derek J Hausenloy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gerd Heusch
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Gary F Baxter
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| | - Rainer Schulz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary (P.F.); Cardiovascular Research Group, Department of Biochemistry, University of Szeged, Szeged and Pharmahungary Group, Szeged, Hungary (P.F.); The Hatter Cardiovascular Institute, University College London, London, United Kingdom (D.J.H.); Institute for Pathophysiology, University of Essen Medical School, Essen, Germany (G.H.); Division of Pharmacology, Cardiff School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Cardiff, United Kingdom (G.F.B.); and Institute of Physiology, Justus-Liebig University, Giessen, Germany (R.S.)
| |
Collapse
|
26
|
Inhibition of MMP-2 expression with siRNA increases baseline cardiomyocyte contractility and protects against simulated ischemic reperfusion injury. BIOMED RESEARCH INTERNATIONAL 2014; 2014:810371. [PMID: 25147815 PMCID: PMC4131446 DOI: 10.1155/2014/810371] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 06/25/2014] [Indexed: 11/17/2022]
Abstract
Matrix metalloproteinases (MMPs) significantly contribute to ischemia reperfusion (I/R) injury, namely, by the degradation of contractile proteins. However, due to the experimental models adopted and lack of isoform specificity of MMP inhibitors, the cellular source and identity of the MMP(s) involved in I/R injury remain to be elucidated. Using isolated adult rat cardiomyocytes, subjected to chemically induced I/R-like injury, we show that specific inhibition of MMP-2 expression and activity using MMP-2 siRNA significantly protected cardiomyocyte contractility from I/R-like injury. This was also associated with increased expression of myosin light chains 1 and 2 (MLC1/2) in comparison to scramble siRNA transfection. Moreover, the positive effect of MMP-2 siRNA transfection on cardiomyocyte contractility and MLC1/2 expression levels was also observed under control conditions, suggesting an important additional role for MMP-2 in physiological sarcomeric protein turnover. This study clearly demonstrates that intracellular expression of MMP-2 plays a significant role in sarcomeric protein turnover, such as MLC1 and MLC2, under aerobic (physiological) conditions. In addition, this study identifies intracellular/autocrine, cardiomyocyte-produced MMP-2, rather than paracrine/extracellular, as responsible for the degradation of MLC1/2 and consequent contractile dysfunction in cardiomyocytes subjected to I/R injury.
Collapse
|
27
|
Hughes BG, Schulz R. Targeting MMP-2 to treat ischemic heart injury. Basic Res Cardiol 2014; 109:424. [PMID: 24986221 DOI: 10.1007/s00395-014-0424-y] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 06/23/2014] [Indexed: 10/24/2022]
Abstract
Matrix metalloproteinase (MMPs) are long understood to be involved in remodeling of the extracellular matrix. However, over the past decade, it has become clear that one of the most ubiquitous MMPs, MMP-2, has numerous intracellular targets in cardiac myocytes. Notably, MMP-2 proteolyzes components of the sarcomere, and its intracellular activity contributes to ischemia-reperfusion injury of the heart. Together with the well documented role played by MMPs in the myocardial remodeling that occurs following myocardial infarction, this has led to great interest in targeting MMPs to treat cardiac ischemic injury. In this review we will describe the expanding understanding of intracellular MMP-2 biology, and how this knowledge may lead to improved treatments for ischemic heart injury. We also critically review the numerous preclinical studies investigating the effects of MMP inhibition in animal models of myocardial infarction and ischemia-reperfusion injury, as well as the recent clinical trials that are part of the effort to translate these results into clinical practice. Acknowledging the disappointing results of past clinical trials of MMP inhibitors for other diseases, we discuss the need for carefully designed preclinical and clinical studies to avoid mistakes that have been previously made. We conclude that inhibition of MMPs, and in particular MMP-2, shows promise as a therapy to prevent the progression from ischemic injury to heart failure. However, it is critical that the full breadth of MMP-2 biology be taken into account as such therapies are developed.
Collapse
Affiliation(s)
- Bryan G Hughes
- Departments of Pediatrics and Pharmacology, Mazankowski Alberta Heart Institute/Cardiovascular Research Centre, University of Alberta, 4-62 HMRC, Edmonton, AB, T6G 2S2, Canada
| | | |
Collapse
|
28
|
Overexpression of the muscle-specific protein, melusin, protects from cardiac ischemia/reperfusion injury. Basic Res Cardiol 2014; 109:418. [PMID: 24859929 DOI: 10.1007/s00395-014-0418-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2013] [Revised: 05/13/2014] [Accepted: 05/16/2014] [Indexed: 02/02/2023]
Abstract
Melusin is a muscle-specific protein which interacts with β1 integrin cytoplasmic domain and acts as chaperone protein. Its overexpression induces improved resistance to cardiac overload delaying left ventricle dilation and reducing the occurrence of heart failure. Here, we investigated possible protective effect of melusin overexpression against acute ischemia/reperfusion (I/R) injury with or without Postconditioning cardioprotective maneuvers. Melusin transgenic (Mel-TG) mice hearts were subjected to 30-min global ischemia followed by 60-min reperfusion. Interestingly, infarct size was reduced in Mel-TG mice hearts compared to wild-type (WT) hearts (40.3 ± 3.5 % Mel-TG vs. 59.5 ± 3.8 % WT hearts; n = 11 animals/group; P < 0.05). The melusin protective effect was also demonstrated by measuring LDH release, which was 50 % lower in Mel-TG compared to WT. Mel-TG hearts had a higher baseline level of AKT, ERK1/2 and GSK3β phosphorylation, and displayed increased phospho-kinases level after I/R compared to WT mice. Post-ischemic Mel-TG hearts displayed also increased levels of the anti-apoptotic factor phospho-BAD. Importantly, pharmacological inhibition of PI3K/AKT (Wortmannin) and ERK1/2 (U0126) pathways abrogated the melusin protective effect. Notably, HSP90, a chaperone known to protect heart from I/R injury, showed high levels of expression in the heart of Mel-TG mice suggesting a possible collaboration of this molecule with AKT/ERK/GSK3β pathways in the melusin-induced protection. Postconditioning, known to activate AKT/ERK/GSK3β pathways, significantly reduced IS and LDH release in WT hearts, but had no additive protective effects in Mel-TG hearts. These findings implicate melusin as an enhancer of AKT and ERK pathways and as a novel player in cardioprotection from I/R injury.
Collapse
|