1
|
Wang JZ, Li XY, Zhang M, Xiao Y, Chen L, Deng MY, Huang S, Zhou XL. Synthesis and biological evaluation of lycoctonine derivatives with cardiotonic and calcium channels inhibitory activities. Bioorg Chem 2024; 146:107297. [PMID: 38503027 DOI: 10.1016/j.bioorg.2024.107297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024]
Abstract
In our previous study, a screening of a variety of lycotonine-type diterpenoid alkaloids were screened for cardiotonic activity revealed that lycoctonine had moderate cardiac effect. In this study, a series of structurally diverse of lycoctonine were synthesized by modifying on B-ring, D-ring, E-ring, F-ring, N-atom or salt formation on lycoctonine skeleton. We evaluated the cardiotonic activity of the derivatives by isolated frog heart, aiming to identify some compounds with significantly enhanced cardiac effects, among which compound 27 with a N-isobutyl group emerged as the most promising cardiotonic candidate. Furthermore, the cardiotonic mechanism of compound 27 was preliminarily investigated. The result suggested that the cardiotonic effect of compound 27 is related to calcium channels. Patch clamp technique confirmed that the compound 27 had inhibitory effects on CaV1.2 and CaV3.2, with inhibition rates of 78.52 % ± 2.26 % and 79.05 % ± 1.59 % at the concentration of 50 μM, respectively. Subsequently, the protective effect of 27 on H9c2 cells injury induced by cobalt chloride was tested. In addition, compound 27 can alleviate CoCl2-induced myocardial injury by alleviating calcium overload. These findings suggest that compound 27 was a new structural derived from lycoctonine, which may serve as a new lead compound for the treatment of heart failure.
Collapse
Affiliation(s)
- Jian-Zhu Wang
- School of Life Science and Engineering Southwest, Jiaotong University, Chengdu, Sichuan, PR China
| | - Xiang-Yu Li
- School of Life Science and Engineering Southwest, Jiaotong University, Chengdu, Sichuan, PR China
| | - Min Zhang
- School of Life Science and Engineering Southwest, Jiaotong University, Chengdu, Sichuan, PR China; Yibin Institute of Southwest Jiaotong University, Yibin, Sichuan, PR China
| | - Yan Xiao
- School of Life Science and Engineering Southwest, Jiaotong University, Chengdu, Sichuan, PR China
| | - Lin Chen
- School of Life Science and Engineering Southwest, Jiaotong University, Chengdu, Sichuan, PR China
| | - Meng-Yi Deng
- School of Life Science and Engineering Southwest, Jiaotong University, Chengdu, Sichuan, PR China; Yibin Institute of Southwest Jiaotong University, Yibin, Sichuan, PR China
| | - Shuai Huang
- School of Life Science and Engineering Southwest, Jiaotong University, Chengdu, Sichuan, PR China; Yibin Institute of Southwest Jiaotong University, Yibin, Sichuan, PR China.
| | - Xian-Li Zhou
- School of Life Science and Engineering Southwest, Jiaotong University, Chengdu, Sichuan, PR China; Yibin Institute of Southwest Jiaotong University, Yibin, Sichuan, PR China.
| |
Collapse
|
2
|
Baglini E, Poggetti V, Cavallini C, Petroni D, Forini F, Nicolini G, Barresi E, Salerno S, Costa B, Iozzo P, Neglia D, Menichetti L, Taliani S, Da Settimo F. Targeting the Translocator Protein (18 kDa) in Cardiac Diseases: State of the Art and Future Opportunities. J Med Chem 2024; 67:17-37. [PMID: 38113353 PMCID: PMC10911791 DOI: 10.1021/acs.jmedchem.3c01716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 11/16/2023] [Accepted: 11/24/2023] [Indexed: 12/21/2023]
Abstract
Mitochondria dysfunctions are typical hallmarks of cardiac disorders (CDs). The multiple tasks of this energy-producing organelle are well documented, but its pathophysiologic involvement in several manifestations of heart diseases, such as altered electromechanical coupling, excitability, and arrhythmias, is still under investigation. The human 18 kDa translocator protein (TSPO) is a protein located on the outer mitochondrial membrane whose expression is altered in different pathological conditions, including CDs, making it an attractive therapeutic and diagnostic target. Currently, only a few TSPO ligands are employed in CDs and cardiac imaging. In this Perspective, we report an overview of the emerging role of TSPO at the heart level, focusing on the recent literature concerning the development of TSPO ligands used for fighting and imaging heart-related disease conditions. Accordingly, targeting TSPO might represent a successful strategy to achieve novel therapeutic and diagnostic strategies to unravel the fundamental mechanisms and to provide solutions to still unanswered questions in CDs.
Collapse
Affiliation(s)
- Emma Baglini
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Valeria Poggetti
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Chiara Cavallini
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Debora Petroni
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Francesca Forini
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Giuseppina Nicolini
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Elisabetta Barresi
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Silvia Salerno
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Barbara Costa
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Patricia Iozzo
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Danilo Neglia
- Fondazione
CNR/Regione Toscana Gabriele Monasterio, Cardiovascular and Imaging
Departments, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Luca Menichetti
- Institute
of Clinical Physiology, National Research Council of Italy, CNR Research Area, Via G. Moruzzi 1, Pisa 56124, Italy
| | - Sabrina Taliani
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| | - Federico Da Settimo
- Department
of Pharmacy, University of Pisa, Via Bonanno 6, Pisa 56126, Italy
| |
Collapse
|
3
|
Piao L, Fang YH, Fisher M, Hamanaka RB, Ousta A, Wu R, Mutlu GM, Garcia AJ, Archer SL, Sharp WW. Dynamin-related protein 1 is a critical regulator of mitochondrial calcium homeostasis during myocardial ischemia/reperfusion injury. FASEB J 2024; 38:e23379. [PMID: 38133921 DOI: 10.1096/fj.202301040rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 11/17/2023] [Accepted: 12/04/2023] [Indexed: 12/23/2023]
Abstract
Dynamin-related protein 1 (Drp1) is a cytosolic GTPase protein that when activated translocates to the mitochondria, meditating mitochondrial fission and increasing reactive oxygen species (ROS) in cardiomyocytes. Drp1 has shown promise as a therapeutic target for reducing cardiac ischemia/reperfusion (IR) injury; however, the lack of specificity of some small molecule Drp1 inhibitors and the reliance on the use of Drp1 haploinsufficient hearts from older mice have left the role of Drp1 in IR in question. Here, we address these concerns using two approaches, using: (a) short-term (3 weeks), conditional, cardiomyocyte-specific, Drp1 knockout (KO) and (b) a novel, highly specific Drp1 GTPase inhibitor, Drpitor1a. Short-term Drp1 KO mice exhibited preserved exercise capacity and cardiac contractility, and their isolated cardiac mitochondria demonstrated increased mitochondrial complex 1 activity, respiratory coupling, and calcium retention capacity compared to controls. When exposed to IR injury in a Langendorff perfusion system, Drp1 KO hearts had preserved contractility, decreased reactive oxygen species (ROS), enhanced mitochondrial calcium capacity, and increased resistance to mitochondrial permeability transition pore (MPTP) opening. Pharmacological inhibition of Drp1 with Drpitor1a following ischemia, but before reperfusion, was as protective as Drp1 KO for cardiac function and mitochondrial calcium homeostasis. In contrast to the benefits of short-term Drp1 inhibition, prolonged Drp1 ablation (6 weeks) resulted in cardiomyopathy. Drp1 KO hearts were also associated with decreased ryanodine receptor 2 (RyR2) protein expression and pharmacological inhibition of the RyR2 receptor decreased ROS in post-IR hearts suggesting that changes in RyR2 may have a role in Drp1 KO mediated cardioprotection. We conclude that Drp1-mediated increases in myocardial ROS production and impairment of mitochondrial calcium handling are key mechanisms of IR injury. Short-term inhibition of Drp1 is a promising strategy to limit early myocardial IR injury which is relevant for the therapy of acute myocardial infarction, cardiac arrest, and heart transplantation.
Collapse
Affiliation(s)
- Lin Piao
- Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Yong-Hu Fang
- Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Michael Fisher
- Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Robert B Hamanaka
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Alaa Ousta
- Department of Emergency Medicine, Duke University, Durham, North Carolina, USA
| | - Rongxu Wu
- Section of Cardiology, Department of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Gökhan M Mutlu
- Section of Pulmonary and Critical Care, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Institute for Integrative Physiology, University of Chicago, Chicago, Illinois, USA
| | - Alfredo J Garcia
- Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Institute for Integrative Physiology, University of Chicago, Chicago, Illinois, USA
- The University of Chicago Neuroscience Institute, University of Chicago, Chicago, Illinois, USA
| | - Stephen L Archer
- Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Willard W Sharp
- Section of Emergency Medicine, Department of Medicine, University of Chicago, Chicago, Illinois, USA
- Institute for Integrative Physiology, University of Chicago, Chicago, Illinois, USA
| |
Collapse
|
4
|
Li MR, Luo XJ, Peng J. Role of sonic hedgehog signaling pathway in the regulation of ion channels: focus on its association with cardio-cerebrovascular diseases. J Physiol Biochem 2023; 79:719-730. [PMID: 37676576 DOI: 10.1007/s13105-023-00982-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 08/25/2023] [Indexed: 09/08/2023]
Abstract
Sonic hedgehog (SHH) signaling is vital for cell differentiation and proliferation during embryonic development, yet its role in cardiac, cerebral, and vascular pathophysiology is under debate. Recent studies have demonstrated that several compounds of SHH signaling regulate ion channels, which in turn affect the behavior of target cells. Some of these ion channels are involved in the cardio-cerebrovascular system. Here, we first reviewed the SHH signaling cascades, then its interaction with ion channels, and their impact on cardio-cerebrovascular diseases. Considering the complex cross talk of SHH signaling with other pathways that also affect ion channels and their potential impact on the cardio-cerebrovascular system, we highlight the necessity of thoroughly studying the effect of SHH signaling on ion homeostasis, which could serve as a novel mechanism for cardio-cerebrovascular diseases. Activation of SHH signaling influence ion channels activity, which in turn influence ion homeostasis, membrane potential, and electrophysiology, could serve as a novel strategy for cardio-cerebrovascular diseases.
Collapse
Affiliation(s)
- Ming-Rui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiu-Ju Luo
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, 410013, China.
| | - Jun Peng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
5
|
Liu GY, Xie WL, Wang YT, Chen L, Xu ZZ, Lv Y, Wu QP. Calpain: the regulatory point of myocardial ischemia-reperfusion injury. Front Cardiovasc Med 2023; 10:1194402. [PMID: 37456811 PMCID: PMC10346867 DOI: 10.3389/fcvm.2023.1194402] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Calpain is a conserved cysteine protease readily expressed in several mammalian tissues, which is usually activated by Ca2+ and with maximum activity at neutral pH. The activity of calpain is tightly regulated because its aberrant activation will nonspecifically cleave various proteins in cells. Abnormally elevation of Ca2+ promotes the abnormal activation of calpain during myocardial ischemia-reperfusion, resulting in myocardial injury and cardiac dysfunction. In this paper, we mainly reviewed the effects of calpain in various programmed cell death (such as apoptosis, mitochondrial-mediated necrosis, autophagy-dependent cell death, and parthanatos) in myocardial ischemia-reperfusion. In addition, we also discussed the abnormal activation of calpain during myocardial ischemia-reperfusion, the effect of calpain on myocardial repair, and the possible future research directions of calpain.
Collapse
Affiliation(s)
- Guo-Yang Liu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Wan-Li Xie
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yan-Ting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Lu Chen
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Zhen-Zhen Xu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Yong Lv
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| | - Qing-Ping Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Anesthesiology and Resuscitation (Huazhong University of Science and Technology), Ministry of Education, Wuhan, China
| |
Collapse
|
6
|
Modification of Ischemia/Reperfusion-Induced Alterations in Subcellular Organelles by Ischemic Preconditioning. Int J Mol Sci 2022; 23:ijms23073425. [PMID: 35408783 PMCID: PMC8998910 DOI: 10.3390/ijms23073425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/17/2022] [Accepted: 03/18/2022] [Indexed: 02/07/2023] Open
Abstract
It is now well established that ischemia/reperfusion (I/R) injury is associated with the compromised recovery of cardiac contractile function. Such an adverse effect of I/R injury in the heart is attributed to the development of oxidative stress and intracellular Ca2+-overload, which are known to induce remodeling of subcellular organelles such as sarcolemma, sarcoplasmic reticulum, mitochondria and myofibrils. However, repeated episodes of brief periods of ischemia followed by reperfusion or ischemic preconditioning (IP) have been shown to improve cardiac function and exert cardioprotective actions against the adverse effects of prolonged I/R injury. This protective action of IP in attenuating myocardial damage and subcellular remodeling is likely to be due to marked reductions in the occurrence of oxidative stress and intracellular Ca2+-overload in cardiomyocytes. In addition, the beneficial actions of IP have been attributed to the depression of proteolytic activities and inflammatory levels of cytokines as well as the activation of the nuclear factor erythroid factor 2-mediated signal transduction pathway. Accordingly, this review is intended to describe some of the changes in subcellular organelles, which are induced in cardiomyocytes by I/R for the occurrence of oxidative stress and intracellular Ca2+-overload and highlight some of the mechanisms for explaining the cardioprotective effects of IP.
Collapse
|
7
|
Wang S, Zhou Y, Luo Y, Kan R, Chen J, Xuan H, Wang C, Chen J, Xu T, Li D. SERCA2a ameliorates cardiomyocyte T-tubule remodeling via the calpain/JPH2 pathway to improve cardiac function in myocardial ischemia/reperfusion mice. Sci Rep 2021; 11:2037. [PMID: 33479390 PMCID: PMC7820433 DOI: 10.1038/s41598-021-81570-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 01/07/2021] [Indexed: 12/21/2022] Open
Abstract
Transverse-tubules (T-tubules) play pivotal roles in Ca2+-induced, Ca2+ release and excitation–contraction coupling in cardiomyocytes. The purpose of this study was to uncover mechanisms where sarco/endoplasmic reticulum Ca2+ ATPase (SERCA2a) improved cardiac function through T-tubule regulation during myocardial ischemia/reperfusion (I/R). SERCA2a protein expression, cytoplasmic [Ca2+]i, calpain activity, junctophilin-2 (JPH2) protein expression and intracellular localization, cardiomyocyte T-tubules, contractility and calcium transients in single cardiomyocytes and in vivo cardiac functions were all examined after SERCA2a knockout and overexpression, and Calpain inhibitor PD150606 (PD) pretreatment, following myocardial I/R. This comprehensive approach was adopted to clarify SERCA2a mechanisms in improving cardiac function in mice. Calpain was activated during myocardial I/R, and led to the proteolytic cleavage of JPH2. This altered the T-tubule network, the contraction function/calcium transients in cardiomyocytes and in vivo cardiac functions. During myocardial I/R, PD pretreatment upregulated JPH2 expression and restored it to its intracellular location, repaired the T-tubule network, and contraction function/calcium transients of cardiomyocytes and cardiac functions in vivo. SERCA2a suppressed calpain activity via [Ca2+]i, and ameliorated these key indices. Our results suggest that SERCA2a ameliorates cardiomyocyte T-tubule remodeling via the calpain/JPH2 pathway, thereby improving cardiac function in myocardial I/R mice.
Collapse
Affiliation(s)
- Shuai Wang
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - You Zhou
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Yuanyuan Luo
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China
| | - Rongsheng Kan
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Jingwen Chen
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China
| | - Haochen Xuan
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China
| | - Chaofan Wang
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China
| | - Junhong Chen
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China
| | - Tongda Xu
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China.
| | - Dongye Li
- Institute of Cardiovascular Disease Research, Xuzhou Medical University, 84 West Huaihai Road, Xuzhou, 221002, Jiangsu, People's Republic of China. .,Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221006, Jiangsu, People's Republic of China.
| |
Collapse
|
8
|
Zeng Z, Ma H, Chen J, Huang N, Zhang Y, Su Y, Zhang H. Knockdown of miR-1275 protects against cardiomyocytes injury through promoting neuromedin U type 1 receptor. Cell Cycle 2020; 19:3639-3649. [PMID: 33323026 DOI: 10.1080/15384101.2020.1860310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The present study aimed to assess the role of miR-1275 in cardiac ischemia reperfusion injury. H9 human embryonic stem cell (hESC)-derived cardiomyocytes stimulated by oxygen-glucose deprivation/reoxygenation (OGD/R) were used to simulate myocardial injury in vitro. miR-1275 expression levels in cells were measured by RT-qPCR. The release of lactate dehydrogenase (LDH) and creatine kinase (CK) was examined through LDH and CK ELISA kits. Cell apoptosis was detected through flow cytometry. A Fura-2 Calcium Flux Assay Kit and a Fluo-4 assay kit were used to determine the Ca2+ concentration. Expression levels of proteins were tested by Western blotting. The binding effect of miR-1275 and neuromedin U type 1 receptor (NMUR1) was detected by dual-luciferase activity assay. The results showed that miR-1275 was upregulated in OGD/R-stimulated cardiomyocytes. Inhibition of miR-1275 suppressed the increased activity of LDH and CK, cell apoptosis, reactive oxygen species (ROS) production, intracellular Ca2+ concentration and sarcoplasmic reticulum (SR) Ca2+ leak induced by OGD/R treatment in cardiomyocytes. miR-1275 directly targets 3'UTR of NMUR1 and negatively regulates NMUR1 expression. Silence of NMUR1 abolished the protecting effect of the miR-1275 antagomir on myocardial OGD/R injury. Our study indicated that the miR-1275 antagomir protects cardiomyocytes from OGD/R injury through the promotion of NMUR1.
Collapse
Affiliation(s)
- Zhu Zeng
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Haixin Ma
- Medical Department, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Jing Chen
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Nina Huang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Yudan Zhang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Yufei Su
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| | - Huifang Zhang
- Department of Emergency, The Affiliated Children Hospital of Xi'an Jiaotong University , Xi'an, Shaanxi, China
| |
Collapse
|
9
|
Wang R, Wang M, He S, Sun G, Sun X. Targeting Calcium Homeostasis in Myocardial Ischemia/Reperfusion Injury: An Overview of Regulatory Mechanisms and Therapeutic Reagents. Front Pharmacol 2020; 11:872. [PMID: 32581817 PMCID: PMC7296066 DOI: 10.3389/fphar.2020.00872] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/27/2020] [Indexed: 12/17/2022] Open
Abstract
Calcium homeostasis plays an essential role in maintaining excitation–contraction coupling (ECC) in cardiomyocytes, including calcium release, recapture, and storage. Disruption of calcium homeostasis may affect heart function, leading to the development of various heart diseases. Myocardial ischemia/reperfusion (MI/R) injury may occur after revascularization, which is a treatment used in coronary heart disease. MI/R injury is a complex pathological process, and the main cause of increased mortality and disability after treatment of coronary heart disease. However, current methods and drugs for treating MI/R injury are very scarce, not ideal, and have limitations. Studies have shown that MI/R injury can cause calcium overload that can further aggravate MI/R injury. Therefore, we reviewed the effects of critical calcium pathway regulators on MI/R injury and drew an intuitive diagram of the calcium homeostasis pathway. We also summarized and analyzed calcium pathway-related or MI/R drugs under research or marketing by searching Therapeutic Target and PubMed Databases. The data analysis showed that six drugs and corresponding targets are used to treat MI/R injury and involved in calcium signaling pathways. We emphasize the relevance of further detailed investigation of MI/R injury and calcium homeostasis and the therapeutic role of calcium homeostasis in MI/R injury, which bridges basic research and clinical applications of MI/R injury.
Collapse
Affiliation(s)
- Ruiying Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,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, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,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, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Shuaibing He
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,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, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Guibo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,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, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaobo Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Beijing Key Laboratory of Innovative Drug Discovery of Traditional Chinese Medicine (Natural Medicine) and Translational Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,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, China.,Key Laboratory of Efficacy Evaluation of Chinese Medicine Against Glycolipid Metabolic Disorders, State Administration of Traditional Chinese Medicine, Institute of Medicinal Plant Development, Peking Union Medical College & Chinese Academy of Medical Sciences, Beijing, China.,Key Laboratory of New Drug Discovery Based on Classic Chinese Medicine Prescription, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
10
|
Zuo W, Liu N, Zeng Y, Liu Y, Li B, Wu K, Xiao Y, Liu Q. CD38: A Potential Therapeutic Target in Cardiovascular Disease. Cardiovasc Drugs Ther 2020; 35:815-828. [PMID: 32472237 DOI: 10.1007/s10557-020-07007-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Substantial research has demonstrated the association between cardiovascular disease and the dysregulation of intracellular calcium, ageing, reduction in nicotinamide adenine dinucleotide NAD+ content, and decrease in sirtuin activity. CD38, which comprises the soluble type, type II, and type III, is the main NADase in mammals. This molecule catalyses the production of cyclic adenosine diphosphate ribose (cADPR), nicotinic acid adenine dinucleotide phosphate (NAADP), and adenosine diphosphate ribose (ADPR), which stimulate the release of Ca2+, accompanied by NAD+ consumption and decreased sirtuin activity. Therefore, the relationship between cardiovascular disease and CD38 has been attracting increased attention. In this review, we summarize the structure, regulation, function, targeted drug development, and current research on CD38 in the cardiac context. More importantly, we provide original views about the as yet elusive mechanisms of CD38 action in certain cardiovascular disease models. Based on our review, we predict that CD38 may serve as a novel therapeutic target in cardiovascular disease in the future.
Collapse
Affiliation(s)
- Wanyun Zuo
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Na Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yunhong Zeng
- Department of Cardiology, Hunan Children's Hospital, No. 86 Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, China
| | - Yaozhong Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Biao Li
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Keke Wu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China
| | - Yunbin Xiao
- Department of Cardiology, Hunan Children's Hospital, No. 86 Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, China.
| | - Qiming Liu
- Department of Cardiovascular Medicine, The Second Xiangya Hospital of Central South University, No. 139 Middle Renmin Road, Furong District, Changsha, 410011, Hunan, China.
| |
Collapse
|
11
|
Ghaleh B, Thireau J, Cazorla O, Soleti R, Scheuermann V, Bizé A, Sambin L, Roubille F, Andriantsitohaina R, Martinez MC, Lacampagne A. Cardioprotective effect of sonic hedgehog ligand in pig models of ischemia reperfusion. Am J Cancer Res 2020; 10:4006-4016. [PMID: 32226535 PMCID: PMC7086352 DOI: 10.7150/thno.40461] [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: 09/19/2019] [Accepted: 01/20/2020] [Indexed: 12/20/2022] Open
Abstract
Sonic hedgehog (SHH) signaling pathway is involved in embryonic tissue patterning and development. Our previous work identified, in small rodent model of ischemia reperfusion, SHH as a specific efficient tool to reduce infarct size and subsequent arrhythmias by preventing ventricular repolarization abnormalities. The goal of the present study was to provide a proof of concept of the cardioprotective effect of SHH ligand in a porcine model of acute ischemia. Methods: The antiarrhythmic effect of SHH, either by a recombinant peptide (N-SHH) or shed membrane microparticles harboring SHH ligand (MPsSHH+), was evaluated in a first set of pigs following a short (25 min) coronary artery occlusion (CAO) followed by 24 hours-reperfusion (CAR) (Protocol A). The infarct-limiting effect was evaluated on a second set of pigs with 40 min of coronary artery occlusion followed by 24 hours reperfusion (Protocol B). Electrocardiogram (ECG) was recorded and arrhythmia's scores were evaluated. Area at risk and myocardial infarct size were quantified. Results: In protocol A, administration of N-SHH 15 min. after the onset of coronary occlusion significantly reduced the occurrence of ventricular fibrillation compared to control group. Evaluation of arrhythmic score showed that N-SHH treatment significantly reduced the overall occurrence of arrhythmias. In protocol B, massive infarction was observed in control animals. Either N-SHH or MPsSHH+ treatment reduced significantly the infarct size with a concomitant increase of salvaged area. The reduction in infarct size was both accompanied by a significant decrease in systemic biomarkers of myocardial injury, i.e., cardiac troponin I and fatty acid-binding protein and an increase of eNOS activation. Conclusions: We show for the first time in a large mammalian model that the activation of the SHH pathway by N-SHH or MPsSHH+ offers a potent protection of the heart to ischemia-reperfusion by preventing the reperfusion arrhythmias, reducing the infarct area and the circulating levels of biomarkers for myocardial injury. These data open up potentially theranostic prospects for patients suffering from myocardial infarction to prevent the occurrence of arrhythmias and reduce myocardial tissue damage.
Collapse
|
12
|
Guidarelli A, Cerioni L, Fiorani M, Catalani A, Cantoni O. Arsenite-Induced Mitochondrial Superoxide Formation: Time and Concentration Requirements for the Effects of the Metalloid on the Endoplasmic Reticulum and Mitochondria. J Pharmacol Exp Ther 2020; 373:62-71. [DOI: 10.1124/jpet.119.262469] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/13/2020] [Indexed: 02/06/2023] Open
|
13
|
Patel P, Karch J. Regulation of cell death in the cardiovascular system. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 353:153-209. [PMID: 32381175 DOI: 10.1016/bs.ircmb.2019.11.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The adult heart is a post-mitotic terminally differentiated organ; therefore, beyond development, cardiomyocyte cell death is maladaptive. Heart disease is the leading cause of death in the world and aberrant cardiomyocyte cell death is the underlying problem for most cardiovascular-related diseases and fatalities. In this chapter, we will discuss the different cell death mechanisms that engage during normal cardiac development, aging, and disease states. The most abundant loss of cardiomyocytes occurs during a myocardial infarction, when the blood supply to the heart is obstructed, and the affected myocardium succumbs to cell death. Originally, this form of cell death was considered to be unregulated; however, research from the last half a century clearly demonstrates that this form of cell death is multifaceted and employees various degrees of regulation. We will explore all of the cell death pathways that have been implicated in this disease state and the potential interplay between them. Beyond myocardial infarction, we also explore the role and mechanisms of cardiomyocyte cell death in heart failure, myocarditis, and chemotherapeutic-induced cardiotoxicity. Inhibition of cardiomyocyte cell death has extensive therapeutic potential that will increase the longevity and health of the human heart.
Collapse
Affiliation(s)
- Pooja Patel
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States
| | - Jason Karch
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, United States; Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
14
|
Yuan M, Meng XW, Ma J, Liu H, Song SY, Chen QC, Liu HY, Zhang J, Song N, Ji FH, Peng K. Dexmedetomidine protects H9c2 cardiomyocytes against oxygen-glucose deprivation/reoxygenation-induced intracellular calcium overload and apoptosis through regulating FKBP12.6/RyR2 signaling. DRUG DESIGN DEVELOPMENT AND THERAPY 2019; 13:3137-3149. [PMID: 31564830 PMCID: PMC6730549 DOI: 10.2147/dddt.s219533] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 08/23/2019] [Indexed: 12/30/2022]
Abstract
Purpose Intracellular calcium ([Ca2+]i) overload is a major cause of cell injury during myocardial ischemia/reperfusion (I/R). Dexmedetomidine (DEX) has been shown to exert anti-inflammatory and organ protective effects. This study aimed to investigate whether pretreatment with DEX could protect H9c2 cardiomyocytes against oxygen-glucose deprivation/reoxygenation (OGD/R) injury through regulating the Ca2+ signaling. Methods H9c2 cardiomyocytes were subjected to OGD for 12 h, followed by 3 h of reoxygenation. DEX was administered 1 h prior to OGD/R. Cell viability, lactate dehydrogenase (LDH) release, level of [Ca2+]i, cell apoptosis, and the expression of 12.6-kd FK506-binding protein/ryanodine receptor 2 (FKBP12.6/RyR2) and caspase-3 were assessed. Results Cells exposed to OGD/R had decreased cell viability, increased LDH release, elevated [Ca2+]i level and apoptosis rate, down-regulated expression of FKBP12.6, and up-regulated expression of phosphorylated-Ser2814-RyR2 and cleaved caspase-3. Pretreatment with DEX significantly blocked the above-mentioned changes, alleviating the OGD/R-induced injury in H9c2 cells. Moreover, knockdown of FKBP12.6 by small interfering RNA abolished the protective effects of DEX. Conclusion This study indicates that DEX pretreatment protects the cardiomyocytes against OGD/R-induced injury by inhibiting [Ca2+]i overload and cell apoptosis via regulating the FKBP12.6/RyR2 signaling. DEX may be used for preventing cardiac I/R injury in the clinical settings.
Collapse
Affiliation(s)
- Mei Yuan
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China.,Department of Anesthesiology, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, Jiangsu 215008, People's Republic of China
| | - Xiao-Wen Meng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Jiao Ma
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Hong Liu
- Department of Anesthesiology and Pain Medicine, University of California Davis Health System, Sacramento, CA 95817, USA
| | - Shao-Yong Song
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Qing-Cai Chen
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Hua-Yue Liu
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Juan Zhang
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Nan Song
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Fu-Hai Ji
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| | - Ke Peng
- Department of Anesthesiology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, People's Republic of China
| |
Collapse
|
15
|
Transient Receptor Potential Canonical Channel Blockers Improve Ventricular Contractile Functions After Ischemia/Reperfusion in a Langendorff-perfused Mouse Heart Model. J Cardiovasc Pharmacol 2019; 71:248-255. [PMID: 29389740 DOI: 10.1097/fjc.0000000000000566] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Reperfusion of ischemic myocardium is accompanied by intracellular Ca overload, leading to cardiac dysfunction. However, the mechanisms underlying intracellular Ca overload have yet to be fully elucidated. The mechanism may involve the activation of store-operated Ca entry, which is primarily mediated through the transient receptor potential canonical (TRPC) channels. This study was undertaken to examine the possible involvement of TRPC channels in the development of contractile dysfunction associated with reperfusion of ischemic myocardium using a mouse heart model. The functional expression of TRPC channels was confirmed in mouse ventricular myocytes using immunocytochemistry, Western blotting, and patch-clamp experiments. The left ventricular functions were assessed by measuring left ventricular end-diastolic pressure, left ventricular developed pressure, and its first derivatives in a Langendorff-perfused mouse heart subjected to 30 minutes of normothermic (37°C) global ischemia followed by 60 minutes of reperfusion. Under control conditions, left ventricular functions were deteriorated during reperfusion, which was significantly ameliorated by administration of the TRPC channel blockers 2-aminoethoxydiphenyl borate and La during initial 5 minutes of reperfusion. Our findings suggest that TRPC channels are involved in mediating contractile dysfunction during reperfusion of ischemic myocardium and detect TRPC channels as a potential therapeutic target for preventing myocardial ischemia/reperfusion injury.
Collapse
|
16
|
Lin HP, Zheng YQ, Zhou ZP, Wang GX, Guo PF. Ryanodine receptor antagonism alleviates skeletal muscle ischemia reperfusion injury by modulating TNF-α and IL-10. Clin Hemorheol Microcirc 2018; 70:51-58. [PMID: 29660904 DOI: 10.3233/ch-170276] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Intracellular calcium overload has been implicated in various pathological conditions including ischemia reperfusion injury. This study aims to explore the effect and probable mechanism of dantrolene, a ryanodine receptor and intracellular calcium antagonist, on the skeletal muscle ischemia reperfusion injury. MATERIALS AND METHODS SD rats were randomly divided into three groups: sham group which underwent anaesthesia and exposure of femoral vein, reperfusion group that received 2 h ischemia and the amount of diluent via femoral vein before 4 h reperfusion, dantrolene group that underwent 2 h ischemia and was given 2 mg/kg dantrolene via femoral vein before 4 h reperfusion. The parameters measured at the end of reperfusion included serum maleic dialdehyde (MDA), tissue myeloperoxidase (MPO) and muscle histology, as well as serum TNF-α and IL-10. RESULTS Levels of MDA, MPO and TNF-α increased in the reperfusion group, whereas the relevant expressions in the dantrolene group decreased significantly. Histological examination demonstrated significant improvements between the same both groups. IL-10 reflected the protection observed above with a significant up-regulation of expression after dantrolene administration. CONCLUSION Ryanodine receptor antagonist dantrolene exerted a significant protective effect against the inflammatory injury of skeletal muscle ischemia reperfusion. The underlying molecular mechanism is probably related to the suppression of TNF-α levels and the increment of IL-10 expression.
Collapse
Affiliation(s)
- Hai-Peng Lin
- Department of General Surgery, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Yan-Qing Zheng
- Department of E.N.T, Quanzhou Women's and Children's Hospital, Quanzhou, China
| | - Zhi-Ping Zhou
- Department of General Surgery, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Gao-Xiong Wang
- Department of General Surgery, the Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Ping-Fan Guo
- Department of Vascular Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
17
|
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
|
18
|
Eraslan E, Tanyeli A, Polat E, Polat E. 8-Br-cADPR, a TRPM2 ion channel antagonist, inhibits renal ischemia-reperfusion injury. J Cell Physiol 2018; 234:4572-4581. [PMID: 30191993 DOI: 10.1002/jcp.27236] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2018] [Accepted: 08/24/2018] [Indexed: 01/25/2023]
Abstract
The transient receptor potential melastatin-2 (TRPM2) channel belongs to the transient receptor potential channel superfamily and is a cation channel permeable to Na+ and Ca 2+ . The TRPM2 ion channel is expressed in the kidney and can be activated by various molecules such as hydrogen peroxide, calcium, and cyclic adenosine diphosphate (ADP)-ribose (cADPR) that are produced during acute kidney injury. In this study, we investigated the role of 8-bromo-cyclic ADP-ribose (8-Br-cADPR; a cADPR antagonist) in renal ischemia-reperfusion injury using biochemical and histopathological parameters. CD38, cADPR, tumor necrosis factor-α, interleukin-1β, and myeloperoxidase (inflammatory markers), urea and creatinine, hydrogen peroxide (oxidant), and catalase (antioxidant enzyme) levels that increase with ischemia-reperfusion injury decreased in the groups treated with 8-Br-cADPR. In addition, renin levels were elevated in the groups treated with 8-Br-cADPR. Histopathological examination revealed that 8-Br-cADPR reduced renal damage and the expression of caspase-3 and TRPM2. Our results suggest that the inhibition of TRPM2 ion channel may be a new treatment modality for ischemic acute kidney injury.
Collapse
Affiliation(s)
- Ersen Eraslan
- Department of Physiology, Faculty of Medicine, University of Bozok, Yozgat, Turkey
| | - Ayhan Tanyeli
- Department of Physiology, Faculty of Medicine, University of Atatürk, Erzurum, Turkey
| | - Elif Polat
- Department of Biochemistry, Faculty of Medicine, University of Atatürk, Erzurum, Turkey
| | - Elif Polat
- Department of Histology and Embryology, Faculty of Medicine, University of Namık Kemal, Tekirdağ, Turkey
| |
Collapse
|
19
|
Two-pore channels and disease. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2018; 1865:1678-1686. [PMID: 29746898 PMCID: PMC6162333 DOI: 10.1016/j.bbamcr.2018.05.004] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/03/2018] [Indexed: 01/25/2023]
Abstract
Two-pore channels (TPCs) are Ca2+-permeable endo-lysosomal ion channels subject to multi-modal regulation. They mediate their physiological effects through releasing Ca2+ from acidic organelles in response to cues such as the second messenger, NAADP. Here, we review emerging evidence linking TPCs to disease. We discuss how perturbing both local and global Ca2+ changes mediated by TPCs through chemical and/or molecular manipulations can induce or reverse disease phenotypes. We cover evidence from models of Parkinson's disease, non-alcoholic fatty liver disease, Ebola infection, cancer, cardiac dysfunction and diabetes. A need for more drugs targeting TPCs is identified.
Collapse
|
20
|
Ragone MI, Bonazzola P, Colareda GA, Lazarte ML, Bruno F, Consolini AE. Cardioprotection of stevioside on stunned rat hearts: A mechano-energetical study. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2017; 35:18-26. [PMID: 28991641 DOI: 10.1016/j.phymed.2017.08.022] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 07/12/2017] [Accepted: 08/20/2017] [Indexed: 06/07/2023]
Abstract
BACKGROUND The sweetener and hypoglycemic properties of stevioside (STV) are well known, as the main component of the plant Stevia rebaudiana. Given its extensive use in diabetic patients, it was of interest to evaluate its effects on the most frequent cardiovascular disease, the coronary insufficiency. PURPOSE To study whether STV could be cardioprotective against ischemia-reperfusion (I/R) in a model of "stunning" in rat hearts. STUDY DESIGN A preclinical study was performed in isolated hearts from rats in the following groups: non-treated rats whose hearts were perfused with STV 0.3 mg/ml and their controls (C) exposed to either moderate stunning (20 min I/45 min R) or severe stunning (30 min I/45 min R), and a group of rats orally treated with STV 25 mg/kg/day in the drink water during 1 week before the experiment of severe stunning in the isolated hearts were done. METHODS The mechano-calorimetrical performance of isolated beating hearts was recorded during stabilization period with control Krebs perfusion inside a calorimeter, with or without 0.3 mg/ml STV before the respective period of I/R. The left ventricular maximal developed pressure (P) and total heat rate (Ht) were continuously measured. RESULTS Both, orally administered and perfused STV improved the post-ischemic contractile recovery (PICR, as % of initial control P) and the total muscle economy (P/Ht) after the severe stunning, but only improved P/Ht in moderate stunning. However, STV increased the diastolic pressure (LVEDP) during I/R in both stunning models. For studying the mechanism of action, ischemic hearts were reperfused with 10 mM caffeine-36 mM Na+-Krebs to induce a contracture dependent on sarcorreticular Ca2+ content, whose relaxation mainly depends on mitochondrial Ca2+ uptake. STV at 0.3 mg/ml increased the area-under-curve of the caffeine-dependent contracture (AUC-LVP). Moreover, at room temperature STV increased the mitochondrial Ca2+ uptake measured by Rhod-2 fluorescence in rat cardiomyocytes, but prevented the [Ca2+]m overload assessed by caffeine-dependent SR release. CONCLUSIONS Results suggest that STV is cardioprotective against I/R under oral administration or direct perfusion in hearts. The mechanism includes the regulation of the myocardial calcium homeostasis and the energetic during I/R in several sites, mainly reducing mitochondrial Ca2+ overload and increasing the sarcorreticular Ca2+ store.
Collapse
Affiliation(s)
- María I Ragone
- Cátedra de Farmacología, Grupo de Farmacología Experimental y Energética Cardíaca, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina; Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Patricia Bonazzola
- Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina; Instituto de Investigaciones Cardiológicas, Facultad de Medicina, Universidad de Buenos Aires (UBA-CONICET), Ciudad Autónoma de Buenos Aires, Argentina
| | - Germán A Colareda
- Cátedra de Farmacología, Grupo de Farmacología Experimental y Energética Cardíaca, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - María Lara Lazarte
- Cátedra de Farmacología, Grupo de Farmacología Experimental y Energética Cardíaca, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - Fiorella Bruno
- Cátedra de Farmacología, Grupo de Farmacología Experimental y Energética Cardíaca, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina
| | - Alicia E Consolini
- Cátedra de Farmacología, Grupo de Farmacología Experimental y Energética Cardíaca, Departamento de Ciencias Biológicas, Facultad de Ciencias Exactas, Universidad Nacional de La Plata, Argentina.
| |
Collapse
|
21
|
Yu X, Zhao XD, Bao RQ, Yu JY, Zhang GX, Chen JW. The modified Yi qi decoction protects cardiac ischemia-reperfusion induced injury in rats. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 17:330. [PMID: 28637456 PMCID: PMC5480198 DOI: 10.1186/s12906-017-1829-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/07/2017] [Indexed: 02/06/2023]
Abstract
BACKGROUND To investigate the effects and involved mechanisms of the modified Yi Qi decoction (MYQ) in cardiac ischemia-reperfusion (IR) induced injury. METHODS Male Sprague-Dawley rats were subjected to a 30-min coronary arterial occlusion followed by reperfusion, low or high dose decoction of MYQ was administrated orally for 1 week or 1 month. RESULTS Both in 1 week and 1 month IR rat groups, cardiac function indexes were significantly impaired compared with sham group rats, accompanied with higher ratio of infarct size to risk size, decreased expressions of sodium calcium exchanger (NCX1) and sarcoplasmic reticulum Ca2+-ATPase (Serca2a), and different expressions of autophagic proteins, Beclin-1 and LC3. Treatment with MYQ (low or high dose) for 1 week showed no marked beneficial effects on cardiac function and cardiac injury (ratio of infarct size to risk size), although expressions of anti-apoptotic protein, Bcl-2, NCX1 and Serca2a were increased. Treatment with MYQ (low or high dose) for 1 month showed significantly improved effects on cardiac function and cardiac injury (ratio of infarct size to risk size), accompanied with increase of Bcl-2, NCX1 and Serca2a expressions, and decrease of Bax (a pro-apoptotic protein) and Beclin-1 expressions. CONCLUSIONS The results show that MYQ have potential therapeutic effects on IR-induced cardiac injury, which may be through regulation of apoptotic proteins, cytosolic Ca2+ handling proteins and autophagic proteins signal pathways.
Collapse
Affiliation(s)
- Xiao Yu
- Laboratory of Cancer Molecular Genetics, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou, 215123 People’s Republic of China
| | - Xiao-Dong Zhao
- Department of Internal Medicine, the Affiliated Suzhou Chinese Traditional Medicine Hospital, Nanjing University of Chinese Medicine, 18 Yang-Su Road, Suzhou, 215003 People’s Republic of China
| | - Rong-Qi Bao
- Department of Internal Medicine, the Affiliated Suzhou Chinese Traditional Medicine Hospital, Nanjing University of Chinese Medicine, 18 Yang-Su Road, Suzhou, 215003 People’s Republic of China
| | - Jia-Yu Yu
- Department of Internal Medicine, the Affiliated Suzhou Chinese Traditional Medicine Hospital, Nanjing University of Chinese Medicine, 18 Yang-Su Road, Suzhou, 215003 People’s Republic of China
| | - Guo-Xing Zhang
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou, 215123 People’s Republic of China
| | - Jing-Wei Chen
- Department of Physiology, Medical College of Soochow University, 199 Ren-Ai Road, Dushu Lake Campus, Suzhou Industrial Park, Suzhou, 215123 People’s Republic of China
- Department of Internal Medicine, the Affiliated Suzhou Chinese Traditional Medicine Hospital, Nanjing University of Chinese Medicine, 18 Yang-Su Road, Suzhou, 215003 People’s Republic of China
| |
Collapse
|
22
|
Zhu S, Xu T, Luo Y, Zhang Y, Xuan H, Ma Y, Pan D, Li D, Zhu H. Luteolin Enhances Sarcoplasmic Reticulum Ca2+-ATPase Activity through p38 MAPK Signaling thus Improving Rat Cardiac Function after Ischemia/Reperfusion. Cell Physiol Biochem 2017; 41:999-1010. [DOI: 10.1159/000460837] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2016] [Accepted: 12/01/2016] [Indexed: 01/15/2023] Open
Abstract
Background/Aims: A major challenge for current therapeutic strategies against ischemia/reperfusion (I/R) is the lack of effective drugs. Considering luteolin enhances the activity of sarcoplasmic reticulum Ca2+-ATPase (SERCA2a) to improve the systolic/diastolic function of rat hearts and cardiomyocytes during the I/R process, we studied the regulatory function of the p38 MAPK pathway in this protective mechanism. Methods: Isolated cardiomyocytes and perfused hearts were separately divided into five groups and used to investigate I/R. The phosphorylation of p38 and phospholamban (p-PLB), the levels and activity of SERCA2a and the levels of proteins related to apoptosis were measured. Apoptotic cells were assessed using the TUNEL assay. Single-cell shortening, Ca2+ transients, and the decay of the mitochondrial membrane potential (Δψm) were detected. Results: The p38 MAPK pathway was activated during the I/R process, and inhibiting it with SB203580 promoted p-PLB, which enhanced the activity of SERCA2a and relieved the calcium overload to promote the recovery of the Δψm and reduce cardiomyocyte apoptosis in I/R. Luteolin also suppressed the activation of the p38 MAPK pathway and showed cardioprotective effects during I/R injury. Conclusions: We conclude that luteolin enhances SERCA2a activity to improve systolic/diastolic function during I/R in rat hearts and cardiomyocytes by attenuating the inhibitive effects of the p38 pathway on p-PLB.
Collapse
|
23
|
Le Page S, Niro M, Fauconnier J, Cellier L, Tamareille S, Gharib A, Chevrollier A, Loufrani L, Grenier C, Kamel R, Sarzi E, Lacampagne A, Ovize M, Henrion D, Reynier P, Lenaers G, Mirebeau-Prunier D, Prunier F. Increase in Cardiac Ischemia-Reperfusion Injuries in Opa1+/- Mouse Model. PLoS One 2016; 11:e0164066. [PMID: 27723783 PMCID: PMC5056696 DOI: 10.1371/journal.pone.0164066] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 09/19/2016] [Indexed: 11/21/2022] Open
Abstract
Background Recent data suggests the involvement of mitochondrial dynamics in cardiac ischemia/reperfusion (I/R) injuries. Whilst excessive mitochondrial fission has been described as detrimental, the role of fusion proteins in this context remains uncertain. Objectives To investigate whether Opa1 (protein involved in mitochondrial inner-membrane fusion) deficiency affects I/R injuries. Methods and Results We examined mice exhibiting Opa1delTTAG mutations (Opa1+/-), showing 70% Opa1 protein expression in the myocardium as compared to their wild-type (WT) littermates. Cardiac left-ventricular systolic function assessed by means of echocardiography was observed to be similar in 3-month-old WT and Opa1+/- mice. After subjection to I/R, infarct size was significantly greater in Opa1+/- than in WTs both in vivo (43.2±4.1% vs. 28.4±3.5%, respectively; p<0.01) and ex vivo (71.1±3.2% vs. 59.6±8.5%, respectively; p<0.05). No difference was observed in the expression of other main fission/fusion protein, oxidative phosphorylation, apoptotic markers, or mitochondrial permeability transition pore (mPTP) function. Analysis of calcium transients in isolated ventricular cardiomyocytes demonstrated a lower sarcoplasmic reticulum Ca2+ uptake, whereas cytosolic Ca2+ removal from the Na+/Ca2+ exchanger (NCX) was increased, whilst SERCA2a, phospholamban, and NCX protein expression levels were unaffected in Opa1+/- compared to WT mice. Simultaneous whole-cell patch-clamp recordings of mitochondrial Ca2+ movements and ventricular action potential (AP) showed impairment of dynamic mitochondrial Ca2+ uptake and a marked increase in the AP late repolarization phase in conjunction with greater occurrence of arrhythmia in Opa1+/- mice. Conclusion Opa1 deficiency was associated with increased sensitivity to I/R, imbalance in dynamic mitochondrial Ca2+ uptake, and subsequent increase in NCX activity.
Collapse
Affiliation(s)
- Sophie Le Page
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- Laboratoire Cardioprotection Remodelage et Thrombose, Angers, France
- Service de Cardiologie, CHU Angers, Angers, France
| | - Marjorie Niro
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- Laboratoire Cardioprotection Remodelage et Thrombose, Angers, France
- Service de Cardiologie, CHU Angers, Angers, France
| | | | - Laura Cellier
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- Laboratoire Cardioprotection Remodelage et Thrombose, Angers, France
| | - Sophie Tamareille
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- Laboratoire Cardioprotection Remodelage et Thrombose, Angers, France
| | | | - Arnaud Chevrollier
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- INSERM UMR_S1083, CNRS UMR_C6214, BNMI, Angers, France
| | - Laurent Loufrani
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- INSERM UMR_S1083, CNRS UMR_C6214, BNMI, Angers, France
| | - Céline Grenier
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- INSERM UMR_S1083, CNRS UMR_C6214, BNMI, Angers, France
| | - Rima Kamel
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- Laboratoire Cardioprotection Remodelage et Thrombose, Angers, France
| | - Emmanuelle Sarzi
- Institut des Neurosciences de Montpellier, INSERM U1051, Université Montpellier I et II, Montpellier, France
| | - Alain Lacampagne
- INSERM U1046, Université Montpellier I et II, Montpellier, France
| | | | - Daniel Henrion
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- INSERM UMR_S1083, CNRS UMR_C6214, BNMI, Angers, France
| | - Pascal Reynier
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- INSERM UMR_S1083, CNRS UMR_C6214, BNMI, Angers, France
| | - Guy Lenaers
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- INSERM UMR_S1083, CNRS UMR_C6214, BNMI, Angers, France
| | - Delphine Mirebeau-Prunier
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- INSERM UMR_S1083, CNRS UMR_C6214, BNMI, Angers, France
| | - Fabrice Prunier
- Institut MITOVASC, Université Angers, CHU Angers, Angers, France
- Laboratoire Cardioprotection Remodelage et Thrombose, Angers, France
- Service de Cardiologie, CHU Angers, Angers, France
- * E-mail:
| |
Collapse
|
24
|
Li X, Li W, Gao Z, Li H. Association of cardiac injury with iron-increased oxidative and nitrative modifications of the SERCA2a isoform of sarcoplasmic reticulum Ca2+-ATPase in diabetic rats. Biochimie 2016; 127:144-52. [DOI: 10.1016/j.biochi.2016.05.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 05/17/2016] [Indexed: 12/21/2022]
|
25
|
Detrimental or beneficial: the role of TRPM2 in ischemia/reperfusion injury. Acta Pharmacol Sin 2016; 37:4-12. [PMID: 26725732 DOI: 10.1038/aps.2015.141] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 10/14/2015] [Indexed: 12/30/2022] Open
Abstract
Ischemia/reperfusion (I/R) injury is the main cause of tissue damage and dysfunction. I/R injury is characterized by Ca(2+) overload and production of reactive oxygen species (ROS), which play critical roles in the process of I/R injury to the brain, heart and kidney, but the underlying mechanisms are largely elusive. Recent evidence demonstrates that TRPM2, a Ca(2+)-permeable cationic channel and ROS sensor, is involved in I/R injury, but whether TRPM2 plays a protective or detrimental role in this process remains controversial. In this review, we discuss the recent progress in understanding the role of TRPM2 in reperfusion process after brain, heart and kidney ischemia and the potential of targeting TRPM2 for the development of therapeutic drugs to treat I/R injury.
Collapse
|
26
|
Morin D, Musman J, Pons S, Berdeaux A, Ghaleh B. Mitochondrial translocator protein (TSPO): From physiology to cardioprotection. Biochem Pharmacol 2015; 105:1-13. [PMID: 26688086 DOI: 10.1016/j.bcp.2015.12.003] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/04/2015] [Indexed: 01/08/2023]
Abstract
The mitochondrial translocator protein (TSPO) is a high affinity cholesterol binding protein which is primarily located in the outer mitochondrial membrane where it has been shown to interact with proteins implicated in mitochondrial permeability transition pore (mPTP) formation. TSPO is found in different species and is expressed at high levels in tissues that synthesize steroids but is also present in other peripheral tissues especially in the heart. TSPO has been involved in the import of cholesterol into mitochondria, a key step in steroidogenesis. This constitutes the main established function of the protein which was recently challenged by genetic studies. TSPO has also been associated directly or indirectly with a wide range of cellular functions such as apoptosis, cell proliferation, differentiation, regulation of mitochondrial function or porphyrin transport. In the heart the role of TSPO remains undefined but a growing body of evidence suggests that TSPO plays a critical role in regulating physiological cardiac function and that TSPO ligands may represent interesting drugs to protect the heart under pathological conditions. This article briefly reviews current knowledge regarding TSPO and discusses its role in the cardiovascular system under physiological and pathologic conditions. More particularly, it provides evidence that TSPO can represent an alternative strategy to develop new pharmacological agents to protect the myocardium against ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Didier Morin
- INSERM U955, Équipe 3, Créteil, France; Université Paris-Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France.
| | - Julien Musman
- INSERM U955, Équipe 3, Créteil, France; Université Paris-Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France.
| | - Sandrine Pons
- INSERM U955, Équipe 3, Créteil, France; Université Paris-Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France.
| | - Alain Berdeaux
- INSERM U955, Équipe 3, Créteil, France; Université Paris-Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France.
| | - Bijan Ghaleh
- INSERM U955, Équipe 3, Créteil, France; Université Paris-Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France.
| |
Collapse
|
27
|
MENG YING, LI WEIZHU, SHI YOUWEI, ZHOU BINGFENG, MA RONG, LI WEIPING. Danshensu protects against ischemia/reperfusion injury and inhibits the apoptosis of H9c2 cells by reducing the calcium overload through the p-JNK-NF-κB-TRPC6 pathway. Int J Mol Med 2015; 37:258-66. [DOI: 10.3892/ijmm.2015.2419] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 11/17/2015] [Indexed: 11/06/2022] Open
|
28
|
Altamirano F, Wang ZV, Hill JA. Cardioprotection in ischaemia-reperfusion injury: novel mechanisms and clinical translation. J Physiol 2015; 593:3773-88. [PMID: 26173176 PMCID: PMC4575567 DOI: 10.1113/jp270953] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 06/23/2015] [Indexed: 12/29/2022] Open
Abstract
In recent decades, robust successes have been achieved in conquering the acutely lethal manifestations of heart disease. Nevertheless, the prevalence of heart disease, especially heart failure, continues to rise. Among the precipitating aetiologies, ischaemic disease is a leading cause of heart failure. In the context of ischaemia, the myocardium is deprived of oxygen and nutrients, which elicits a cascade of events that provokes cell death. This ischaemic insult is typically coupled with reperfusion, either spontaneous or therapeutically imposed, wherein blood supply is restored to the previously ischaemic tissue. While this intervention limits ischaemic injury, it triggers a new cascade of events that is also harmful, viz. reperfusion injury. In recent years, novel insights have emerged regarding mechanisms of ischaemia-reperfusion injury, and some hold promise as targets of therapeutic relevance. Here, we review a select number of these pathways, focusing on recent discoveries and highlighting prospects for therapeutic manipulation for clinical benefit.
Collapse
Affiliation(s)
- Francisco Altamirano
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical CenterDallas, TX, 75390, USA
| | - Zhao V Wang
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical CenterDallas, TX, 75390, USA
| | - Joseph A Hill
- Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical CenterDallas, TX, 75390, USA
- Department of Molecular Biology, University of Texas Southwestern Medical CenterDallas, TX, 75390, USA
| |
Collapse
|
29
|
Svennerholm K, Rodsand P, Hellman U, Lundholm M, Waldenström A, Biber B, Ronquist G, Haney M. Myocardial ischemic preconditioning in a porcine model leads to rapid changes in cardiac extracellular vesicle messenger RNA content. IJC HEART & VASCULATURE 2015; 8:62-67. [PMID: 28785681 PMCID: PMC5497283 DOI: 10.1016/j.ijcha.2015.05.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 04/28/2015] [Accepted: 05/05/2015] [Indexed: 11/18/2022]
Affiliation(s)
- Kristina Svennerholm
- Anesthesiology and Intensive Care Medicine, Department of Clinical Science, Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Pouria Rodsand
- Anesthesiology and Intensive Care Medicine, Department of Surgical and Perioperative Science, Umeå University, 90185 Umeå, Sweden
| | - Urban Hellman
- Cardiology, Heart Centre and Department of Public Health and Clinical Medicine, Umeå University, 90185 Umeå, Sweden
| | - Marie Lundholm
- Pathology, Department of Medical Biosciences, Umeå University, 90185 Umeå, Sweden
| | - Anders Waldenström
- Cardiology, Heart Centre and Department of Public Health and Clinical Medicine, Umeå University, 90185 Umeå, Sweden
| | - Björn Biber
- Anesthesiology and Intensive Care Medicine, Department of Clinical Science, Sahlgrenska Academy, University of Gothenburg, 41345 Gothenburg, Sweden
| | - Gunnar Ronquist
- Department of Medical Sciences, Clinical Chemistry, Uppsala University, 75185 Uppsala, Sweden
| | - Michael Haney
- Anesthesiology and Intensive Care Medicine, Department of Surgical and Perioperative Science, Umeå University, 90185 Umeå, Sweden
| |
Collapse
|
30
|
Roussel J, Thireau J, Brenner C, Saint N, Scheuermann V, Lacampagne A, Le Guennec JY, Fauconnier J. Palmitoyl-carnitine increases RyR2 oxidation and sarcoplasmic reticulum Ca2+ leak in cardiomyocytes: Role of adenine nucleotide translocase. Biochim Biophys Acta Mol Basis Dis 2015; 1852:749-58. [DOI: 10.1016/j.bbadis.2015.01.011] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 01/16/2015] [Accepted: 01/18/2015] [Indexed: 12/30/2022]
|
31
|
Montaigne D, Maréchal X, Lacroix D, Staels B. From cardiac mitochondrial dysfunction to clinical arrhythmias. Int J Cardiol 2015; 184:597-599. [PMID: 25769006 DOI: 10.1016/j.ijcard.2015.03.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2015] [Accepted: 03/01/2015] [Indexed: 02/02/2023]
Affiliation(s)
- David Montaigne
- Université Lille 2, F-59000 Lille, France; Inserm, U1011, F-59000 Lille, France; EGID, F-59000 Lille, France; Institut Pasteur de Lille, F-59019 Lille, France; Service d'Explorations Fonctionnelles CardioVasculaires, University Hospital of Lille, France.
| | - Xavier Maréchal
- Université Lille 2, F-59000 Lille, France; Inserm, U1011, F-59000 Lille, France; EGID, F-59000 Lille, France; Institut Pasteur de Lille, F-59019 Lille, France
| | - Dominique Lacroix
- Clinique de Cardiologie, Service de Rythmologie, University Hospital of Lille, France
| | - Bart Staels
- Université Lille 2, F-59000 Lille, France; Inserm, U1011, F-59000 Lille, France; EGID, F-59000 Lille, France; Institut Pasteur de Lille, F-59019 Lille, France
| |
Collapse
|
32
|
Activation of Sonic hedgehog signaling in ventricular cardiomyocytes exerts cardioprotection against ischemia reperfusion injuries. Sci Rep 2015; 5:7983. [PMID: 25613906 PMCID: PMC4303926 DOI: 10.1038/srep07983] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 12/24/2014] [Indexed: 11/09/2022] Open
Abstract
Sonic hedgehog (SHH) is a conserved protein involved in embryonic tissue patterning and development. SHH signaling has been reported as a cardio-protective pathway via muscle repair–associated angiogenesis. The goal of this study was to investigate the role of SHH signaling pathway in the adult myocardium in physiological situation and after ischemia-reperfusion. We show in a rat model of ischemia-reperfusion that stimulation of SHH pathway, either by a recombinant peptide or shed membranes microparticles harboring SHH ligand, prior to reperfusion reduces both infarct size and subsequent arrhythmias by preventing ventricular repolarization abnormalities. We further demonstrate in healthy animals a reduction of QTc interval mediated by NO/cGMP pathway leading to the shortening of ventricular cardiomyocytes action potential duration due to the activation of an inward rectifying potassium current sharing pharmacological and electrophysiological properties with ATP-dependent potassium current. Besides its effect on both angiogenesis and endothelial dysfunction we demonstrate here a novel cardio-protective effect of SHH acting directly on the cardiomyocytes. This emphasizes the pleotropic effect of SHH pathway as a potential cardiac therapeutic target.
Collapse
|
33
|
Roberge S, Roussel J, Andersson DC, Meli AC, Vidal B, Blandel F, Lanner JT, Le Guennec JY, Katz A, Westerblad H, Lacampagne A, Fauconnier J. TNF-α-mediated caspase-8 activation induces ROS production and TRPM2 activation in adult ventricular myocytes. Cardiovasc Res 2014; 103:90-9. [PMID: 24802330 DOI: 10.1093/cvr/cvu112] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
AIMS TRPM2 is a Ca(2+)-permeable cationic channel of the transient receptor potential (TRP) superfamily that is linked to apoptotic signalling. Its involvement in cardiac pathophysiology is unknown. The aim of this study was to determine whether the pro-apoptotic cytokine tumour necrosis factor-α (TNF-α) induces a TRPM2-like current in murine ventricular cardiomyocytes. METHODS AND RESULTS Adult isolated cardiomyocytes from C57BL/6 mice were exposed to TNF-α (10 ng/mL). Western blotting showed TRPM2 expression, which was not changed after TNF-α incubation. Using patch clamp in whole-cell configuration, a non-specific cation current was recorded after exposure to TNF-α (ITNF), which reached maximal steady-state amplitude after 3 h incubation. ITNF was inhibited by the caspase-8 inhibitor z-IETD-fmk, the antioxidant N-acetylcysteine, and the TRPM2 inhibitors clotrimazole, N-(P-amylcinnamoyl) anthranilic acid and flufenamic acid (FFA). TRPM2 has previously been shown to be activated by ADP-ribose, which is produced by poly(ADP-ribose) polymerase 1 (PARP-1). TNF-α exposure resulted in increased poly-ADP-ribosylation of proteins and the PARP-1 inhibitor 3-aminobenzamide inhibited ITNF. TNF-α exposure increased the mitochondrial production of reactive oxygen species (ROS; measured with the fluorescent indicator MitoSOX Red), and this increase was blocked by the caspase-8 inhibitor z-IETD-fmk. Clotrimazole and TRPM2 inhibitory antibody decreased TNF-α-induced cardiomyocyte death. CONCLUSION These results demonstrate that TNF-α induces a TRPM2 current in adult ventricular cardiomyocytes. TNF-α induces caspase-8 activation leading to ROS production, PARP-1 activation, and ADP-ribose production. TNF-induced TRPM2 activation may contribute to cardiomyocyte cell death.
Collapse
Affiliation(s)
- Stéphanie Roberge
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier, France
| | - Julien Roussel
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier, France
| | - Daniel C Andersson
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden Department of Medicine, Karolinska Institutet, Stockholm, Sweden Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Albano C Meli
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier, France
| | - Bastien Vidal
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier, France
| | - Florence Blandel
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier, France
| | - Johanna T Lanner
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jean-Yves Le Guennec
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier, France
| | - Abram Katz
- Department of Physical Therapy, School of Health Sciences, Ariel University, Ariel, Israel
| | - Håkan Westerblad
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Alain Lacampagne
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier, France
| | - Jérémy Fauconnier
- INSERM U1046, Université Montpellier 1, Université Montpellier 2, Montpellier, France
| |
Collapse
|
34
|
Calcium signaling in pancreatic ductal epithelial cells: an old friend and a nasty enemy. Cell Calcium 2014; 55:337-45. [PMID: 24602604 DOI: 10.1016/j.ceca.2014.02.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 02/04/2014] [Accepted: 02/05/2014] [Indexed: 12/20/2022]
Abstract
Ductal epithelial cells of the exocrine pancreas secrete HCO3(-) rich, alkaline pancreatic juice, which maintains the intraluminal pH and washes the digestive enzymes out from the ductal system. Importantly, damage of this secretory process can lead to pancreatic diseases such as acute and chronic pancreatitis. Intracellular Ca(2+) signaling plays a central role in the physiological regulation of HCO3(-) secretion, however uncontrolled Ca(2+) release can lead to intracellular Ca(2+) overload and toxicity, including mitochondrial damage and impaired ATP production. Recent findings suggest that the most common pathogenic factors leading to acute pancreatitis, such as bile acids, or ethanol and ethanol metabolites can evoke different types of intracellular Ca(2+) signals, which can stimulate or inhibit ductal HCO3(-) secretion. Therefore, understanding the intracellular Ca(2+) pathways and the mechanisms which can switch a good signal to a bad signal in pancreatic ductal epithelial cells are crucially important. This review summarizes the variety of Ca(2+) signals both in physiological and pathophysiological aspects and highlight molecular targets which may strengthen our old friend or release our nasty enemy.
Collapse
|
35
|
Roubille F, Lacampagne A. New drug avenues for cardioprotection in patients with acute myocardial infarction. Am J Cardiovasc Drugs 2014; 14:73-7. [PMID: 24105018 DOI: 10.1007/s40256-013-0049-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- François Roubille
- Montreal Heart Institute, Université de Montréal, Montreal, QC, Canada,
| | | |
Collapse
|