1
|
Mendoza A, Patel P, Robichaux D, Ramirez D, Karch J. Inhibition of the mPTP and Lipid Peroxidation Is Additively Protective Against I/R Injury. Circ Res 2024; 134:1292-1305. [PMID: 38618716 PMCID: PMC11081482 DOI: 10.1161/circresaha.123.323882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND During myocardial ischemia/reperfusion (I/R) injury, high levels of matrix Ca2+ and reactive oxygen species (ROS) induce the opening of the mitochondrial permeability transition pore (mPTP), which causes mitochondrial dysfunction and ultimately necrotic death. However, the mechanisms of how these triggers individually or cooperatively open the pore have yet to be determined. METHODS Here, we use a combination of isolated mitochondrial assays and in vivo I/R surgery in mice. We challenged isolated liver and heart mitochondria with Ca2+, ROS, and Fe2+ to induce mitochondrial swelling. Using inhibitors of the mPTP (cyclosporine A or ADP) lipid peroxidation (ferrostatin-1, MitoQ), we determined how the triggers elicit mitochondrial damage. Additionally, we used the combination of inhibitors during I/R injury in mice to determine if dual inhibition of these pathways is additivity protective. RESULTS In the absence of Ca2+, we determined that ROS fails to trigger mPTP opening. Instead, high levels of ROS induce mitochondrial dysfunction and rupture independently of the mPTP through lipid peroxidation. As expected, Ca2+ in the absence of ROS induces mPTP-dependent mitochondrial swelling. Subtoxic levels of ROS and Ca2+ synergize to induce mPTP opening. Furthermore, this synergistic form of Ca2+- and ROS-induced mPTP opening persists in the absence of CypD (cyclophilin D), suggesting the existence of a CypD-independent mechanism for ROS sensitization of the mPTP. These ex vivo findings suggest that mitochondrial dysfunction may be achieved by multiple means during I/R injury. We determined that dual inhibition of the mPTP and lipid peroxidation is significantly more protective against I/R injury than individually targeting either pathway alone. CONCLUSIONS In the present study, we have investigated the relationship between Ca2+ and ROS, and how they individually or synergistically induce mitochondrial swelling. Our findings suggest that Ca2+ mediates mitochondrial damage through the opening of the mPTP, although ROS mediates its damaging effects through lipid peroxidation. However, subtoxic levels both Ca2+ and ROS can induce mPTP-mediated mitochondrial damage. Targeting both of these triggers to preserve mitochondria viability unveils a highly effective therapeutic approach for mitigating I/R injury.
Collapse
Affiliation(s)
- Arielys Mendoza
- Department of Integrative Physiology (A.M., P.P., D.R., D.R., J.K.), Baylor College of Medicine, Houston TX
| | - Pooja Patel
- Department of Integrative Physiology (A.M., P.P., D.R., D.R., J.K.), Baylor College of Medicine, Houston TX
| | - Dexter Robichaux
- Department of Integrative Physiology (A.M., P.P., D.R., D.R., J.K.), Baylor College of Medicine, Houston TX
| | - Daniel Ramirez
- Department of Integrative Physiology (A.M., P.P., D.R., D.R., J.K.), Baylor College of Medicine, Houston TX
| | - Jason Karch
- Department of Integrative Physiology (A.M., P.P., D.R., D.R., J.K.), Baylor College of Medicine, Houston TX
- the Cardiovascular Research Institute (J.K.), Baylor College of Medicine, Houston TX
| |
Collapse
|
2
|
Morel O. Potential of Epigenetic Therapy in Alleviating Cardiac Death and Fibrotic Remodeling in Myocardial Infarction. JACC Basic Transl Sci 2024; 9:649-651. [PMID: 38984044 PMCID: PMC11228388 DOI: 10.1016/j.jacbts.2024.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Affiliation(s)
- Olivier Morel
- Department of Cardiology, University Hospital of Strasbourg, Strasbourg, France
- UR3074 Translational Cardiovascular Medicine, University of Strasbourg, Strasbourg, France
- Medical University of Hanoi, Hanoi, Vietnam
| |
Collapse
|
3
|
Pantos CI, Trikas AG, Pissimisis EG, Grigoriou KP, Stougiannos PN, Dimopoulos AK, Linardakis SI, Alexopoulos NA, Evdoridis CG, Gavrielatos GD, Patsourakos NG, Papakonstantinou ND, Theodosis-Georgilas AD, Mourouzis IS. Effects of Acute Triiodothyronine Treatment in Patients with Anterior Myocardial Infarction Undergoing Primary Angioplasty: Evidence from a Pilot Randomized Clinical Trial (ThyRepair Study). Thyroid 2022; 32:714-724. [PMID: 35297659 DOI: 10.1089/thy.2021.0596] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Background: Thyroid hormone has a differential action on healthy and ischemic heart. Triiodothyronine (T3) administration improved postischemic cardiac function while it limited apoptosis in experimentally induced ischemia. Thus, the present study investigated the potential effects of acute liothyronine (LT3) treatment in patients with anterior myocardial infarction. Methods: This study is a pilot, randomized, double-blind, placebo-controlled trial (ThyRepair study). We randomized 52 patients and analyzed data from 37 patients (n = 16 placebo and n = 21 LT3), per prespecified per protocol analysis. We excluded three patients who had died of cardiovascular causes (one in placebo and two in LT3 arm), four with small infarct size below a pre-specified threshold (in the placebo arm), and the rest, who lacked follow-up data. LT3 treatment started after stenting as an intravenous (i.v.) bolus injection of 0.8 μg/kg of LT3 followed by a constant infusion of 0.113 μg/kg/h i.v. for 48 hours. All patients had cardiac magnetic resonance (CMR) at hospital discharge and 6 months follow-up. The primary end point was CMR left ventricular (LV) ejection fraction (LVEF) and secondary endpoints were LV volumes, infarct volume (IV), and safety. Results: The CMR LVEF% at 6 months was 53.6 ± 9.5 for the LT3-treated group and 48.6 ± 11 for placebo, p = 0.15. Acute LT3 treatment resulted in a significantly lower LV end-diastolic volume index (92.2 ± 16.8 mL/m2 vs. 107.5 ± 22.2, p = 0.022) and LV systolic volume index (47.5 ± 13.9 mL/m2 vs. 61.3 ± 21.7, p = 0.024) at hospital discharge, but not at 6 months. There was no statistically significant difference in CMR IV at hospital discharge between the groups (p = 0.24). CMR IV tended to be lower in the LT3-treated group at 6 months (18.7 ± 9.5 vs. 25.9 ± 11.7, in placebo, p = 0.05). Serious, life-threatening events related to LT3 treatment were not observed. A tendency for an increased incidence of atrial fibrillation (AF) was found in the LT3 group during the first 48 hours (19% for T3 group vs. 5% for placebo, p = 0.13). Conclusion: This pilot randomized, placebo-controlled trial study suggests potential favorable effects (acute cardiac dilatation and 6-month IV) as well as potential concerns regarding a higher risk of AF after LT3 administration early after myocardial infarction, which should be tested in a larger scale study.
Collapse
Affiliation(s)
- Constantinos I Pantos
- Department of Pharmacology, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | | | | | | | - Nikolaos A Alexopoulos
- Department of Radiology, IASO Hospital of Athens, Athens, Greece
- Cardiovascular Imaging Unit, Department of Radiology, Athens Euroclinic, Athens, Greece
| | - Costas G Evdoridis
- Department of Cardiology, ELPIS General Hospital of Athens, Athens, Greece
| | | | | | | | | | - Iordanis S Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
4
|
Singhanat K, Apaijai N, Sumneang N, Maneechote C, Arunsak B, Chunchai T, Chattipakorn SC, Chattipakorn N. Therapeutic potential of a single-dose melatonin in the attenuation of cardiac ischemia/reperfusion injury in prediabetic obese rats. Cell Mol Life Sci 2022; 79:300. [PMID: 35588335 PMCID: PMC11072751 DOI: 10.1007/s00018-022-04330-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 01/05/2023]
Abstract
Although acute melatonin treatment effectively reduces cardiac ischemia/reperfusion (I/R) injury in lean rats by modulating melatonin receptor 2 (MT2), there is no information regarding the temporal effects of melatonin administration during cardiac I/R injury in prediabetic obese rats. Prediabetic obese rats induced by chronic consumption of a high-fat diet (HFD) were used. The rats underwent a cardiac I/R surgical procedure (30-min of ischemia, followed by 120-min of reperfusion) and were randomly assigned to receive either vehicle or melatonin treatment. In the melatonin group, rats were divided into 3 different subgroups: (1) pretreatment, (2) treatment during ischemic period, (3) treatment at the reperfusion onset. In the pretreatment subgroup either a nonspecific MT blocker (Luzindole) or specific MT2 blocker (4-PPDOT) was also given to the rats prior to melatonin treatment. Pretreatment with melatonin (10 mg/kg) effectively reduced cardiac I/R injury by reducing infarct size, arrhythmia, and LV dysfunction. Reduction in impaired mitochondrial function, mitochondrial dynamic balance, oxidative stress, defective autophagy, and apoptosis were observed in rats pretreated with melatonin. Unfortunately, the cardioprotective benefits were not observed when 10-mg/kg of melatonin was acutely administered to the rats after cardiac ischemia. Thus, we increased the dose of melatonin to 20 mg/kg, and it was administered to the rats during ischemia or at the onset of reperfusion. The results showed that 20-mg/kg of melatonin effectively reduced cardiac I/R injury to a similar extent to the 10-mg/kg pretreatment regimen. The MT2 blocker inhibited the protective effects of melatonin. Acute melatonin treatment during cardiac I/R injury exerted protective effects in prediabetic obese rats. However, a higher dose of melatonin is required when given after the onset of cardiac ischemia. These effects of melatonin were mainly mediated through activation of MT2.
Collapse
Affiliation(s)
- Kodchanan Singhanat
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nattayaporn Apaijai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Natticha Sumneang
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Busarin Arunsak
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Titikorn Chunchai
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
5
|
Fernandez Rico C, Konate K, Josse E, Nargeot J, Barrère-Lemaire S, Boisguérin P. Therapeutic Peptides to Treat Myocardial Ischemia-Reperfusion Injury. Front Cardiovasc Med 2022; 9:792885. [PMID: 35252383 PMCID: PMC8891520 DOI: 10.3389/fcvm.2022.792885] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 01/07/2022] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVD) including acute myocardial infarction (AMI) rank first in worldwide mortality and according to the World Health Organization (WHO), they will stay at this rank until 2030. Prompt revascularization of the occluded artery to reperfuse the myocardium is the only recommended treatment (by angioplasty or thrombolysis) to decrease infarct size (IS). However, despite beneficial effects on ischemic lesions, reperfusion leads to ischemia-reperfusion (IR) injury related mainly to apoptosis. Improvement of revascularization techniques and patient care has decreased myocardial infarction (MI) mortality however heart failure (HF) morbidity is increasing, contributing to the cost-intense worldwide HF epidemic. Currently, there is no treatment for reperfusion injury despite promising results in animal models. There is now an obvious need to develop new cardioprotective strategies to decrease morbidity/mortality of CVD, which is increasing due to the aging of the population and the rising prevalence rates of diabetes and obesity. In this review, we will summarize the different therapeutic peptides developed or used focused on the treatment of myocardial IR injury (MIRI). Therapeutic peptides will be presented depending on their interacting mechanisms (apoptosis, necroptosis, and inflammation) reported as playing an important role in reperfusion injury following myocardial ischemia. The search and development of therapeutic peptides have become very active, with increasing numbers of candidates entering clinical trials. Their optimization and their potential application in the treatment of patients with AMI will be discussed.
Collapse
Affiliation(s)
- Carlota Fernandez Rico
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Karidia Konate
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Emilie Josse
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| | - Joël Nargeot
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Stéphanie Barrère-Lemaire
- Institut de Génomique Fonctionnelle, Université de Montpellier, CNRS, INSERM, Montpellier, France
- Laboratory of Excellence Ion Channel Science and Therapeutics, Valbonne, France
| | - Prisca Boisguérin
- PHYMEDEXP, Université de Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
6
|
Cardioprotective Natural Compound Pinocembrin Attenuates Acute Ischemic Myocardial Injury via Enhancing Glycolysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4850328. [PMID: 33178386 PMCID: PMC7644300 DOI: 10.1155/2020/4850328] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 09/14/2020] [Accepted: 09/23/2020] [Indexed: 12/11/2022]
Abstract
Purpose Emerging evidence has shown that pinocembrin protects the myocardium from ischemic injury in animals. However, it is unknown whether it has cardioprotection when given at the onset of reperfusion. Also, mechanisms mediating the cardioprotective actions of pinocembrin were largely unknown. Thus, this study is aimed at investigating the effects of pinocembrin postconditioning on ischemia-reperfusion (I/R) injury and the underlying mechanisms. Methods The in vivo mouse model of myocardial I/R injury, ex vivo isolated rat heart with global I/R, and in vitro hypoxia/reoxygenation (H/R) injury model for primary cardiomyocytes were used. Results We found that pinocembrin postconditioning significantly reduced the infarct size and improved cardiac contractile function after acute myocardial I/R. Mechanically, in primary cardiomyocytes, we found that pinocembrin may confer protection in part via direct stimulation of cardiac glycolysis via promoting the expression of the glycolytic enzyme, PFKFB3. Besides, PFKFB3 inhibition abolished pinocembrin-induced glycolysis and protection in cardiomyocytes. More importantly, PFKFB3 knockdown via cardiotropic adeno-associated virus (AAV) abrogated cardioprotective effects of pinocembrin. Moreover, we demonstrated that HIF1α is a key transcription factor driving pinocembrin-induced PFKFB3 expression in cardiomyocytes. Conclusions In conclusion, these results established that the acute cardioprotective benefits of pinocembrin are mediated in part via enhancing PFKFB3-mediated glycolysis via HIF1α, which may provide a new therapeutic target to impede the progression of myocardial I/R injury.
Collapse
|
7
|
Li RW, Deng Y, Pham HN, Weiss S, Chen M, Smith PN. Riluzole protects against skeletal muscle ischaemia-reperfusion injury in a porcine model. Injury 2020; 51:178-184. [PMID: 31882236 DOI: 10.1016/j.injury.2019.12.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/09/2019] [Accepted: 12/16/2019] [Indexed: 02/02/2023]
Abstract
INTRODUCTION Skeletal muscle ischaemia-reperfusion injury (IRI) can be a life threatening condition. It is relevant to various aspects of the management of trauma and surgical patients. Currently there lacks a pharmacological agent that can be used to dampen the effects of IRI. Riluzole has been shown to reduce the effects of IRI on various organ systems, but there have yet to be any studies on the effects in IRI of skeletal muscle. Our aim was to investigate the effects of Riluzole on IRI in the skeletal muscle of pigs. METHODS Twenty-two pigs were randomly divided into groups. Riluzole was administered before ligation of the femoral artery to produce ischaemia in the tibialis anterior muscle in the experimental group but not the control group. The microscopic appearance of muscles were recorded, a TUNEL assay was used to identify DNA damage and glutathione levels were measured. RESULTS In the Riluzole group, muscle fibres appeared less wavy and less oedematous compared to the control group. The Riluzole group also had less evidence of DNA fragmentation on the TUNEL assay. The glutathione levels in the Riluzole group were also significantly greater than the control group. DISCUSSION Our findings suggest that Riluzole can potentially reduce the effects of IRI on skeletal muscle. This is potentially due to the ability of Riluzole to block sodium channels, decreasing action potentials and therefore glutamate release. It also acts to decrease intracellular calcium levels, which prevents apoptosis. Riluzole is a promising drug for the prevention of IRI in skeletal muscle, but further research is required.
Collapse
Affiliation(s)
- Rachel W Li
- The Medical School, the Australian National UNiversity, Canberra, ACT 2601, Australia; John Curtin School of Medical Research, The Australian National University, Garran Rd, Canberra, ACT 2601 Australia.
| | - Yi Deng
- The Medical School, the Australian National UNiversity, Canberra, ACT 2601, Australia; Canberra Hospital, Yamba Dr, Canberra, ACT 2605 Australia
| | - Hai Nam Pham
- The Medical School, the Australian National UNiversity, Canberra, ACT 2601, Australia
| | - Steven Weiss
- John Curtin School of Medical Research, The Australian National University, Garran Rd, Canberra, ACT 2601 Australia
| | - Mingming Chen
- The Medical School, the Australian National UNiversity, Canberra, ACT 2601, Australia
| | - Paul N Smith
- Canberra Hospital, Yamba Dr, Canberra, ACT 2605 Australia
| |
Collapse
|
8
|
Andreas M, Oeser C, Kainz FM, Shabanian S, Aref T, Bilban M, Messner B, Heidtmann J, Laufer G, Kocher A, Wolzt M. Intravenous Heme Arginate Induces HO-1 (Heme Oxygenase-1) in the Human Heart. Arterioscler Thromb Vasc Biol 2019; 38:2755-2762. [PMID: 30354231 DOI: 10.1161/atvbaha.118.311832] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Objective- HO-1 (heme oxygenase-1) induction may prevent or reduce ischemia-reperfusion injury. We previously evaluated its in vivo induction after a single systemic administration of heme arginate in peripheral blood mononuclear cells. The current trial was designed to assess the pharmacological tissue induction of HO-1 in the human heart with heme arginate in vivo. Approach and Results- Patients planned for conventional aortic valve replacement received placebo (n=8), 1 mg/kg (n=7) or 3 mg/kg (n=9) heme arginate infused intravenously 24 hours before surgery. A biopsy of the right ventricle was performed directly before aortic cross-clamping and after cross-clamp release. In addition, the right atrial appendage was partially removed for analysis. HO-1 protein and mRNA concentrations were measured in tissue samples and in peripheral blood mononuclear cells before to and up to 72 hours after surgery. No study medication-related adverse events occurred. A strong, dose-dependent effect on myocardial HO-1 mRNA levels was observed (right ventricle: 7.9±5.0 versus 88.6±49.1 versus 203.6±148.7; P=0.002 and right atrium: 10.8±8.8 versus 229.8±173.1 versus 392.7±195.7; P=0.001). This was paralleled by a profound increase of HO-1 protein concentration in atrial tissue (8401±3889 versus 28 585±10 692 versus 29 022±8583; P<0.001). Surgery and heme arginate infusion significantly increased HO-1 mRNA concentration in peripheral blood mononuclear cells ( P<0.001). HO-1 induction led to a significant increase of postoperative carboxyhemoglobin (1.7% versus 1.4%; P=0.041). No effect on plasma HO-1 protein levels could be detected. Conclusions- Myocardial HO-1 mRNA and protein can be dose-dependently induced by heme arginate. Protective effects of this therapeutic strategy should be evaluated in upcoming clinical trials. Clinical Trial Registration- URL: http://www.clinicaltrials.gov . Unique identifier: NCT02314780.
Collapse
Affiliation(s)
- Martin Andreas
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Claudia Oeser
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Frieda-Maria Kainz
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Shiva Shabanian
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Tandis Aref
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Martin Bilban
- Department of Laboratory Medicine (M.B.), Medical University of Vienna, Austria
- Department of Clinical Pharmacology (M.B., M.W.), Medical University of Vienna, Austria
| | - Barbara Messner
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Julian Heidtmann
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Guenther Laufer
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Alfred Kocher
- From the Department of Cardiac Surgery (M.A., C.O., F.-M.K., S.S., T.A., B.M., J.H., G.L., A.K.), Medical University of Vienna, Austria
| | - Michael Wolzt
- Department of Clinical Pharmacology (M.B., M.W.), Medical University of Vienna, Austria
| |
Collapse
|
9
|
Zhang WY, Yu Y, Yan LL, Li C, Han JY, Qin ZF, Dai Y, Yao ZH, Zhou H, Yao XS. Discovery of cardio-protective constituents of Gualou Xiebai Decoction, a classical traditional Chinese medicinal formula. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 54:318-327. [PMID: 30060904 DOI: 10.1016/j.phymed.2018.04.047] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Revised: 03/19/2018] [Accepted: 04/16/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUD Finding effective compounds of TCMs has always been the basis for achieving marker-based quality control which is currently most widely used quality control strategy. Gualou Xiebai Decoction (GLXB), a classical TCM formula, is recorded and proven as a therapy for curing coronary heart disease but the effective constituents are unidentified and the substantial basis of the therapeutic effects is not clear. PURPOSE The present research is an investigation on the chemistry of this formula aiming at finding and precisely identifying effective compounds. STUDY DESIGN AND METHODS This research started with screening for effective fractions of GLXB by rat myocardial infarction model and H9c2 cell hypoxia/reoxygenation model, then compounds in effective fractions were isolated and identified by phytochemical and spectroscopic methods. The cardio-protective activities of the compounds were tested in vitro and one of the effective compounds was taken as example to investigate the mechanisms. RESULTS The water-insoluble parts of GLXB were identified as effective parts in both in vitro and in vivo experiments. Systematic isolation of compounds in the effective fractions resulted in the isolation of 34 compounds including 7 new compounds, whereas 8 compounds were effective in protecting H9c2 cells against hypoxia/reoxygenation injury. One of the effective compounds, macrostemonoside P (MP) possibly exerted its effect by activating RISK pathway and attenuating apoptosis. CONCLUSION An array of effective constituents of GLXB were discovered, and discovery of these compounds contributed to elucidating the substantial basis for the therapeutic effects of this formula, and provides fundaments for establishing Q-markers for further reliable quality control of GLXB.
Collapse
Affiliation(s)
- Wei-Yang Zhang
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macau, PR China
| | - Yang Yu
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Lu-Lu Yan
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macau, PR China; Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, PR China
| | - Chong Li
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macau, PR China
| | - Jing-Yan Han
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, PR China; Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, PR China
| | - Zi-Fei Qin
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Yi Dai
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China
| | - Zhi-Hong Yao
- College of Pharmacy, Jinan University, Guangzhou 510632, PR China; State key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, PR China
| | - Hua Zhou
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macau, PR China.
| | - Xin-Sheng Yao
- Faculty of Chinese Medicine, Macau University of Science and Technology and State Key Laboratory of Quality Research in Chinese Medicine (Macau University of Science and Technology), Taipa, Macau, PR China; College of Pharmacy, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
10
|
Hu S, Zhu P, Zhou H, Zhang Y, Chen Y. Melatonin-Induced Protective Effects on Cardiomyocytes Against Reperfusion Injury Partly Through Modulation of IP3R and SERCA2a Via Activation of ERK1. Arq Bras Cardiol 2018. [PMID: 29538523 PMCID: PMC5831301 DOI: 10.5935/abc.20180008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background Melatonin is a neuroendocrine hormone synthesized primarily by the pineal
gland that is indicated to effectively prevent myocardial reperfusion
injury. It is unclear whether melatonin protects cardiac function from
reperfusion injury by modulating intracellular calcium homeostasis. Objective Demonstrate that melatonin protect against myocardial reperfusion injury
through modulating IP3R and SERCA2a to maintain calcium homeostasis via
activation of ERK1 in cardiomyocytes. Methods In vitro experiments were performed using H9C2 cells undergoing simulative
hypoxia/reoxygenation (H/R) induction. Expression level of ERK1, IP3R and
SERCA2a were assessed by Western Blots. Cardiomyocytes apoptosis was
detected by TUNEL. Phalloidin-staining was used to assess alteration of
actin filament organization of cardiomyocytes. Fura-2 /AM was used to
measure intracellular Ca2+ concentration. Performing in vivo
experiments, myocardial expression of IP3R and SERCA2a were detected by
immunofluorescence staining using myocardial ischemia/ reperfusion (I/R)
model in rats. Results In vitro results showed that melatonin induces ERK1 activation in
cardiomyocytes against H/R which was inhibited by PD98059 (ERK1 inhibitor).
The results showed melatonin inhibit apoptosis of cardiomyocytes and improve
actin filament organization in cardiomyocytes against H/R, because both
could be reversed by PD98059. Melatonin was showed to reduce calcium
overload, further to inhibit IP3R expression and promote SERCA2a expression
via ERK1 pathway in cardiomyocytes against H/R. Melatonin induced lower IP3R
and higher SERCA2a expression in myocardium that were reversed by
PD98059. Conclusion melatonin-induced cardioprotection against reperfusion injury is at least
partly through modulation of IP3R and SERCA2a to maintain intracellular
calcium homeostasis via activation of ERK1.
Collapse
Affiliation(s)
- Shunying Hu
- Chinese PLA General Hospital, Beijing - China
| | - Pingjun Zhu
- Chinese PLA General Hospital, Beijing - China
| | - Hao Zhou
- Chinese PLA General Hospital, Beijing - China
| | - Ying Zhang
- Chinese PLA General Hospital, Beijing - China
| | - Yundai Chen
- Chinese PLA General Hospital, Beijing - China
| |
Collapse
|
11
|
Combined Treatment With Exenatide and Cyclosporine A or Parstatin 1-26 Results in Enhanced Reduction of Infarct Size in a Rabbit Model. J Cardiovasc Pharmacol 2017; 70:34-41. [DOI: 10.1097/fjc.0000000000000492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
12
|
Berbamine postconditioning protects the heart from ischemia/reperfusion injury through modulation of autophagy. Cell Death Dis 2017; 8:e2577. [PMID: 28151484 PMCID: PMC5386498 DOI: 10.1038/cddis.2017.7] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Revised: 12/17/2016] [Accepted: 12/27/2016] [Indexed: 12/22/2022]
Abstract
Pretreatment of berbamine protects the heart from ischemia/reperfusion (I/R) injury. However it is unknown whether it has cardioprotection when given at the onset of reperfusion (postconditioning (PoC)), a protocol with more clinical impact. Autophagy is upregulated in I/R myocardium and exacerbates cardiomyocyte death during reperfusion. However, it is unknown whether the autophagy during reperfusion is regulated by berbamine. Here we investigated whether berbamine PoC (BMPoC) protects the heart through regulation of autophagy by analyzing the effects of BMPoC on infarct size and/or cell death, functional recovery and autophagy in perfused rat hearts and isolated cardiomyocytes subjected to I/R. Berbamine from 10 to 100 nM given during the first 5 min of reperfusion concentration-dependently improved post-ischemic myocardial function and attenuated cell death. Similar protections were observed in cardiomyocytes subjected to simulated I/R. Meanwhile, BMPoC prevented I/R-induced impairment of autophagosome processing in cardiomyocytes, characterized by increased LC3-II level and GFP-LC3 puncta, and decreased p62 degradation. Besides, lysosomal inhibitor chloroquine did not induce additional increase of LC3-II and P62 abundance after I/R but it reversed the effects of BMPoC in those parameters in cardiomyocytes, suggesting that I/R-impaired autophagic flux is restored by BMPoC. Moreover, I/R injury was accompanied by enhanced expression of Beclin 1, which was significantly inhibited by BMPoC. In vitro and in vivo adenovirus-mediated knockdown of Beclin 1 in myocardium and cardiomyocytes restored I/R-impaired autophagosome processing, associated with an improvement of post-ischemic recovery of myocardial contractile function and a reduction of cell death, but it did not have additive effects to BMPoC. Conversely, overexpression of Beclin 1 abolished the cardioprotection of BMPoC as did by overexpression of an essential autophagy gene Atg5. Furthermore, BMPoC-mediated cardioprotection was abolished by a specific Akt1/2 inhibitor A6730. Our results demonstrate that BMPoC confers cardioprotection by modulating autophagy during reperfusion through the activation of PI3K/Akt signaling pathway.
Collapse
|
13
|
Abstract
A core feature of ischemic heart disease is injury to cardiomyocytes (CMC). Ischemic CMC manifest the molecular mechanisms to undergo the major forms of cell injury and death, namely, oncotic necrosis, necroptosis, apoptosis and unregulated autophagy. Important modulators of ischemic injury are reperfusion and conditioning. Mitochondria have a major role in mediating the injury to CMC through membrane protein complexes referred to as death channels. Apoptosis is mediated by activation of a channel regulated by the Bcl-2 protein family leading to mitochondrial outer membrane permeabilization (MOMP). Oncotic type injury is mediated by opening of the mitochondrial permeability transition pore (mPTP). Mitochondria also have a reperfusion salvage kinase pathway (RISK). With cyclosporine A serving as a prototype, ongoing research is aimed at developing pharmacological approaches to condition and preserve mitochondrial integrity in order to promote CMC survival during episodes of myocardial ischemia.
Collapse
|
14
|
Fancelli D, Abate A, Amici R, Bernardi P, Ballarini M, Cappa A, Carenzi G, Colombo A, Contursi C, Di Lisa F, Dondio G, Gagliardi S, Milanesi E, Minucci S, Pain G, Pelicci PG, Saccani A, Storto M, Thaler F, Varasi M, Villa M, Plyte S. Cinnamic Anilides as New Mitochondrial Permeability Transition Pore Inhibitors Endowed with Ischemia-Reperfusion Injury Protective Effect in Vivo. J Med Chem 2014; 57:5333-47. [DOI: 10.1021/jm500547c] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Daniele Fancelli
- Genextra Group, Congenia s.r.l., Via Adamello 16, 20139 Milan, Italy
| | - Agnese Abate
- Genextra Group, DAC s.r.l., ,
Via Adamello 16, 20139 Milan, Italy
| | - Raffaella Amici
- Genextra Group, Congenia s.r.l., Via Adamello 16, 20139 Milan, Italy
| | - Paolo Bernardi
- Department
of Biomedical Sciences, University of Padua, Via Ugo Bassi 58/B, 35121 Padua, Italy
| | - Marco Ballarini
- Genextra Group, Congenia s.r.l., Via Adamello 16, 20139 Milan, Italy
- Department
of Experimental Oncology, European Institute of Oncology IEO, Via
Adamello 16, 20139 Milan, Italy
| | - Anna Cappa
- Genextra Group, DAC s.r.l., ,
Via Adamello 16, 20139 Milan, Italy
| | - Giacomo Carenzi
- Genextra Group, DAC s.r.l., ,
Via Adamello 16, 20139 Milan, Italy
| | - Andrea Colombo
- NiKem Research s.r.l., Via
Zambeletti 25, 20021 Baranzate, MI, Italy
| | - Cristina Contursi
- Genextra Group, Congenia s.r.l., Via Adamello 16, 20139 Milan, Italy
| | - Fabio Di Lisa
- Department
of Biomedical Sciences, University of Padua, Viale G. Colombo 3, 35131 Padua, Italy
| | - Giulio Dondio
- NiKem Research s.r.l., Via
Zambeletti 25, 20021 Baranzate, MI, Italy
| | | | - Eva Milanesi
- Genextra Group, Congenia s.r.l., Via Adamello 16, 20139 Milan, Italy
| | - Saverio Minucci
- Department
of Experimental Oncology, European Institute of Oncology IEO, Via
Adamello 16, 20139 Milan, Italy
- Department
of Biosciences, University of Milan, 20100 Milan, Italy
| | - Gilles Pain
- Genextra Group, Congenia s.r.l., Via Adamello 16, 20139 Milan, Italy
| | - Pier Giuseppe Pelicci
- Department
of Experimental Oncology, European Institute of Oncology IEO, Via
Adamello 16, 20139 Milan, Italy
| | | | - Mariangela Storto
- Genextra Group, Congenia s.r.l., Via Adamello 16, 20139 Milan, Italy
- Department
of Experimental Oncology, European Institute of Oncology IEO, Via
Adamello 16, 20139 Milan, Italy
| | - Florian Thaler
- Genextra Group, Congenia s.r.l., Via Adamello 16, 20139 Milan, Italy
| | - Mario Varasi
- Genextra Group, DAC s.r.l., ,
Via Adamello 16, 20139 Milan, Italy
| | - Manuela Villa
- Genextra Group, Congenia s.r.l., Via Adamello 16, 20139 Milan, Italy
| | - Simon Plyte
- Genextra Group, Congenia s.r.l., Via Adamello 16, 20139 Milan, Italy
| |
Collapse
|
15
|
Longnus SL, Mathys V, Dornbierer M, Dick F, Carrel TP, Tevaearai HT. Heart transplantation with donation after circulatory determination of death. Nat Rev Cardiol 2014; 11:354-63. [DOI: 10.1038/nrcardio.2014.45] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
16
|
Diez ER, Renna NF, Prado NJ, Lembo C, Ponce Zumino AZ, Vazquez-Prieto M, Miatello RM. Melatonin, given at the time of reperfusion, prevents ventricular arrhythmias in isolated hearts from fructose-fed rats and spontaneously hypertensive rats. J Pineal Res 2013; 55:166-73. [PMID: 23635352 DOI: 10.1111/jpi.12059] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2012] [Accepted: 04/12/2013] [Indexed: 01/04/2023]
Abstract
Melatonin reduces reperfusion arrhythmias when administered before coronary occlusion, but in the clinical context of acute coronary syndromes, most of the therapies are administered at the time of reperfusion. Patients frequently have physiological modifications that can reduce the response to therapeutic interventions. This work determined whether acute melatonin administration starting at the moment of reperfusion protects against ventricular arrhythmias in Langendorff-perfused hearts isolated from fructose-fed rats (FFR), a dietary model of metabolic syndrome, and from spontaneous hypertensive rats (SHR). In both experimental models, we confirmed metabolic alterations, a reduction in myocardial total antioxidant capacity and an increase in arterial pressure and NADPH oxidase activity, and in FFR, we also found a decrease in eNOS activity. Melatonin (50 μm) initiated at reperfusion after 15-min regional ischemia reduced the incidence of ventricular fibrillation from 83% to 33% for the WKY strain, from 92% to 25% in FFR, and from 100% to 33% in SHR (P = 0.0361, P = 0.0028, P = 0.0013, respectively, by Fisher's exact test, n = 12 each). Although, ventricular tachycardia incidence was high at the beginning of reperfusion, the severity of the arrhythmias progressively declined in melatonin-treated hearts. Melatonin induced a shortening of the action potential duration at the beginning of reperfusion and in the SHR group also a faster recovery of action potential amplitude. We conclude that melatonin protects against ventricular fibrillation when administered at reperfusion, and these effects are maintained in hearts from rats exposed to major cardiovascular risk factors. These results further support the ongoing translation to clinical trials of this agent.
Collapse
Affiliation(s)
- Emiliano Raúl Diez
- Instituto de Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina.
| | | | | | | | | | | | | |
Collapse
|
17
|
Roubille F, Prunier F, Barrère-Lemaire S, Leclercq F, Piot C, Kritikou EA, Rhéaume E, Busseuil D, Tardif JC. What is the Role of Erythropoietin in Acute Myocardial Infarct? Bridging the Gap Between Experimental Models and Clinical Trials. Cardiovasc Drugs Ther 2013; 27:315-31. [DOI: 10.1007/s10557-013-6461-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
18
|
Coronary artery disease and ventricular tachyarrhythmia: pathophysiology and treatment. Curr Opin Pharmacol 2013; 13:210-7. [PMID: 23357129 DOI: 10.1016/j.coph.2013.01.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 01/04/2013] [Accepted: 01/07/2013] [Indexed: 01/01/2023]
Abstract
Ventricular tachyarrhythmias are common consequences of coronary artery disease. During the prehospital phase of acute myocardial infarction (MI), ischemia-induced electrophysiological changes and genetic factors are responsible for their occurrence, but the precise pathophysiologic mechanisms are ill-understood. Primary percutaneous coronary interventions (PCIs) have decreased the incidence of ventricular tachyarrhythmias during subsequent stages, and future treatments ameliorating reperfusion injury may provide further progress. In the chronic phase, antiarrhythmic drug therapy targeted toward arrhythmogenic substrate has relatively limited value, but alternative approaches are still uncertain. By contrast, prompt arrhythmia termination by implantable cardioverter-defibrillators (ICDs) is highly effective, although risk-stratification algorithms in candidate patients are inadequate. This review explores current views in the pathophysiology and treatment of ventricular tachyarrhythmias at different clinical stages.
Collapse
|
19
|
|