1
|
Xu Z, Song R, Chen Z, Sun Y, Xia Y, Miao H, Wang W, Zhang Y, Jiang X, Chen G. Hydrogen generators-protected mesenchymal stem cells reverse articular redox imbalance-induced immune dysfunction for osteoarthritis treatment. Biomaterials 2025; 320:123239. [PMID: 40054376 DOI: 10.1016/j.biomaterials.2025.123239] [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: 10/26/2024] [Revised: 02/07/2025] [Accepted: 03/03/2025] [Indexed: 04/06/2025]
Abstract
Stem cell therapy has revolutionized the management of osteoarthritis (OA), but the articular dysregulated redox status diminishes cell engraftment efficiency and disrupts immune homeostasis, therefore compromising the overall therapeutic efficacy. Here, we present hydrogen (H2) generators-backpacked mesenchymal stem cells (MSCs) which preserve the biological functions and survival of transplanted cells and reverse articular immune dysfunction, mitigating OA. Specifically, post systemic transplantation, H2 generators-laden MSCs home to OA joints, and upon stimulation in acidic OA environment, H2 produced from the generators remodels articular redox balance, thereby relieving the loss of mitochondrial membrane potential, decreasing cell apoptosis rate, and maintaining pluripotent and paracrine functions of MSCs. Furthermore, the reactive oxygen species scavenging by H2 in combination with paracrine effects of the MSCs promote macrophage polarization towards the anti-inflammatory M2 phenotype, which contributes to reversing synovial immune disorder. In severe OA model, the backpacked MSCs reduce osteoarthritic degeneration, osteophyte formation and joint inflammation, and promote cartilage regeneration. In sum, our work demonstrates that arming with H2 generators effectively boosts the therapeutic efficacy of MSCs, which hold great potential for alleviating redox imbalance-related tissue lesions, including but not limited to OA.
Collapse
Affiliation(s)
- Zhou Xu
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, China; Shandong Provincial Key Medical and Health Laboratory of Neuro-oncology of Innovative Integrated Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Qingdao, 266024, China; Northern Jiangsu People's Hospital, Yangzhou, 225001, China; Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Ruilong Song
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Zhiling Chen
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, China
| | - Yu Sun
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Yinhe Xia
- Institute of Comparative Medicine, College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China
| | - Haixiang Miao
- Department of Orthopedics, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou, 225001, China
| | - Weijie Wang
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, China
| | - Yuankai Zhang
- Department of Orthopaedic Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Xinyi Jiang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products and Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Gang Chen
- Qingdao Key Laboratory of Materials for Tissue Repair and Rehabilitation, School of Rehabilitation Sciences and Engineering, University of Health and Rehabilitation Sciences, Qingdao, 266024, China; Shandong Provincial Key Medical and Health Laboratory of Neuro-oncology of Innovative Integrated Medicine, Qingdao Central Hospital, University of Health and Rehabilitation Sciences (Qingdao Central Medical Group), Qingdao, Qingdao, 266024, China; State Key Laboratory on Technologies for Chinese Medicine Pharmaceutical Process Control and Intelligent Manufacture, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Baranovicova E, Kalenska D, Lehotsky J. Glutamate/GABA/glutamine ratios in intact and ischemia reperfusion challenged rat brain subregions, the effect of ischemic preconditioning. Metab Brain Dis 2025; 40:121. [PMID: 39918622 PMCID: PMC11805880 DOI: 10.1007/s11011-024-01511-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 12/16/2024] [Indexed: 02/11/2025]
Abstract
The proper function of the brain is entirely dependent on intact neurotransmission, where glutamate (Glu) and γ-aminobutyric acid (GABA) are the two most present neurotransmitters. Maintenance of these neurotransmitters pools is strictly relying on the de novo synthesis of glutamine in astrocytes. Cerebral ischemic events disrupt the balance in uptake and re-synthesis, altering Glu, GABA, and glutamine (Gln) levels. We focused on the determining of the ratios of glutamate, GABA and glutamine in the brain of rats in the intact state, the early changes and temporal development of changes towards the recovery after disruption of balance by global cerebral ischemia. Animals underwent 15 min of global cerebral ischemia, and changes in Glu/GABA/Gln ratios in the hippocampus, cortex, and cerebellum were assessed at 3 h, 24 h, and 72 h post-reperfusion using high-resolution NMR. Ischemic preconditioning was also used to induce tolerance. In an intact rat brain, glutamate level was about twice that of glutamine in all substructures, about sevenfold compared to GABA in the hippocampus and cortex, and almost eightfold compared to GABA in the cerebellum. There were three to four times as much glutamine compared to GABA. After severe cerebral ischemia, Glu/Gln as well as GABA/Gln ratios extensively dropped in early reperfusion (3 h) and gradually increased in 72 h reperfusion time, however, only the Glu/Gln ratio recovered to the level of controls. Glu/GABA ratio remained in all three reperfusion times over the level of control animals. We observed a decrease in glutathione NMR peak in brain tissue homogenates after ischemia. The obtained data suggest the accelerated accumulation of intraparenchymal glutamate after ischemia, which was even more pronounced in the preconditioned animals three days after an ischemic event. The postischemic GABA level restoration did not achieve the level before ischemia in 72 h reperfusion, which could be one of the limiting factors in the complete postischemic GABA transmission recovery. Presented data may be of advantage not only when comparing glutamate and GABA homeostasis and neurotransmission, but also for glutamine reserve display as neurotransmitter precursor and ammonia transfer buffer in glutamate/GABA/glutamine cycle within the intact brain substructures as well after ischemic insult in rats.
Collapse
Affiliation(s)
- Eva Baranovicova
- Biomedical Centre Martin, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine in Martin, Comenius University Bratislava, Mala Hora 4, 036 01, Martin, Slovakia.
| |
Collapse
|
3
|
Khaliulin I, Hamoudi W, Amal H. The multifaceted role of mitochondria in autism spectrum disorder. Mol Psychiatry 2025; 30:629-650. [PMID: 39223276 PMCID: PMC11753362 DOI: 10.1038/s41380-024-02725-z] [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: 03/10/2024] [Revised: 08/21/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
Normal brain functioning relies on high aerobic energy production provided by mitochondria. Failure to supply a sufficient amount of energy, seen in different brain disorders, including autism spectrum disorder (ASD), may have a significant negative impact on brain development and support of different brain functions. Mitochondrial dysfunction, manifested in the abnormal activities of the electron transport chain and impaired energy metabolism, greatly contributes to ASD. The aberrant functioning of this organelle is of such high importance that ASD has been proposed as a mitochondrial disease. It should be noted that aerobic energy production is not the only function of the mitochondria. In particular, these organelles are involved in the regulation of Ca2+ homeostasis, different mechanisms of programmed cell death, autophagy, and reactive oxygen and nitrogen species (ROS and RNS) production. Several syndromes originated from mitochondria-related mutations display ASD phenotype. Abnormalities in Ca2+ handling and ATP production in the brain mitochondria affect synaptic transmission, plasticity, and synaptic development, contributing to ASD. ROS and Ca2+ regulate the activity of the mitochondrial permeability transition pore (mPTP). The prolonged opening of this pore affects the redox state of the mitochondria, impairs oxidative phosphorylation, and activates apoptosis, ultimately leading to cell death. A dysregulation between the enhanced mitochondria-related processes of apoptosis and the inhibited autophagy leads to the accumulation of toxic products in the brains of individuals with ASD. Although many mitochondria-related mechanisms still have to be investigated, and whether they are the cause or consequence of this disorder is still unknown, the accumulating data show that the breakdown of any of the mitochondrial functions may contribute to abnormal brain development leading to ASD. In this review, we discuss the multifaceted role of mitochondria in ASD from the various aspects of neuroscience.
Collapse
Affiliation(s)
- Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Wajeha Hamoudi
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem, Israel.
| |
Collapse
|
4
|
Lu A, Xu Z, Zhao Z, Yan Y, Jiang L, Geng J, Jin H, Wang X, Liu X, Zhu Y, Shi Y, Liu L, Dai H, Wang JC. Double Braking Effects of Nanomedicine on Mitochondrial Permeability Transition Pore for Treating Idiopathic Pulmonary Fibrosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2405406. [PMID: 39475000 DOI: 10.1002/advs.202405406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 10/08/2024] [Indexed: 12/19/2024]
Abstract
Mitochondrial permeability transition pore (mPTP) opening is a key hallmark of injured type II alveolar epithelial cells (AECIIs) in idiopathic pulmonary fibrosis (IPF). Inhibiting mPTP opening in AECIIs is considered a potential IPF treatment. Herein, a "double braking" strategy on mPTP by cyclosporin A (CsA) derived ionizable lipid with 3D structure (3D-lipid) binding cyclophilin D (CypD) and siRNA downregulating mitochondrial calcium uniporter (MCU) expression is proposed for treating IPF. 3D-lipid and MCU targeting siRNA (siMCU) are co-assembled to form stable 3D-LNP/siMCU nanoparticles (NPs), along with helper lipids. In vitro results demonstrated that these NPs effectively inhibit mPTP opening by 3D-lipid binding with CypD and siRNA downregulating MCU expression, thereby decreasing damage-associated molecular patterns (DAMPs) release and suppressing epithelial-to-mesenchymal transition (EMT) process in bleomycin-induced A549 cells. In vivo results revealed that 3D-LNP/siMCU NPs effectively ameliorated collagen deposition, pro-fibrotic factors secretion, and fibroblast activation in bleomycin-induced pulmonary fibrosis (PF) mouse models. Moreover, compared to the commercial MC3-based formulation, optimized Opt-MC3/siRNA NPs with incorporating 3D-lipid as the fifth component, showed superior therapeutic efficacy against PF due to their enhanced stability and higher gene silencing efficiency. Overall, the nanomedicine containing 3D-lipid and siMCU will be a promising and potential approach for IPF treatment.
Collapse
Affiliation(s)
- An Lu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zhiyi Xu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zhixia Zhao
- Department of Pharmacy, Clinical Trial Research Center, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Yi Yan
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Linxia Jiang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Jing Geng
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Hongwei Jin
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiangyu Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Xiaoyan Liu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yuanjun Zhu
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yujie Shi
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Lihong Liu
- Department of Pharmacy, Clinical Trial Research Center, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Huaping Dai
- National Center for Respiratory Medicine, State Key Laboratory of Respiratory Health and Multimorbidity, National Clinical Research Center for Respiratory Diseases, Institute of Respiratory Medicine, Chinese Academy of Medical Sciences, Peking Union Medical College, Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, 100029, China
| | - Jian-Cheng Wang
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
- Laboratory of Innovative Formulations and Pharmaceutical Excipients, Peking University Ningbo Institute of Marine Medicine, Ningbo, 315832, China
| |
Collapse
|
5
|
Wang Y, Du B, Han X, Qu L. Molecular mechanism underlying the protective effects of ischemic preconditioning in total knee arthroplasty. Chin J Traumatol 2024:S1008-1275(24)00153-6. [PMID: 39551662 DOI: 10.1016/j.cjtee.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 02/08/2024] [Accepted: 02/10/2024] [Indexed: 11/19/2024] Open
Abstract
PROPOSE To investigate the molecular mechanisms underlying the protective effects of ischemic preconditioning (IPC) in patients undergoing total knee arthroplasty. METHODS GSE21164 was extracted from an online database, followed by an investigation of differentially expressed genes (DEGs) between IPC treatment samples at 2 time points (T0T and T1T). Function and pathway enrichment analyses were performed on the DEGs. A protein-protein interaction network was constructed to identify hub genes according to 5 different algorithms, followed by enrichment analysis. In addition, long noncoding RNAs (lncRNAs) were identified between the T0T and T1T samples. Furthermore, a competing endogenous RNA network was predicted based on the identified lncRNA-messenger RNA (mRNA), lncRNA-microRNA (miRNA), and mRNA-miRNA relationships revealed in this study. Finally, a drug-gene network was investigated. Statistical analyses were performed using GraphPad Prism 8.0. Differences between groups were determined using an unpaired t-test. p < 0.05 was considered significant. RESULTS A total of 343 DEGs at T0 and 10 DEGs at T1 were identified and compared with their respective control groups, followed by 100 DEGs between T0T and T1T. Based on these 100 DEGs, protein-protein interaction network analysis revealed 9 hub genes, mainly with mitochondria-related functions and the carbon metabolism pathway. Six differentially expressed lncRNAs were investigated between T0T and T1T. A competing endogenous RNA network was constructed using 259 lncRNA-miRNA-mRNA interactions, including alpha-2-macroglobulin antisense RNA 1-miR-7161-5p-iron-sulfur cluster scaffold. Finally, 13 chemical drugs associated with the hub genes were explored. CONCLUSION Iron-sulfur cluster scaffold may promote IPC-induced ischemic tolerance mediated by alpha-2-macroglobulin antisense RNA 1-miR-7161-5p axis. Moreover, IPC may induce a protective response after total knee arthroplasty via mitochondria-related functions and the carbon metabolism pathway, which should be further validated in the near future.
Collapse
Affiliation(s)
- Yongli Wang
- Department of Anesthesiology, the 80th Group Military Hospital of the Chinese People's Liberation Army, Weifang, 261000, Shandong province, China
| | - Bencai Du
- Orthopedic Center, Sunshine Union Hospital, Weifang, 261000, Shandong province, China
| | - Xueliang Han
- Orthopedic Center, Sunshine Union Hospital, Weifang, 261000, Shandong province, China
| | - Lianjun Qu
- Orthopedic Center, Sunshine Union Hospital, Weifang, 261000, Shandong province, China.
| |
Collapse
|
6
|
Kim HJ, Norton CE, Zawieja SD, Castorena-Gonzalez JA, Davis MJ. Acute Metabolic Stress Induces Lymphatic Dysfunction Through KATP Channel Activation. FUNCTION 2024; 5:zqae033. [PMID: 39075985 PMCID: PMC11384908 DOI: 10.1093/function/zqae033] [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: 03/31/2024] [Revised: 06/26/2024] [Accepted: 07/13/2024] [Indexed: 07/31/2024] Open
Abstract
Lymphatic dysfunction is an underlying component of multiple metabolic diseases, including diabetes, obesity, and metabolic syndrome. We investigated the roles of KATP channels in lymphatic contractile dysfunction in response to acute metabolic stress induced by inhibition of the mitochondrial electron transport chain. Ex vivo popliteal lymphatic vessels from mice were exposed to the electron transport chain inhibitors antimycin A and rotenone, or the oxidative phosphorylation inhibitor/protonophore, CCCP. Each inhibitor led to a significant reduction in the frequency of spontaneous lymphatic contractions and calculated pump flow, without a significant change in contraction amplitude. Contraction frequency was restored by the KATP channel inhibitor, glibenclamide. Lymphatic vessels from mice with global Kir6.1 deficiency or expressing a smooth muscle-specific dominant negative Kir6.1 channel were resistant to inhibition. Antimycin A inhibited the spontaneous action potentials generated in lymphatic muscle and this effect was reversed by glibenclamide, confirming the role of KATP channels. Antimycin A, but not rotenone or CCCP, increased dihydrorhodamine fluorescence in lymphatic muscle, indicating ROS production. Pretreatment with tiron or catalase prevented the effect of antimycin A on wild-type lymphatic vessels, consistent with its action being mediated by ROS. Our results support the conclusion that KATP channels in lymphatic muscle can be directly activated by reduced mitochondrial ATP production or ROS generation, consequent to acute metabolic stress, leading to contractile dysfunction through inhibition of the ionic pacemaker controlling spontaneous lymphatic contractions. We propose that a similar activation of KATP channels contributes to lymphatic dysfunction in metabolic disease.
Collapse
Affiliation(s)
- Hae Jin Kim
- Department of Medical Pharmacology & Physiology, University of Missouri, Columbia, MO 65212, USA
| | - Charles E Norton
- Department of Medical Pharmacology & Physiology, University of Missouri, Columbia, MO 65212, USA
| | - Scott D Zawieja
- Department of Medical Pharmacology & Physiology, University of Missouri, Columbia, MO 65212, USA
| | | | - Michael J Davis
- Department of Medical Pharmacology & Physiology, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
7
|
de Souza IIA, da Silva Barenco T, Pavarino MEMF, Couto MT, de Resende GO, de Oliveira DF, Ponte CG, Nascimento JHM, Maciel L. A potent and selective activator of large-conductance Ca 2+-activated K + channels induces preservation of mitochondrial function after hypoxia and reoxygenation by handling of calcium and transmembrane potential. Acta Physiol (Oxf) 2024; 240:e14151. [PMID: 38676357 DOI: 10.1111/apha.14151] [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: 10/31/2023] [Revised: 02/15/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024]
Abstract
AIMS Ischaemic heart disease remains a significant cause of mortality globally. A pharmacological agent that protects cardiac mitochondria against oxygen deprivation injuries is welcome in therapy against acute myocardial infarction. Here, we evaluate the effect of large-conductance Ca2+-activated K+ channels (BKCa) activator, Compound Z, in isolated mitochondria under hypoxia and reoxygenation. METHODS Mitochondria from mice hearts were obtained by differential centrifugation. The isolated mitochondria were incubated with a BKCa channel activator, Compound Z, and subjected to normoxia or hypoxia/reoxygenation. Mitochondrial function was evaluated by measurement of O2 consumption in the complexes I, II, and IV in the respiratory states 1, 2, 3, and by maximal uncoupled O2 uptake, ATP production, ROS production, transmembrane potential, and calcium retention capacity. RESULTS Incubation of isolated mitochondria with Compound Z under normoxia conditions reduced the mitochondrial functions and induced the production of a significant amount of ROS. However, under hypoxia/reoxygenation, the Compound Z prevented a profound reduction in mitochondrial functions, including reducing ROS production over the hypoxia/reoxygenation group. Furthermore, hypoxia/reoxygenation induced a large mitochondria depolarization, which Compound Z incubation prevented, but, even so, Compound Z created a small depolarization. The mitochondrial calcium uptake was prevented by the BKCa activator, extruding the mitochondrial calcium present before Compound Z incubation. CONCLUSION The Compound Z acts as a mitochondrial BKCa channel activator and can protect mitochondria function against hypoxia/reoxygenation injury, by handling mitochondrial calcium and transmembrane potential.
Collapse
Affiliation(s)
- Itanna Isis Araujo de Souza
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Programa de Pós-Graduação Em Cardiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Thais da Silva Barenco
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Programa de Pós-Graduação Em Cardiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | | | - Marcos Tadeu Couto
- Instituto Federal de Educação, Ciência e Tecnologia do Rio de Janeiro, Rio de Janeiro, Brasil
| | | | | | | | - José Hamilton Matheus Nascimento
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Programa de Pós-Graduação Em Cardiologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
| | - Leonardo Maciel
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brasil
- Universidade Federal do Rio de Janeiro, Duque de Caxias, Brasil
| |
Collapse
|
8
|
French C, Robbins D, Gernigon M, Gordon D. The effects of lower limb ischaemic preconditioning: a systematic review. Front Physiol 2024; 14:1323310. [PMID: 38274048 PMCID: PMC10808809 DOI: 10.3389/fphys.2023.1323310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 12/28/2023] [Indexed: 01/27/2024] Open
Abstract
Ischaemic preconditioning (IPC) involves the use of repeated occlusions and reperfusions of the peripheral muscle blood supply at a limb. This systematic literature review examines the typical responses in response to the method of application during an IPC applied at the lower limb. This review focuses on the physiological responses for VO2max, haemoglobin, metabolic and genetic responses to various IPC interventions. The literature search was performed using four databases and assessed using the PRISMA search strategy and COSMIN to assess the quality of the articles. Seventeen articles were included in the review, with a total of 237 participants. While there is variation in the method of application, the average occlusion pressure was 222 ± 34 mmHg, ranging from 170 to 300 mmHg typically for 3 or 4 occlusion cycles. The distribution of this pressure is influenced by cuff width, although 8 studies failed to report cuff width. The majority of studies applies IPC at the proximal thigh with 16/17 studies applying an occlusion below this location. The results highlighted the disparities and conflicting findings in response to various IPC methods. While there is some agreement in certain aspects of the IPC manoeuvre such as the location of the occlusion during lower limb IPC, there is a lack of consensus in the optimal protocol to elicit the desired responses. This offers the opportunity for future research to refine the protocols, associated responses, and mechanisms responsible for these changes during the application of IPC.
Collapse
Affiliation(s)
- Chloe French
- Cambridge Centre for Sport and Exercise Sciences (CCSES), Faculty of Science and Engineering, Anglia Ruskin University, Cambridge, United Kingdom
- CIAMS, Université Paris-Saclay, Orsay Cedex, France
- CIAMS, Université d’Orléans, Orléans, France
| | - Dan Robbins
- Medical Technology Research Centre, Faculty of Health, Education, Medicine and Social Care, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Marie Gernigon
- CIAMS, Université Paris-Saclay, Orsay Cedex, France
- CIAMS, Université d’Orléans, Orléans, France
| | - Dan Gordon
- Cambridge Centre for Sport and Exercise Sciences (CCSES), Faculty of Science and Engineering, Anglia Ruskin University, Cambridge, United Kingdom
| |
Collapse
|
9
|
Seefeldt JM, Libai Y, Berg K, Jespersen NR, Lassen TR, Dalsgaard FF, Ryhammer P, Pedersen M, Ilkjaer LB, Hu MA, Erasmus ME, Nielsen RR, Bøtker HE, Caspi O, Eiskjær H, Moeslund N. Effects of ketone body 3-hydroxybutyrate on cardiac and mitochondrial function during donation after circulatory death heart transplantation. Sci Rep 2024; 14:757. [PMID: 38191915 PMCID: PMC10774377 DOI: 10.1038/s41598-024-51387-y] [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: 10/18/2023] [Accepted: 01/04/2024] [Indexed: 01/10/2024] Open
Abstract
Normothermic regional perfusion (NRP) allows assessment of therapeutic interventions prior to donation after circulatory death transplantation. Sodium-3-hydroxybutyrate (3-OHB) increases cardiac output in heart failure patients and diminishes ischemia-reperfusion injury, presumably by improving mitochondrial metabolism. We investigated effects of 3-OHB on cardiac and mitochondrial function in transplanted hearts and in cardiac organoids. Donor pigs (n = 14) underwent circulatory death followed by NRP. Following static cold storage, hearts were transplanted into recipient pigs. 3-OHB or Ringer's acetate infusions were initiated during NRP and after transplantation. We evaluated hemodynamics and mitochondrial function. 3-OHB mediated effects on contractility, relaxation, calcium, and conduction were tested in cardiac organoids from human pluripotent stem cells. Following NRP, 3-OHB increased cardiac output (P < 0.0001) by increasing stroke volume (P = 0.006), dP/dt (P = 0.02) and reducing arterial elastance (P = 0.02). Following transplantation, infusion of 3-OHB maintained mitochondrial respiration (P = 0.009) but caused inotropy-resistant vasoplegia that prevented weaning. In cardiac organoids, 3-OHB increased contraction amplitude (P = 0.002) and shortened contraction duration (P = 0.013) without affecting calcium handling or conduction velocity. 3-OHB had beneficial cardiac effects and may have a potential to secure cardiac function during heart transplantation. Further studies are needed to optimize administration practice in donors and recipients and to validate the effect on mitochondrial function.
Collapse
Affiliation(s)
- Jacob Marthinsen Seefeldt
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark.
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark.
| | - Yaara Libai
- The Laboratory for Cardiovascular Precision Medicine, Rapport Faculty of Medicine, Technion and Rambam's Cardiovascular Research and Innovation Center, 2 Efron St, Haifa, Israel
| | - Katrine Berg
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark
| | - Nichlas Riise Jespersen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Thomas Ravn Lassen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Frederik Flyvholm Dalsgaard
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark
| | - Pia Ryhammer
- Department of Anesthesiology, Regional Hospital Silkeborg, Falkevej 1A, 8600, Silkeborg, Denmark
| | - Michael Pedersen
- Comparative Medicine Lab, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark
| | - Lars Bo Ilkjaer
- Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Palle Juul-Jensens, Boulevard 99, 8200, Aarhus N, Denmark
| | - Michiel A Hu
- Department of Cardiothoracic Surgery, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Michiel E Erasmus
- Department of Cardiothoracic Surgery, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - Roni R Nielsen
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Hans Erik Bøtker
- Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 82, 8200, Aarhus N, Denmark
| | - Oren Caspi
- The Laboratory for Cardiovascular Precision Medicine, Rapport Faculty of Medicine, Technion and Rambam's Cardiovascular Research and Innovation Center, 2 Efron St, Haifa, Israel
| | - Hans Eiskjær
- Department of Cardiology, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, 8200, Aarhus N, Denmark
| | - Niels Moeslund
- Department of Cardiothoracic and Vascular Surgery, Aarhus University Hospital, Palle Juul-Jensens, Boulevard 99, 8200, Aarhus N, Denmark
| |
Collapse
|
10
|
Tallo FS, de Santana PO, Pinto SAG, Lima RY, de Araújo EA, Tavares JGP, Pires-Oliveira M, Nicolau LAD, Medeiros JVR, Taha MO, David AI, Luna-Filho B, Filho CEB, Barbosa AHP, Silva CMC, Wanderley AG, Caixeta A, Caricati-Neto A, Menezes-Rodrigues FS. Pharmacological Modulation of the Ca 2+/cAMP/Adenosine Signaling in Cardiac Cells as a New Cardioprotective Strategy to Reduce Severe Arrhythmias in Myocardial Infarction. Pharmaceuticals (Basel) 2023; 16:1473. [PMID: 37895945 PMCID: PMC10610028 DOI: 10.3390/ph16101473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Revised: 10/09/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
Acute myocardial infarction (AMI) is the main cause of morbidity and mortality worldwide and is characterized by severe and fatal arrhythmias induced by cardiac ischemia/reperfusion (CIR). However, the molecular mechanisms involved in these arrhythmias are still little understood. To investigate the cardioprotective role of the cardiac Ca2+/cAMP/adenosine signaling pathway in AMI, L-type Ca2+ channels (LTCC) were blocked with either nifedipine (NIF) or verapamil (VER), with or without A1-adenosine (ADO), receptors (A1R), antagonist (DPCPX), or cAMP efflux blocker probenecid (PROB), and the incidence of ventricular arrhythmias (VA), atrioventricular block (AVB), and lethality (LET) induced by CIR in rats was evaluated. VA, AVB and LET incidences were evaluated by ECG analysis and compared between control (CIR group) and intravenously treated 5 min before CIR with NIF 1, 10, and 30 mg/kg and VER 1 mg/kg in the presence or absence of PROB 100 mg/kg or DPCPX 100 µg/kg. The serum levels of cardiac injury biomarkers total creatine kinase (CK) and CK-MB were quantified. Both NIF and VER treatment were able to attenuate cardiac arrhythmias caused by CIR; however, these antiarrhythmic effects were abolished by pretreatment with PROB and DPCPX. The total serum CK and CK-MB were similar in all groups. These results indicate that the pharmacological modulation of Ca2+/cAMP/ADO in cardiac cells by means of attenuation of Ca2+ influx via LTCC and the activation of A1R by endogenous ADO could be a promising therapeutic strategy to reduce the incidence of severe and fatal arrhythmias caused by AMI in humans.
Collapse
Affiliation(s)
- Fernando Sabia Tallo
- Department of Urgency and Emergency Care, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil
| | - Patricia Oliveira de Santana
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil; (P.O.d.S.); (S.A.G.P.); (R.Y.L.); (E.A.d.A.); (B.L.-F.); (C.E.B.F.); (A.H.P.B.); (C.M.C.S.); (A.C.)
| | - Sandra Augusta Gordinho Pinto
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil; (P.O.d.S.); (S.A.G.P.); (R.Y.L.); (E.A.d.A.); (B.L.-F.); (C.E.B.F.); (A.H.P.B.); (C.M.C.S.); (A.C.)
| | - Rildo Yamaguti Lima
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil; (P.O.d.S.); (S.A.G.P.); (R.Y.L.); (E.A.d.A.); (B.L.-F.); (C.E.B.F.); (A.H.P.B.); (C.M.C.S.); (A.C.)
| | - Erisvaldo Amarante de Araújo
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil; (P.O.d.S.); (S.A.G.P.); (R.Y.L.); (E.A.d.A.); (B.L.-F.); (C.E.B.F.); (A.H.P.B.); (C.M.C.S.); (A.C.)
| | - José Gustavo Padrão Tavares
- Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil; (J.G.P.T.); (A.C.-N.)
| | - Marcelo Pires-Oliveira
- União Metropolitana de Educação e Cultura—School of Medicine (UNIME), Lauro de Freitas 42700-000, BA, Brazil;
| | - Lucas Antonio Duarte Nicolau
- Department of Biotechnology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba 64202-020, PI, Brazil; (L.A.D.N.); (J.V.R.M.)
| | - Jand Venes Rolim Medeiros
- Department of Biotechnology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba 64202-020, PI, Brazil; (L.A.D.N.); (J.V.R.M.)
| | - Murched Omar Taha
- Department of Surgery, Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-900, SP, Brazil; (M.O.T.); (A.I.D.)
| | - André Ibrahim David
- Department of Surgery, Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-900, SP, Brazil; (M.O.T.); (A.I.D.)
| | - Bráulio Luna-Filho
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil; (P.O.d.S.); (S.A.G.P.); (R.Y.L.); (E.A.d.A.); (B.L.-F.); (C.E.B.F.); (A.H.P.B.); (C.M.C.S.); (A.C.)
| | - Carlos Eduardo Braga Filho
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil; (P.O.d.S.); (S.A.G.P.); (R.Y.L.); (E.A.d.A.); (B.L.-F.); (C.E.B.F.); (A.H.P.B.); (C.M.C.S.); (A.C.)
| | - Adriano Henrique Pereira Barbosa
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil; (P.O.d.S.); (S.A.G.P.); (R.Y.L.); (E.A.d.A.); (B.L.-F.); (C.E.B.F.); (A.H.P.B.); (C.M.C.S.); (A.C.)
| | - Célia Maria Camelo Silva
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil; (P.O.d.S.); (S.A.G.P.); (R.Y.L.); (E.A.d.A.); (B.L.-F.); (C.E.B.F.); (A.H.P.B.); (C.M.C.S.); (A.C.)
| | - Almir Gonçalves Wanderley
- Department of Pharmaceutical Sciences, Universidade Federal de São Paulo (UNIFESP), Diadema 09913-030, SP, Brazil;
| | - Adriano Caixeta
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil; (P.O.d.S.); (S.A.G.P.); (R.Y.L.); (E.A.d.A.); (B.L.-F.); (C.E.B.F.); (A.H.P.B.); (C.M.C.S.); (A.C.)
| | - Afonso Caricati-Neto
- Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil; (J.G.P.T.); (A.C.-N.)
| | - Francisco Sandro Menezes-Rodrigues
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil; (P.O.d.S.); (S.A.G.P.); (R.Y.L.); (E.A.d.A.); (B.L.-F.); (C.E.B.F.); (A.H.P.B.); (C.M.C.S.); (A.C.)
- Department of Surgery, Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-900, SP, Brazil; (M.O.T.); (A.I.D.)
| |
Collapse
|
11
|
Filho CEB, Barbosa AHP, Nicolau LAD, Medeiros JVR, Pires-Oliveira M, dos Santos Póvoa RM, Govato TCP, Júnior HJF, de Carvalho RG, Luna-Filho B, Sabia Tallo F, de Araújo EA, Padrão Tavares JG, Arida RM, Caricati-Neto A, Menezes-Rodrigues FS. Pharmacological Modulation by Low Molecular Weight Heparin of Purinergic Signaling in Cardiac Cells Prevents Arrhythmia and Lethality Induced by Myocardial Infarction. J Cardiovasc Dev Dis 2023; 10:jcdd10030103. [PMID: 36975867 PMCID: PMC10058697 DOI: 10.3390/jcdd10030103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Revised: 02/22/2023] [Accepted: 02/24/2023] [Indexed: 03/02/2023] Open
Abstract
Background: Although several studies suggest that heparins prevent arrhythmias caused by acute myocardial infarction (AMI), the molecular mechanisms involved remain unclear. To investigate the involvement of pharmacological modulation of adenosine (ADO) signaling in cardiac cells by a low-molecular weight heparin (enoxaparin; ENOX) used in AMI therapy, the effects of ENOX on the incidences of ventricular arrhythmias (VA), atrioventricular block (AVB), and lethality (LET) induced by cardiac ischemia and reperfusion (CIR) were evaluated, with or without ADO signaling blockers. Methods: To induce CIR, adult male Wistar rats were anesthetized and subjected to CIR. Electrocardiogram (ECG) analysis was used to evaluate CIR-induced VA, AVB, and LET incidence, after treatment with ENOX. ENOX effects were evaluated in the absence or presence of an ADO A1-receptor antagonist (DPCPX) and/or an inhibitor of ABC transporter-mediated cAMP efflux (probenecid, PROB). Results: VA incidence was similar between ENOX-treated (66%) and control rats (83%), but AVB (from 83% to 33%) and LET (from 75% to 25%) incidences were significantly lower in rats treated with ENOX. These cardioprotective effects were blocked by either PROB or DPCPX. Conclusion: These results indicate that ENOX was effective in preventing severe and lethal arrhythmias induced by CIR due to pharmacological modulation of ADO signaling in cardiac cells, suggesting that this cardioprotective strategy could be promising in AMI therapy.
Collapse
Affiliation(s)
- Carlos Eduardo Braga Filho
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil
| | | | | | - Jand Venes Rolim Medeiros
- Department of Biotechnology, Universidade Federal do Delta do Parnaíba (UFDPar), Parnaíba 64202-020, PI, Brazil
| | - Marcelo Pires-Oliveira
- União Metropolitana de Educação e Cultura–School of Medicine (UNIME), Lauro de Freitas 42700-000, BA, Brazil
| | - Rui Manuel dos Santos Póvoa
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil
| | | | - Hézio Jadir Fernandes Júnior
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil
| | - Rafael Guzella de Carvalho
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil
| | - Bráulio Luna-Filho
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil
| | - Fernando Sabia Tallo
- Department of Urgency and Emergency Care, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil
| | - Erisvaldo Amarante de Araújo
- Postgraduate Program in Cardiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04024-000, SP, Brazil
| | | | - Ricardo Mario Arida
- Department of Physiology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil
| | - Afonso Caricati-Neto
- Department of Pharmacology, Universidade Federal de São Paulo (UNIFESP), São Paulo 04023-062, SP, Brazil
| | | |
Collapse
|
12
|
Methane Admixture Protects Liver Mitochondria and Improves Graft Function after Static Cold Storage and Reperfusion. Antioxidants (Basel) 2023; 12:antiox12020271. [PMID: 36829829 PMCID: PMC9951982 DOI: 10.3390/antiox12020271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/21/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023] Open
Abstract
Mitochondria are targets of cold ischemia-reperfusion (IR), the major cause of cell damage during static cold preservation of liver allografts. The bioactivity of methane (CH4) has recently been recognized in various hypoxic and IR conditions as having influence on many aspects of mitochondrial biology. We therefore hypothesized that cold storage of liver grafts in CH4-enriched preservation solution can provide an increased defence against organ dysfunction in a preclinical rat model of liver transplantation. Livers were preserved for 24 h in cold histidine-tryptophan-ketoglutarate (HTK) or CH4-enriched HTK solution (HTK-CH4) (n = 24 each); then, viability parameters were monitored for 60 min during normothermic isolated reperfusion and perfusate and liver tissue were collected. The oxidative phosphorylation capacity and extramitochondrial Ca2+ movement were measured by high resolution respirometry. Oxygen and glucose consumption increased significantly while hepatocellular damage was decreased in the HTK-CH4 grafts compared to the HTK group. Mitochondrial oxidative phosphorylation capacity was more preserved (128.8 ± 31.5 pmol/s/mL vs 201.3 ± 54.8 pmol/s/mL) and a significantly higher Ca2+ flux was detected in HTK-CH4 storage (2.9 ± 0.1 mV/s) compared to HTK (2.3 ± 0.09 mV/s). These results demonstrate the direct effect of CH4 on hepatic mitochondrial function and extramitochondrial Ca2+ fluxes, which may have contributed to improved graft functions and a preserved histomorphology after cold IR.
Collapse
|
13
|
Baranovicova E, Kalenska D, Kovalska M, Lehotsky J. Hippocampal metabolic recovery as a manifestation of the protective effect of ischemic preconditioning in rats. Neurochem Int 2022; 160:105419. [PMID: 36113578 DOI: 10.1016/j.neuint.2022.105419] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 08/25/2022] [Accepted: 09/04/2022] [Indexed: 10/31/2022]
Abstract
The ever-present risk of brain ischemic events in humans and its full prevention make the detailed studies of an organism's response to ischemia at different levels essential to understanding the mechanism of the injury as well as protection. We used the four-vessel occlusion as an animal model of forebrain ischemia to investigate its impact on the metabolic alterations in both the hippocampus and the blood plasma to see changes on the systemic level. By inducing sublethal ischemic stimuli, we focused on the endogenous phenomena known as ischemic tolerance. NMR spectroscopy was used to analyze relative metabolite levels in tissue extracts from rats' hippocampus and blood plasma in three various ischemic/reperfusion times: 3 h, 24 h, and 72 h. Hippocampal tissues were characterized by postischemically decreased glutamate and GABA (4-aminobutyrate) tissue content balanced with increased glutamine level, with most pronounced changes at 3 h reperfusion time. Glutamate (as well as glutamine) levels recovered towards the control levels on the third day, as if the glutamate re-synthesis would be firstly preferred before GABA. These results are indicating the higher feasibility of re-establishing of glutamatergic transmission three days after an ischemic event, in contrast to GABA-ergic. Tissue levels of N-acetylaspartate (NAA), as well as choline, were decreased without the tendency to recover three days after the ischemic event. Metabolomic analysis of blood plasma revealed that ischemically preconditioned rats, contrary to the non-preconditioned animals, did not show hyperglycemic conditions. Ischemically induced semi-ketotic state, manifested in increased plasma ketone bodies 3-hydroxybutyrate and acetoacetate, seems to be programmed to support the brain tissue revitalization after the ischemic event. These and other metabolites changes found in blood plasma as well as in the hippocampus were observed to a lower extent or recovered faster in preconditioned animals. Some metabolomic changes in hippocampal tissue extract were so strong that even single metabolites were able to differentiate between ischemic, ischemically preconditioned, and control brain tissues.
Collapse
Affiliation(s)
- Eva Baranovicova
- Biomedical Center BioMed, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Dagmar Kalenska
- Department of Anatomy, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Maria Kovalska
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia
| | - Jan Lehotsky
- Department of Medical Biochemistry, Jessenius Faculty of Medicine, Comenius University in Bratislava, Mala Hora 4, 036 01, Martin, Slovakia.
| |
Collapse
|
14
|
Li W, Xiang Z, Xing Y, Li S, Shi S. Mitochondria bridge HIF signaling and ferroptosis blockage in acute kidney injury. Cell Death Dis 2022; 13:308. [PMID: 35387983 PMCID: PMC8986825 DOI: 10.1038/s41419-022-04770-4] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/15/2022] [Accepted: 03/25/2022] [Indexed: 11/09/2022]
Abstract
AbstractFerroptosis, a form of regulated cell death, plays an important role in acute kidney injury (AKI). Previous studies have shown that prolyl hydroxylase domain protein (PHD) inhibitors that activate HIF signaling provide strong protection against AKI, which is characterized by marked cell death. However, the relationship between PHD inhibition/HIF signaling and ferroptosis in AKI has not been elucidated. Here, we review recent studies to explore the issue. First, we will review the literature concerning the functions of HIF in promoting mitophagy, suppressing mitochondrial respiration and modulating redox homeostasis. Second, we will describe the current understanding of ferroptosis and its role in AKI, particularly from the perspective of mitochondrial dysfunction. Finally, we will discuss the possibility that mitochondria link PHD inhibition/HIF signaling and ferroptosis in AKI. In conclusion, we propose that HIF may protect renal cells against ferroptosis in AKI by reducing mitochondrial oxidative stress and damage.
Collapse
|
15
|
Xin Y, Zhang X, Li J, Gao H, Li J, Li J, Hu W, Li H. New Insights Into the Role of Mitochondria Quality Control in Ischemic Heart Disease. Front Cardiovasc Med 2021; 8:774619. [PMID: 34901234 PMCID: PMC8661033 DOI: 10.3389/fcvm.2021.774619] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 11/09/2021] [Indexed: 02/05/2023] Open
Abstract
IHD is a significant cause of mortality and morbidity worldwide. In the acute phase, it's demonstrated as myocardial infarction and ischemia-reperfusion injury, while in the chronic stage, the ischemic heart is mainly characterised by adverse myocardial remodelling. Although interventions such as thrombolysis and percutaneous coronary intervention could reduce the death risk of these patients, the underlying cellular and molecular mechanisms need more exploration. Mitochondria are crucial to maintain the physiological function of the heart. During IHD, mitochondrial dysfunction results in the pathogenesis of ischemic heart disease. Ischemia drives mitochondrial damage not only due to energy deprivation, but also to other aspects such as mitochondrial dynamics, mitochondria-related inflammation, etc. Given the critical roles of mitochondrial quality control in the pathological process of ischemic heart disease, in this review, we will summarise the efforts in targeting mitochondria (such as mitophagy, mtROS, and mitochondria-related inflammation) on IHD. In addition, we will briefly revisit the emerging therapeutic targets in this field.
Collapse
Affiliation(s)
- Yanguo Xin
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xiaodong Zhang
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,National Clinical Research Center for Digestive Diseases, Beijing, China
| | - Jingye Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Hui Gao
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jiayu Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Junli Li
- Laboratory of Heart Valve Disease, West China Hospital, Sichuan University, Chengdu, China
| | - Wenyu Hu
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hongwei Li
- Department of Cardiology, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China.,Beijing Key Laboratory of Metabolic Disorder Related Cardiovascular Disease, Beijing, China.,Department of Geriatrics, Cardiovascular Center, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
16
|
Yi T, Chen H, Zhan J, Li Y, Long Z, Wu Z, Yang M, Peng T, Li H. Ubiquinol-cytochrome c reductase core protein 1 contributes to cardiac tolerance to acute exhaustive exercise. Exp Biol Med (Maywood) 2021; 247:165-173. [PMID: 34648372 DOI: 10.1177/15353702211046546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Ubiquinol-cytochrome c reductase core protein 1 (UQCRC1) is an indispensable component of mitochondrial complex III. It plays a key role in cardioprotection and maintaining mitochondrion function. However, the exact role of UQCRC1 in maintaining cardiac function has not been reported by in vivo models. Also, the exact biological functions of UQCRC1 are far from fully understood. UQCRC1+/- mice had decreased both mRNA and protein expression of UQCRC1 in the left ventricular myocardia, and these mice had reduced tolerance to acute exhaustive exercise including decreased time and distance with higher apoptosis rate, higher expression level of cleaved CASPASE 3, and higher ratio of cleaved PARP1 to full-length PARP1. Moreover, UQCRC1 knockdown led to increased LV interventricular septal thicknesses both at systole and diastole, as well as decreased LV volume both at end-systole and end-diastole. Finally, UQCRC1 gene disruption resulted in mitochondrial vacuolation, fibril disarrangement, and more severe morphological and structural changes in mitochondria after acute exhaustive exercise. In conclusion, UQCRC1 contributes to cardiac tolerance to acute exhaustive exercise in mice, and it may be an essential component of complex III, playing a crucial role in maintaining cardiac functions.
Collapse
Affiliation(s)
- Tingting Yi
- Department of Anesthesiology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China.,Department of Anesthesiology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Huifang Chen
- Department of Anesthesiology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China.,Department of Radiology, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, Chongqing 400030, China
| | - Jian Zhan
- Department of Anesthesiology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China.,Department of Anesthesiology, Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan, China
| | - Yu Li
- Department of Anesthesiology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Zonghong Long
- Department of Anesthesiology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Zhuoxi Wu
- Department of Anesthesiology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Mi Yang
- Department of Anesthesiology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Taotao Peng
- Department of Anesthesiology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Hong Li
- Department of Anesthesiology, Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
17
|
Morio A, Tsutsumi R, Kondo T, Miyoshi H, Kato T, Narasaki S, Satomi S, Nakaya E, Kuroda M, Sakaue H, Kitamura T, Tsutsumi YM. Leucine induces cardioprotection in vitro by promoting mitochondrial function via mTOR and Opa-1 signaling. Nutr Metab Cardiovasc Dis 2021; 31:2979-2986. [PMID: 34362635 DOI: 10.1016/j.numecd.2021.06.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 01/30/2023]
Abstract
BACKGROUND AND AIMS Coronary heart disease is a major global health concern. Further, severity of this condition is greatly influenced by myocardial ischemia/reperfusion (I/R) injury. Branched-chain amino acids (BCAAs) have cardioprotective effects against I/R via mammalian target of rapamycin (mTOR) activity, wherein Leu is considered to particularly regulate mTOR activation. However, the mechanism underlying cardioprotective effects of Leu via mTOR activity is not fully elucidated. Here, we aimed to study the signaling pathway of cardioprotection and mitochondrial function induced by Leu treatment. METHODS AND RESULTS Cardiac myocytes isolated from adult male Wistar rats were incubated and exposed to simulated I/R (SI/R) injury by replacing the air content. Cardiac myocytes were treated with Leu and subsequently, their survival rate was calculated. To elucidate the signaling pathway and mitochondrial function, immunoblots and mitochondrial permeability transition pore were examined. Cell survival rate was decreased with SI/R but improved by 160 μM Leu (38.5 ± 3.6% vs. 64.5 ± 4.2%, respectively, p < 0.001). Although rapamycin (mTOR inhibitor) prevented this cardioprotective effect induced by Leu, wortmannin (PI3K inhibitor) did not interfere with this effect. In addition, we indicated that overexpression of Opa-1 and mitochondrial function are ameliorated via Leu-induced mitochondrial biogenesis. In contrast, knockdown of Opa-1 suppressed Leu-induced cardioprotection. CONCLUSION Leu treatment is critical in rendering a cardioprotective effect exhibited by BCAAs via mTOR signaling. Furthermore, Leu improved mitochondrial function.
Collapse
Affiliation(s)
- Atsushi Morio
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Rie Tsutsumi
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Takashi Kondo
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Hirotsugu Miyoshi
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Takahiro Kato
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Soshi Narasaki
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Shiho Satomi
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan
| | - Erika Nakaya
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Masashi Kuroda
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Hiroshi Sakaue
- Department of Nutrition and Metabolism, Institute of Biomedical Sciences, Tokushima University, Tokushima, Japan
| | - Tadahiro Kitamura
- Laboratory of Metabolic Signal, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Yasuo M Tsutsumi
- Department of Anesthesiology and Critical Care, Hiroshima University, Hiroshima, Japan.
| |
Collapse
|
18
|
Kulawiak B, Bednarczyk P, Szewczyk A. Multidimensional Regulation of Cardiac Mitochondrial Potassium Channels. Cells 2021; 10:1554. [PMID: 34205420 PMCID: PMC8235349 DOI: 10.3390/cells10061554] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
Mitochondria play a fundamental role in the energetics of cardiac cells. Moreover, mitochondria are involved in cardiac ischemia/reperfusion injury by opening the mitochondrial permeability transition pore which is the major cause of cell death. The preservation of mitochondrial function is an essential component of the cardioprotective mechanism. The involvement of mitochondrial K+ transport in this complex phenomenon seems to be well established. Several mitochondrial K+ channels in the inner mitochondrial membrane, such as ATP-sensitive, voltage-regulated, calcium-activated and Na+-activated channels, have been discovered. This obliges us to ask the following question: why is the simple potassium ion influx process carried out by several different mitochondrial potassium channels? In this review, we summarize the current knowledge of both the properties of mitochondrial potassium channels in cardiac mitochondria and the current understanding of their multidimensional functional role. We also critically summarize the pharmacological modulation of these proteins within the context of cardiac ischemia/reperfusion injury and cardioprotection.
Collapse
Affiliation(s)
- Bogusz Kulawiak
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland;
| | - Piotr Bednarczyk
- Department of Physics and Biophysics, Institute of Biology, Warsaw University of Life Sciences-SGGW, Nowoursynowska 159, 02-776 Warsaw, Poland;
| | - Adam Szewczyk
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Pasteura 3, 02-093 Warsaw, Poland;
| |
Collapse
|
19
|
Orlandi M, Masi S, Bhowruth D, Leira Y, Georgiopoulos G, Yellon D, Hingorani A, Chiesa ST, Hausenloy DJ, Deanfield J, D'Aiuto F. Remote Ischemic Preconditioning Protects Against Endothelial Dysfunction in a Human Model of Systemic Inflammation: A Randomized Clinical Trial. Arterioscler Thromb Vasc Biol 2021; 41:e417-e426. [PMID: 34107730 DOI: 10.1161/atvbaha.121.316388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Marco Orlandi
- Periodontology Unit, UCL Eastman Dental Institute and Hospital (M.O., Y.L., F.D.), University College London, United Kingdom
| | - Stefano Masi
- National Centre for Cardiovascular Prevention and Outcomes Institute of Cardiovascular Science (S.M., D.B., S.T.C., J.D.), University College London, United Kingdom.,Internal Medicine Unit, University of Pisa, Italy (S.M.)
| | - Devina Bhowruth
- National Centre for Cardiovascular Prevention and Outcomes Institute of Cardiovascular Science (S.M., D.B., S.T.C., J.D.), University College London, United Kingdom
| | - Yago Leira
- Periodontology Unit, UCL Eastman Dental Institute and Hospital (M.O., Y.L., F.D.), University College London, United Kingdom.,Periodontology Unit, Faculty of Odontology, University of Santiago de Compostela and Medical-Surgical Dentistry Research Group (Y.L.), Health Research Institute of Santiago de Compostela, Spain.,Clinical Neurosciences Research Laboratory (Y.L.), Health Research Institute of Santiago de Compostela, Spain
| | - Georgios Georgiopoulos
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas Hospital, United Kingdom (G.G.)
| | - Derek Yellon
- The Hatter Cardiovascular Institute (D.Y., D.J.H.), University College London, United Kingdom
| | - Aroon Hingorani
- Institute of Cardiovascular Science (A.H.), University College London, United Kingdom
| | - Scott T Chiesa
- National Centre for Cardiovascular Prevention and Outcomes Institute of Cardiovascular Science (S.M., D.B., S.T.C., J.D.), University College London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute (D.Y., D.J.H.), University College London, United Kingdom.,Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore (D.J.H.).,National Heart Research Institute Singapore, National Heart Centre, Singapore (D.J.H.).,Yong Loo Lin School of Medicine, National University Singapore, Singapore (D.J.H.).,Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taiwan (D.J.H.)
| | - John Deanfield
- National Centre for Cardiovascular Prevention and Outcomes Institute of Cardiovascular Science (S.M., D.B., S.T.C., J.D.), University College London, United Kingdom
| | - Francesco D'Aiuto
- Periodontology Unit, UCL Eastman Dental Institute and Hospital (M.O., Y.L., F.D.), University College London, United Kingdom
| |
Collapse
|
20
|
Hadrava Vanova K, Kraus M, Neuzil J, Rohlena J. Mitochondrial complex II and reactive oxygen species in disease and therapy. Redox Rep 2021; 25:26-32. [PMID: 32290794 PMCID: PMC7178880 DOI: 10.1080/13510002.2020.1752002] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Increasing evidence points to the respiratory Complex II (CII) as a source and modulator of reactive oxygen species (ROS). Both functional loss of CII as well as its pharmacological inhibition can lead to ROS generation in cells, with a relevant impact on the development of pathophysiological conditions, i.e. cancer and neurodegenerative diseases. While the basic framework of CII involvement in ROS production has been defined, the fine details still await clarification. It is important to resolve these aspects to fully understand the role of CII in pathology and to explore its therapeutic potential in cancer and other diseases.
Collapse
Affiliation(s)
| | - Michal Kraus
- Institute of Biotechnology of the Czech Academy of Sciences, Prague-West, Czech Republic
| | - Jiri Neuzil
- Institute of Biotechnology of the Czech Academy of Sciences, Prague-West, Czech Republic.,School of Medical Science, Griffith University, Southport, Qld, Australia
| | - Jakub Rohlena
- Institute of Biotechnology of the Czech Academy of Sciences, Prague-West, Czech Republic
| |
Collapse
|
21
|
Liu Y, Song Y, Li S, Mo L. Cardioprotective Effect of Quercetin against Ischemia/Reperfusion Injury Is Mediated Through NO System and Mitochondrial K-ATP Channels. CELL JOURNAL 2021; 23:184-190. [PMID: 34096219 PMCID: PMC8181321 DOI: 10.22074/cellj.2021.7183] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 10/30/2019] [Indexed: 02/05/2023]
Abstract
Objective Quercetin (Que) is a plant-derived polyphenolic compound, that was shown to possess anti-inflammatory
activity in myocardial ischemia/reperfusion (I/R) models in vivo; however, detailed mechanisms of its anti-inflammatory
effects remain unclear. This study aimed to examine the effects of quercetin postconditioning (QPC) on I/R-induced
inflammatory response in a rat model and evaluate the role of the mitochondrial K-ATP (mitoKATP) channels and NO
system in this regard.
Materials and Methods In this experimental study, hearts of male Wistar rats (250 ± 20 g) perused by Langendorff
apparatus, were subjected to 30 minutes of global ischemia followed by 55 minutes reperfusion, and Que was added
to the perfusion solution immediately at the onset of reperfusion. Creatine kinase (CK) levels in the coronary effluent
were measured by spectrophotometry. Interleukin-1 (IL-1β), IL-6, and tumor necrosis factor-alpha (TNF-α) levels were
analyzed by an enzyme-linked immunosorbent assay (ELISA) rat specific kit to assess the inflammatory condition of
the myocardial tissue.
Results Our results showed that QPC significantly improved left ventricular developed pressure (LVDP) (P<0.05), and
decreased the CK release into the coronary effluent vs. control group (P<0.01). The levels of IL-1β (P<0.01), TNF-α
(P<0.01), and IL-6 (P<0.05) were significantly diminished in Que-treated groups when compared to the control group.
Inhibiting mitoKATPchannels by 100 μM 5-hydroxydecanoate and blocking NO system by 100 μM L-NAME reversed the
cardioprotective effects of Que.
Conclusion The findings of this study suggested that QPC exerts cardioprotective effects on myocardial I/R injury
(MIRI) through inhibition of inflammatory reactions and improvement of contractility potential. Also, mitoKATP channels
and NO system might be involved in this anti-inflammatory effect.
Collapse
Affiliation(s)
- Ying Liu
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Song
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Siyuan Li
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Li Mo
- The Center of Gerontology and Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
22
|
Khaliulin I, Ascione R, Maslov LN, Amal H, Suleiman MS. Preconditioning or Postconditioning with 8-Br-cAMP-AM Protects the Heart against Regional Ischemia and Reperfusion: A Role for Mitochondrial Permeability Transition. Cells 2021; 10:1223. [PMID: 34067674 PMCID: PMC8155893 DOI: 10.3390/cells10051223] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 05/12/2021] [Accepted: 05/13/2021] [Indexed: 01/15/2023] Open
Abstract
The cAMP analogue 8-Br-cAMP-AM (8-Br) confers marked protection against global ischaemia/reperfusion of isolated perfused heart. We tested the hypothesis that 8-Br is also protective under clinically relevant conditions (regional ischaemia) when applied either before ischemia or at the beginning of reperfusion, and this effect is associated with the mitochondrial permeability transition pore (MPTP). 8-Br (10 μM) was administered to Langendorff-perfused rat hearts for 5 min either before or at the end of 30 min regional ischaemia. Ca2+-induced mitochondria swelling (a measure of MPTP opening) and binding of hexokinase II (HKII) to mitochondria were assessed following the drug treatment at preischaemia. Haemodynamic function and ventricular arrhythmias were monitored during ischaemia and 2 h reperfusion. Infarct size was evaluated at the end of reperfusion. 8-Br administered before ischaemia attenuated ventricular arrhythmias, improved haemodynamic function, and reduced infarct size during ischaemia/reperfusion. Application of 8-Br at the end of ischaemia protected the heart during reperfusion. 8-Br promoted binding of HKII to the mitochondria and reduced Ca2+-induced mitochondria swelling. Thus, 8-Br protects the heart when administered before regional ischaemia or at the beginning of reperfusion. This effect is associated with inhibition of MPTP via binding of HKII to mitochondria, which may underlie the protective mechanism.
Collapse
Affiliation(s)
- Igor Khaliulin
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Pharmacy Building, Ein Karem, Jerusalem 91120, Israel;
- Bristol Medical School (THS), Faculty of Health Sciences, University of Bristol, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK; (R.A.); (M.S.S.)
| | - Raimondo Ascione
- Bristol Medical School (THS), Faculty of Health Sciences, University of Bristol, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK; (R.A.); (M.S.S.)
| | - Leonid N. Maslov
- Cardiology Research Institute, Tomsk National Research Medical Center, The Russian Academy of Sciences, 111 a, Kievskaya Street, 634012 Tomsk, Russia;
| | - Haitham Amal
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, Hebrew University of Jerusalem, Pharmacy Building, Ein Karem, Jerusalem 91120, Israel;
| | - M. Saadeh Suleiman
- Bristol Medical School (THS), Faculty of Health Sciences, University of Bristol, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK; (R.A.); (M.S.S.)
| |
Collapse
|
23
|
Connexins in the Heart: Regulation, Function and Involvement in Cardiac Disease. Int J Mol Sci 2021; 22:ijms22094413. [PMID: 33922534 PMCID: PMC8122935 DOI: 10.3390/ijms22094413] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 04/12/2021] [Accepted: 04/20/2021] [Indexed: 12/20/2022] Open
Abstract
Connexins are a family of transmembrane proteins that play a key role in cardiac physiology. Gap junctional channels put into contact the cytoplasms of connected cardiomyocytes, allowing the existence of electrical coupling. However, in addition to this fundamental role, connexins are also involved in cardiomyocyte death and survival. Thus, chemical coupling through gap junctions plays a key role in the spreading of injury between connected cells. Moreover, in addition to their involvement in cell-to-cell communication, mounting evidence indicates that connexins have additional gap junction-independent functions. Opening of unopposed hemichannels, located at the lateral surface of cardiomyocytes, may compromise cell homeostasis and may be involved in ischemia/reperfusion injury. In addition, connexins located at non-canonical cell structures, including mitochondria and the nucleus, have been demonstrated to be involved in cardioprotection and in regulation of cell growth and differentiation. In this review, we will provide, first, an overview on connexin biology, including their synthesis and degradation, their regulation and their interactions. Then, we will conduct an in-depth examination of the role of connexins in cardiac pathophysiology, including new findings regarding their involvement in myocardial ischemia/reperfusion injury, cardiac fibrosis, gene transcription or signaling regulation.
Collapse
|
24
|
Preventing Myocardial Injury Following Non-Cardiac Surgery: A Potential Role for Preoperative Antioxidant Therapy with Ubiquinone. Antioxidants (Basel) 2021; 10:antiox10020276. [PMID: 33579045 PMCID: PMC7916807 DOI: 10.3390/antiox10020276] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 02/07/2023] Open
Abstract
Over 240 million non-cardiac operations occur each year and are associated with a 15-20% incidence of adverse perioperative cardiovascular events. Unfortunately, preoperative therapies that have been useful for chronic ischemic heart diseases, such as coronary artery revascularization, antiplatelet agents, and beta-blockers have failed to improve outcomes. In a pre-clinical swine model of ischemic heart disease, we showed that daily administration of ubiquinone (coenzyme Q10, CoQ10) enhances the antioxidant status of mitochondria within chronically ischemic heart tissue, potentially via a PGC1α-dependent mechanism. In a randomized controlled trial, among high-risk patients undergoing elective vascular surgery, we showed that NT Pro-BNP levels are an important means of risk-stratification during the perioperative period and can be lowered with administration of CoQ10 (400 mg/day) for 3 days prior to surgery. The review provides background information for the role of oxidant stress and inflammation during high-risk operations and the potential novel application of ubiquinone as a preoperative antioxidant therapy that might reduce perioperative adverse cardiovascular outcomes.
Collapse
|
25
|
Røsand Ø, Høydal MA. Cardiac Exosomes in Ischemic Heart Disease- A Narrative Review. Diagnostics (Basel) 2021; 11:diagnostics11020269. [PMID: 33572486 PMCID: PMC7916440 DOI: 10.3390/diagnostics11020269] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/04/2021] [Accepted: 02/07/2021] [Indexed: 12/15/2022] Open
Abstract
Ischemic heart disease (IHD) is the primary cause of death globally. IHD is associated with the disruption of blood supply to the heart muscles, which often results in myocardial infarction (MI) that further may progress to heart failure (HF). Exosomes are a subgroup of extracellular vesicles that can be secreted by virtually all types of cells, including cardiomyocytes, cardiac fibroblasts, endothelial cells, and stem and progenitor cells. Exosomes represent an important means of cell–cell communication through the transport of proteins, coding and non-coding RNA, and other bioactive molecules. Several studies show that exosomes play an important role in the progression of IHD, including endothelial dysfunction, the development of arterial atherosclerosis, ischemic reperfusion injury, and HF development. Recently, promising data have been shown that designates exosomes as carriers of cardioprotective molecules that enhance the survival of recipient cells undergoing ischemia. In this review, we summarize the functional involvement of exosomes regarding IHD. We also highlight the cardioprotective effects of native and bioengineered exosomes to IHD, as well as the possibility of using exosomes as natural biomarkers of cardiovascular diseases. Lastly, we discuss the opportunities and challenges that need to be addressed before exosomes can be used in clinical applications.
Collapse
|
26
|
Rameshrad M, Omidkhoda SF, Razavi BM, Hosseinzadeh H. Evaluating the possible role of mitochondrial ATP-sensitive potassium channels in the cardioprotective effects of morin in the isolated rat heart. Life Sci 2021; 264:118659. [PMID: 33148418 DOI: 10.1016/j.lfs.2020.118659] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 12/28/2022]
Abstract
AIMS During heart ischemia, the lack of oxygen in the myocardial cells causes pH and ion disturbances and cell death through opening mitochondrial permeability transition pores (mPTP). Considering the inhibitory effects of mitochondrial ATP-dependent potassium channels (mt-KATP) on these pores and anti-ischemic effects of morin, we hypothesized that it may exert its positive effects via activating mt-KATP as well as its anti-oxidative effects. MAIN METHODS Isolated rat hearts were perfused by Krebs-Henseleit solution enriched with the morin (0.25, 0.5 and 1 mg/L) or 5-hydroxydecanoate (5-HD, a mt-KATP blocker;100 μM) or both as needed 5 min before starting regional ischemia till the first 10 min of the reperfusion period. The reperfusion was developed with Krebs-Henseleit solution 60 or 120 min respectively for biochemical evaluations (lactate dehydrogenase and malondialdehyde level) or the assessment of myocardial infarct size. During the experiments, hemodynamic functions were recorded and cardiac arrhythmias were determined. KEY FINDINGS Our findings demonstrated that morin reduced the infarct size. Also, morin perfusion could remarkably prevent the malondialdehyde over-production during ischemia. Total ventricular ectopic beats had the same significant changes as the malondialdehyde level, in both ischemia and reperfusion phases. Morin could also relatively improve the ischemia-induced hemodynamic dysfunction. All mentioned protective effects of morin were reversed by concomitant perfusion of 5-HD. SIGNIFICANCE Morin has protective effects against ischemic hearts through anti-oxidative effects. It also suggests a link between the cardioprotective effects of morin and mt-KATP. However, additional studies are required to prove this preliminary hypothesis.
Collapse
Affiliation(s)
- Maryam Rameshrad
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Seyedeh Farzaneh Omidkhoda
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Bibi Marjan Razavi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Targeted Drug Delivery Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hossein Hosseinzadeh
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
27
|
Sabet Sarvestani F, Azarpira N, Al-Abdullah IH, Tamaddon AM. microRNAs in liver and kidney ischemia reperfusion injury: insight to improve transplantation outcome. Biomed Pharmacother 2020; 133:110944. [PMID: 33227704 DOI: 10.1016/j.biopha.2020.110944] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 10/03/2020] [Accepted: 10/25/2020] [Indexed: 12/26/2022] Open
Abstract
Ischemia reperfusion injury (IRI) is a condition that occurs wherever blood flow and oxygen is reduced or absent, such as trauma, vascular disease, stroke, and solid organ transplantation. This condition can lead to tissue damage, especially during organ transplantation. Under such circumstances, some signaling pathways are activated, leading to up- or down- regulation of several genes such as microRNAs (miRNAs) that might attenuate or ameliorate this status. Therefore, by manipulating miRNAs level, they can be used as a biomarker for early diagnosis of IRI or suggestive to be therapeutic agents in clinical situation in future.
Collapse
Affiliation(s)
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ismail H Al-Abdullah
- Department of Translational Research and Cellular Therapeutics, Diabetes and Metabolism Research Institute, Beckman Research Institute of City of Hope, Duarte, USA.
| | - Ali-Mohammad Tamaddon
- Department of Pharmaceutics and Center for Nanotechnology in Drug Delivery, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
28
|
Brown SM, Larsen NK, Thankam FG, Agrawal DK. Fetal cardiomyocyte phenotype, ketone body metabolism, and mitochondrial dysfunction in the pathology of atrial fibrillation. Mol Cell Biochem 2020; 476:1165-1178. [PMID: 33188453 DOI: 10.1007/s11010-020-03980-8] [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] [Received: 07/29/2020] [Accepted: 11/06/2020] [Indexed: 10/23/2022]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia diagnosed in clinical practice. Even though hypertension, congestive heart failure, pulmonary disease, and coronary artery disease are the potential risk factors for AF, the underlying molecular pathology is largely unknown. The reversion of the mature cardiomyocytes to fetal phenotype, impaired ketone body metabolism, mitochondrial dysfunction, and the cellular effect of reactive oxygen species (ROS) are the major underlying biochemical events associated with the molecular pathology of AF. On this background, the present manuscript sheds light into these biochemical events in regard to the metabolic derangements in cardiomyocyte leading to AF, especially with respect to structural, contractile, and electrophysiological properties. In addition, the article critically reviews the current understanding, potential demerits, and translational strategies in the management of AF.
Collapse
Affiliation(s)
- Sean M Brown
- Creighton University School of Medicine, Omaha, NE, 68178, USA
| | | | - Finosh G Thankam
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
29
|
The association between left ventricular mass index and serum sirtuin 3 level in patients with hypertension. Cardiovasc Endocrinol Metab 2020; 10:99-105. [PMID: 34113795 DOI: 10.1097/xce.0000000000000231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/15/2020] [Indexed: 11/26/2022]
Abstract
Objectives Sirtuin 3 (SIRT3) can protect cardiomyocytes from oxidative stress-mediated cell damage and prevent cardiac hypertrophy development. The aim of this study was to evaluate whether a relationship existed between left ventricular mass index (LVMI) and serum SIRT3 levels in patients with hypertension. Patients and methods This study was conducted as a cross-sectional study of 83 patients between April 2018 and October 2018. The LVMI of all patients was calculated using the formula of the American Echocardiography Association and patients were divided into two groups according to results (increased LVMI and normal LVMI). Results Increased LVMI was determined in 37.3% of patients, whereas 62.7% had normal LVMI. There was no significant difference between serum SIRT3 levels between those with increased LVMI and normal LVMI (5.8 versus 5.4 ng/ml; P = 0.914). Serum pro-brain natriuretic peptide levels (69 versus 41 ng/ml; P = 0.019) were found to be higher in patients with increased LVMI than in those with normal LVMI. A positive correlation between SIRT3 levels and Sm (myocardial systolic) velocity was also determined (r = 0.338; P = 0.002). Conclusion The serum levels of SIRT3, a molecule which has been proposed to have protective properties against myocardial hypertrophy, were not found to be correlated with LVMI values; however, SIRT3 levels were found to be correlated with Sm velocity, which is accepted to be an indicator of myocardial early diastolic dysfunction.
Collapse
|
30
|
Ubiquinol-cytochrome c reductase core protein 1 overexpression protects H9c2 cardiac cells against mimic ischemia/reperfusion injury through PI3K/Akt/GSK-3β pathway. Biochem Biophys Res Commun 2020; 529:904-909. [PMID: 32819597 DOI: 10.1016/j.bbrc.2020.06.089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 11/20/2022]
Abstract
Ubiquinol-cytochrome c reductase core protein 1 (UQCRC1) plays a key role in influencing mitochondrial function. Increasing evidence supports that UQCRC1 overexpression takes part in cardioprotection. However, it remains unclear about the signaling pathway mediating the protective role of UQCRC1 overexpression. Thus, the current study aimed to investigate the signaling pathway. Inhibition of PI3K completely abolished the protective effects of UQCRC1 overexpression on cell viability and mitochondrial membrane potential after OGD or hydrogen peroxide injury in H9c2 cardiac cells, while inhibition of ERK only partially abolished these effects. Moreover, UQCRC1 overexpression dramatically increased the phosphorylation of PI3K downstream signal molecules including Akt and GSK-3β. Finally, UQCRC1 overexpression upregulated the expression of antiapoptotic protein Bcl-2, downregulated the expression of proapoptotic protein Bax, decreased active caspase 3 expression and cell apoptosis, which were completely abolished by inhibition of PI3K. In conclusion, UQCRC1 overexpression protects H9c2 cardiac cells against mimic ischemia/reperfusion injury through mediating PI3K/Akt/GSK-3β pathway to regulate apoptosis-related proteins.
Collapse
|
31
|
Menezes-Rodrigues FS, Tavares JGP, Vasques ER, Errante PR, Araújo EAD, Pires-Oliveira M, Scorza CA, Scorza FA, Taha MO, Caricati-Neto A. Cardioprotective effects of pharmacological blockade of the mitochondrial calcium uniporter on myocardial ischemia-reperfusion injury. Acta Cir Bras 2020; 35:e202000306. [PMID: 32692797 PMCID: PMC7251977 DOI: 10.1590/s0102-865020200030000006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Accepted: 02/22/2020] [Indexed: 05/13/2023] Open
Abstract
PURPOSE To evaluate whether the attenuation of mitochondrial Ca2+ overload produced by pharmacological blockade of mitochondrial Ca2+ uniporter (MCU) protects the myocardium against injuries caused by cardiac ischemia and reperfusion (CIR). METHODS CIR was induced in adult male Wistar rats (300-350 g) by occlusion of the left anterior descendent coronary artery (10 min), followed by reperfusion (120 min). Rats were treated with different doses of MCU blocker ruthenium red (RuR), administered 5 min before ischemia or reperfusion. RESULTS In untreated rats, the incidences of ventricular arrhythmias (VA), atrioventricular block (AVB) and the lethality (LET) induced by CIR were 85%, 79% and 70%, respectively. In rats treated with RuR before ischemia, the incidences of VA, AVB and LET were significantly reduced to 62%, 25% and 25%, respectively. In rats treated with RuR after ischemia, the incidences of VA, AVB and LET were significantly reduced to 50%, 25% and 25%, respectively. CONCLUSION The significant reduction of the incidence of CIR-induced VA, AVB and LET produced by the treatment with RuR indicates that the attenuation of mitochondrial Ca2+ overload produced by pharmacological blockade of MCU can protect the myocardium against injuries caused by CIR.
Collapse
|
32
|
The Role of Oxidative Stress in Cardiac Disease: From Physiological Response to Injury Factor. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:5732956. [PMID: 32509147 PMCID: PMC7244977 DOI: 10.1155/2020/5732956] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2019] [Revised: 01/11/2020] [Accepted: 04/22/2020] [Indexed: 02/07/2023]
Abstract
Reactive oxygen species (ROS) are highly reactive chemical species containing oxygen, controlled by both enzymatic and nonenzymatic antioxidant defense systems. In the heart, ROS play an important role in cell homeostasis, by modulating cell proliferation, differentiation, and excitation-contraction coupling. Oxidative stress occurs when ROS production exceeds the buffering capacity of the antioxidant defense systems, leading to cellular and molecular abnormalities, ultimately resulting in cardiac dysfunction. In this review, we will discuss the physiological sources of ROS in the heart, the mechanisms of oxidative stress-related myocardial injury, and the implications of experimental studies and clinical trials with antioxidant therapies in cardiovascular diseases.
Collapse
|
33
|
Maciel L, de Oliveira DF, Monnerat G, Campos de Carvalho AC, Nascimento JHM. Exogenous 10 kDa-Heat Shock Protein Preserves Mitochondrial Function After Hypoxia/Reoxygenation. Front Pharmacol 2020; 11:545. [PMID: 32431608 PMCID: PMC7214810 DOI: 10.3389/fphar.2020.00545] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/09/2020] [Indexed: 02/05/2023] Open
Abstract
Humoral factors released during ischemic preconditioning (IPC) protect the myocardium against ischemia/reperfusion (I/R) injury. We have recently identified 10 kDa-heat shock protein (HSP10) and a fraction of small 5-10 kDa peptides (5-10-sP) in the coronary effluent of IPC-treated hearts and demonstrated their cardioprotective potential. We here used our isolated mitochondria model to characterize the impact of exogenous HSP10 and 5-10-sP on mitochondria function from myocardium subjected to I/R injury. Isolated perfused rat hearts were submitted to 30-min global ischemia and 10-min reperfusion. Before ischemia, isolated hearts were infused with saline or 5-10-sP, with or without a mitochondrial ATP-sensitive-K+-channel blocker (5HD 10 μmol·L-1) or PKC inhibitor (chelerythrine 10 μmol·L-1), before I/R. HSP10 (1 µmol·L-1) was infused into isolated hearts before I/R without blockers. At 10-min reperfusion, the mitochondria were isolated and mitochondrial function was assessed. In a subset of experiments, freshly isolated mitochondria were directly incubated with HSP10 or 5-10-sP with or without 5HD or chelerythrine before in vitro hypoxia/reoxygenation. Infusion of 5-10-sP (n = 5) and HSP10 (n = 5) into isolated hearts before I/R improved mitochondrial ADP-stimulated respiration, ATP production and prevented mitochondrial ROS formation compared to the I/R group (n = 5); this effect was abrogated by 5HD and chelerythrine. In freshly isolated mitochondria with in vitro hypoxia/reoxygenation, HSP10 (n = 16) and 5-10-sP (n = 16) incubation prevented reductions of mitochondrial ADP-stimulated respiration (91.5 ± 5.1 nmol O2/min/mg PTN), ATP production (250.1 ± 9.3 μmol ATP/200μg PTN), and prevented mitochondrial ROS production (219.7 ± 9.0 nmol H2O2/200μg PTN) induced by hypoxia/reoxygenation (n = 12, 51.5 ± 5.0 nmol O2/min/mg PTN; 187 ± 21.7 μmol ATP/200 μg PTN; 339.0 ± 14.3 nmol H2O2/200 μg PTN, p < 0.001, respectively). 5HD reduced the ADP-stimulated respiration in the HSP10 group (65.84 ± 3.3 nmol O2/min/mg PTN), ATP production (193.7 ± 12.1 μmol ATP/200μg PTN) and increased ROS in the 5-10-sP group (274.4 ± 21.7 nmol H2O2/200 μg PTN). Mitochondria are a target of the cardioprotection induced by 5-10-sP and HSP10. This protection is dependent of PKC and mKATP activation. HSP10 can act directly on mitochondria and protects against hypoxia/reoxygenation injury by mKATP activation.
Collapse
Affiliation(s)
- Leonardo Maciel
- Laboratory of Cardiac Electrophysiology Antônio Paes de Carvalho, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Dahienne Ferreira de Oliveira
- Laboratory of Cardiac Electrophysiology Antônio Paes de Carvalho, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Gustavo Monnerat
- Laboratory of Cardiac Electrophysiology Antônio Paes de Carvalho, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Cardiology, Rio de Janeiro, Brazil
| | - Antonio Carlos Campos de Carvalho
- Laboratory of Cardiac Electrophysiology Antônio Paes de Carvalho, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
- National Institute of Cardiology, Rio de Janeiro, Brazil
| | - Jose Hamilton Matheus Nascimento
- Laboratory of Cardiac Electrophysiology Antônio Paes de Carvalho, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
34
|
Gallyas Jr. F, Sumegi B. Mitochondrial Protection by PARP Inhibition. Int J Mol Sci 2020; 21:ijms21082767. [PMID: 32316192 PMCID: PMC7215481 DOI: 10.3390/ijms21082767] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Inhibitors of the nuclear DNA damage sensor and signalling enzyme poly(ADP-ribose) polymerase (PARP) have recently been introduced in the therapy of cancers deficient in double-strand DNA break repair systems, and ongoing clinical trials aim to extend their use from other forms of cancer non-responsive to conventional treatments. Additionally, PARP inhibitors were suggested to be repurposed for oxidative stress-associated non-oncological diseases resulting in a devastating outcome, or requiring acute treatment. Their well-documented mitochondria- and cytoprotective effects form the basis of PARP inhibitors’ therapeutic use for non-oncological diseases, yet can limit their efficacy in the treatment of cancers. A better understanding of the processes involved in their protective effects may improve the PARP inhibitors’ therapeutic potential in the non-oncological indications. To this end, we endeavoured to summarise the basic features regarding mitochondrial structure and function, review the major PARP activation-induced cellular processes leading to mitochondrial damage, and discuss the role of PARP inhibition-mediated mitochondrial protection in several oxidative stress-associated diseases.
Collapse
Affiliation(s)
- Ferenc Gallyas Jr.
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary;
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary
- HAS-UP Nuclear-Mitochondrial Interactions Research Group, 1245 Budapest, Hungary
- Correspondence: ; Tel.: +36-72-536-278
| | - Balazs Sumegi
- Department of Biochemistry and Medical Chemistry, University of Pecs Medical School, 7624 Pecs, Hungary;
- Szentagothai Research Centre, University of Pecs, 7624 Pecs, Hungary
- HAS-UP Nuclear-Mitochondrial Interactions Research Group, 1245 Budapest, Hungary
| |
Collapse
|
35
|
Satomi S, Morio A, Miyoshi H, Nakamura R, Tsutsumi R, Sakaue H, Yasuda T, Saeki N, Tsutsumi YM. Branched-chain amino acids-induced cardiac protection against ischemia/reperfusion injury. Life Sci 2020; 245:117368. [DOI: 10.1016/j.lfs.2020.117368] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 01/11/2020] [Accepted: 01/26/2020] [Indexed: 02/08/2023]
|
36
|
Sampieri R, Fuentes E, Carrillo ED, Hernández A, García MC, Sánchez JA. Pharmacological Preconditioning Using Diazoxide Regulates Store-Operated Ca 2 + Channels in Adult Rat Cardiomyocytes. Front Physiol 2020; 10:1589. [PMID: 32009985 PMCID: PMC6972595 DOI: 10.3389/fphys.2019.01589] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/19/2019] [Indexed: 01/31/2023] Open
Abstract
Voltage-dependent Ca2+ channels and store-operated Ca2+ channels (SOCs) are the major routes of Ca2+ entry into mammalian cells. Previously, we reported that pharmacological preconditioning (PPC) leads to a decrease in the amplitude of L-type calcium channel current in the heart. In this study, we examined PPC-associated changes in SOC function. We measured adult cardiomyocyte membrane currents using the whole-cell patch-clamp technique, and we evaluated reactive oxygen species (ROS) production and intracellular Ca2+ levels in cardiomyocytes using fluorescent probes. Diazoxide (Dzx) and thapsigargin (Tg) were used to induce PPC and to deplete internal stores of Ca2+, respectively. Ca2+ store depletion generated inward currents with strong rectification, which were suppressed by the SOC blocker GSK-7975-A. These currents were completely abolished by PPC, an effect that could be countered with 5-hydroxydecanoate (5-HD; a selective mitochondrial ATP-sensitive K+ channel blocker), an intracellular mitochondrial energizing solution, or Ni2+ [a blocker of sodium-calcium exchanger (NCX)]. Buffering of ROS and intracellular Ca2+ also prevented PPC effects on SOC currents. Refilling of intracellular stores was largely suppressed by PPC, as determined by measuring intracellular Ca2+ with a fluorescent Ca2+ indicator. These results indicate that influx of Ca2+ through SOCs is inhibited by their ROS and Ca2+-dependent inactivation during PPC and that NCX is a likely source of PPC-inactivating Ca2+. We further showed that NCX associates with Orai1. Down-regulation of SOCs by PPC may play a role in cardioprotection following ischemia-reperfusion.
Collapse
Affiliation(s)
- Raúl Sampieri
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Eridani Fuentes
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Elba D Carrillo
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Ascención Hernández
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - María C García
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| | - Jorge A Sánchez
- Departamento de Farmacología, Centro de Investigación y de Estudios Avanzados del IPN, Mexico City, Mexico
| |
Collapse
|
37
|
Bonzon-Kulichenko E, Camafeita E, López JA, Gómez-Serrano M, Jorge I, Calvo E, Núñez E, Trevisan-Herraz M, Bagwan N, Bárcena JA, Peral B, Vázquez J. Improved integrative analysis of the thiol redox proteome using filter-aided sample preparation. J Proteomics 2019; 214:103624. [PMID: 31874222 DOI: 10.1016/j.jprot.2019.103624] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 12/20/2019] [Accepted: 12/20/2019] [Indexed: 02/08/2023]
Abstract
Changes in the oxidation state of protein Cys residues are involved in cell signalling and play a key role in a variety of pathophysiological states. We had previously developed GELSILOX, an in-gel method that enables the large-scale, parallel analysis of dynamic alterations to the redox state of Cys sites and protein abundance changes. Here we present FASILOX, a further development of the GELSILOX approach featuring: i) significantly increased peptide recovery, ii) enhanced sensitivity for the detection of Cys oxidative alterations, and iii) streamlined workflow that results in shortened assay duration. In mitochondria isolated from the adipose tissue of obese, diabetic patients, FASILOX revealed a sexually dimorphic trait of Cys oxidation involving mainly mitochondrial oxidative phosphorylation complexes. These results provide the first evidence for a decreased efficiency in the antioxidant response of men as compared to women.
Collapse
Affiliation(s)
- Elena Bonzon-Kulichenko
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Emilio Camafeita
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain.
| | - Juan Antonio López
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - María Gómez-Serrano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain; Instituto de Investigaciones Biomédicas, Alberto Sols, (IIBM), Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Inmaculada Jorge
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Enrique Calvo
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Estefanía Núñez
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Marco Trevisan-Herraz
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Navratan Bagwan
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - José Antonio Bárcena
- Dept. Biochemistry and Molecular Biology, University of Córdoba, Córdoba, Spain; Maimónides Biomedical Research Institute of Córdoba (IMIBIC), Córdoba, Spain
| | - Belén Peral
- Instituto de Investigaciones Biomédicas, Alberto Sols, (IIBM), Consejo Superior de Investigaciones Científicas & Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Jesús Vázquez
- Cardiovascular Proteomics Laboratory, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
38
|
Prokudina E, Naryzhnaya N, Mukhomedzyanov A, Gorbunov A, Zhang Y, Jaggi A, Tsibulnikov S, Nesterov E, Lishmanov Y, Suleiman M, Oeltgen P, Maslov L. Effect of Chronic Continuous Normobaric Hypoxia on Functional State of Cardiac Mitochondria and Tolerance of Isolated Rat Heart to Ischemia and Reperfusion: Role of µ and δ2 Opioid Receptors. Physiol Res 2019; 68:909-920. [DOI: 10.33549/physiolres.933945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Chronic continuous normobaric hypoxia (CNH) increases cardiac tolerance to ischemia/reperfusion injury in vivo and this effect is mediated via µ and δ2 opioid receptors (ORs) activation. CNH has also been shown to be cardioprotective in isolated rat heart. In this study, we hypothesize that this cardioprotective effect of CNH is mediated by activation of µ and δ2 ORs and preservation of mitochondrial function. Hearts from rats adapted to CNH (12 % oxygen) for 3 weeks were extracted, perfused in the Langendorff mode and subjected to 45 min of global ischemia and 30 min of reperfusion. Intervention groups were pretreated for 10 min with antagonists for different OR types: naloxone (300 nmol/l), the selective δ OR antagonist TIPP(ψ) (30 nmol/l), the selective δ1 OR antagonist BNTX (1 nmol/l), the selective δ2 OR antagonist naltriben (1 nmol/l), the selective peptide μ OR antagonist CTAP (100 nmol/l) and the selective κ OR antagonist nor-binaltorphimine (3 nmol/l). Creatine kinase activity in coronary effluent and cardiac contractile function were monitored to assess cardiac injury and functional impairment. Additionally, cardiac tissue was collected to measure ATP and to isolate mitochondria to measure respiration rate and calcium retention capacity. Adaptation to CNH decreased myocardial creatine kinase release during reperfusion and improved the postischemic recovery of contractile function. Additionally, CNH improved mitochondrial state 3 and uncoupled respiration rates, ADP/O, mitochondrial transmembrane potential and calcium retention capacity and myocardial ATP level during reperfusion compared to the normoxic group. These protective effects were completely abolished by naloxone, TIPP(ψ), naltriben, CTAP but not BNTX or nor-binaltorphimine. These results suggest that cardioprotection associated with adaptation to CNH is mediated by µ and δ2 opioid receptors activation and preservation of mitochondrial function.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - L.N. Maslov
- Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Centre, Russian Academy of Sciences, Tomsk, Russia.
| |
Collapse
|
39
|
Soares ROS, Losada DM, Jordani MC, Évora P, Castro-E-Silva O. Ischemia/Reperfusion Injury Revisited: An Overview of the Latest Pharmacological Strategies. Int J Mol Sci 2019; 20:ijms20205034. [PMID: 31614478 PMCID: PMC6834141 DOI: 10.3390/ijms20205034] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 10/03/2019] [Accepted: 10/08/2019] [Indexed: 02/08/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) permeates a variety of diseases and is a ubiquitous concern in every transplantation proceeding, from whole organs to modest grafts. Given its significance, efforts to evade the damaging effects of both ischemia and reperfusion are abundant in the literature and they consist of several strategies, such as applying pre-ischemic conditioning protocols, improving protection from preservation solutions, thus providing extended cold ischemia time and so on. In this review, we describe many of the latest pharmacological approaches that have been proven effective against IRI, while also revisiting well-established concepts and presenting recent pathophysiological findings in this ever-expanding field. A plethora of promising protocols has emerged in the last few years. They have been showing exciting results regarding protection against IRI by employing drugs that engage several strategies, such as modulating cell-surviving pathways, evading oxidative damage, physically protecting cell membrane integrity, and enhancing cell energetics.
Collapse
Affiliation(s)
| | - Daniele M Losada
- Department of Anatomic Pathology, Faculty of Medical Sciences, University of Campinas, 13083-970 Campinas, Brazil.
| | - Maria C Jordani
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
| | - Paulo Évora
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
- Department of Gastroenterology, São Paulo Medical School, University of São Paulo, 01246-903 São Paulo, Brazil.
| | - Orlando Castro-E-Silva
- Department of Surgery & Anatomy, Ribeirão Preto Medical School, University of São Paulo, 14049-900 Ribeirão Preto, Brazil.
- Department of Gastroenterology, São Paulo Medical School, University of São Paulo, 01246-903 São Paulo, Brazil.
| |
Collapse
|
40
|
Recent Advances in Pharmacological and Non-Pharmacological Strategies of Cardioprotection. Int J Mol Sci 2019; 20:ijms20164002. [PMID: 31426434 PMCID: PMC6720817 DOI: 10.3390/ijms20164002] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/30/2019] [Accepted: 07/30/2019] [Indexed: 02/08/2023] Open
Abstract
Ischemic heart diseases (IHD) are the leading cause of death worldwide. Although the principal form of treatment of IHD is myocardial reperfusion, the recovery of coronary blood flow after ischemia can cause severe and fatal cardiac dysfunctions, mainly due to the abrupt entry of oxygen and ionic deregulation in cardiac cells. The ability of these cells to protect themselves against injury including ischemia and reperfusion (I/R), has been termed “cardioprotection”. This protective response can be stimulated by pharmacological agents (adenosine, catecholamines and others) and non-pharmacological procedures (conditioning, hypoxia and others). Several intracellular signaling pathways mediated by chemical messengers (enzymes, protein kinases, transcription factors and others) and cytoplasmic organelles (mitochondria, sarcoplasmic reticulum, nucleus and sarcolemma) are involved in cardioprotective responses. Therefore, advancement in understanding the cellular and molecular mechanisms involved in the cardioprotective response can lead to the development of new pharmacological and non-pharmacological strategies for cardioprotection, thus contributing to increasing the efficacy of IHD treatment. In this work, we analyze the recent advances in pharmacological and non-pharmacological strategies of cardioprotection.
Collapse
|
41
|
Chen Q, Thompson J, Hu Y, Dean J, Lesnefsky EJ. Inhibition of the ubiquitous calpains protects complex I activity and enables improved mitophagy in the heart following ischemia-reperfusion. Am J Physiol Cell Physiol 2019; 317:C910-C921. [PMID: 31411917 DOI: 10.1152/ajpcell.00190.2019] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Activation of calpain 1 (CPN1) and calpain 2 (CPN2) contributes to cardiac injury during ischemia (ISC) and reperfusion (REP). Complex I activity is decreased in heart mitochondria following ISC-REP. CPN1 and CPN2 are ubiquitous calpains that exist in both cytosol (cs)-CPN1 and 2 and mitochondria (mit)-CPN1 and 2. Recent work shows that the complex I subunit (NDUFS7) is a potential substrate of the mit-CPN1. We asked whether ISC-REP led to decreased complex I activity via proteolysis of the NDUFS7 subunit via activation of mit-CPN1 and -2. Activation of cs-CPN1 and -2 decreases mitophagy in hepatocytes following ISC-REP. We asked whether activation of cs-CPN1 and -2 impaired mitophagy in the heart following ISC-REP. Buffer-perfused rat hearts underwent 25 min of global ISC and 30 min of REP. MDL-28170 (MDL; 10 µM) was used to inhibit CPN1 and -2. Cytosol, subsarcolemmal mitochondria (SSM), and interfibrillar mitochondria (IFM) were isolated at the end of heart perfusion. Cardiac ISC-REP led to decreased complex I activity with a decrease in the content of NDUFS7 in both SSM and IFM. ISC-REP also resulted in a decrease in cytosolic beclin-1 content, a key component of the autophagy pathway required to form autophagosomes. MDL treatment protected the contents of cytosolic beclin-1 and mitochondrial NDUFS7 in hearts following ISC-REP. These results support that activation of both cytosolic and mitochondrial calpains impairs mitochondria during cardiac ISC-REP. Mitochondria-localized calpains impair complex I via cleavage of a key subunit. Activation of cytosolic calpains contributes to mitochondrial dysfunction by impairing removal of the impaired mitochondria through depletion of a key component of the mitophagy process.
Collapse
Affiliation(s)
- Qun Chen
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Jeremy Thompson
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Ying Hu
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Joseph Dean
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia
| | - Edward J Lesnefsky
- Department of Medicine, Division of Cardiology, Virginia Commonwealth University, Richmond, Virginia.,Department of Biochemistry and Molecular Biology Virginia Commonwealth University, Richmond, Virginia.,Department of Physiology and Biophysics, Virginia Commonwealth University, Richmond, Virginia.,McGuire Department of Veterans Affairs Medical Center, Richmond, Virginia
| |
Collapse
|
42
|
STIM2 knockdown protects against ischemia/reperfusion injury through reducing mitochondrial calcium overload and preserving mitochondrial function. Life Sci 2019; 247:116560. [PMID: 31200000 DOI: 10.1016/j.lfs.2019.116560] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2019] [Revised: 06/04/2019] [Accepted: 06/10/2019] [Indexed: 01/03/2023]
Abstract
Mitochondrial dysfunction caused by calcium overload is a vital factor for mediating cardiomyocyte death following ischemia/reperfusion (I/R) injury. The stromal interactive molecule 2 (STIM2) is a calcium sensor protein that regulates the store-operated calcium entry (SOCE). Whereas, whether STIM2 is associated with I/R injury remains largely unclear. We report here that STIM2, but not its homologue STIM1, is upregulated in cultured H9c2 cells, a cell model for cardiomyocytes, following I/R injury. In addition, the knockdown of STIM2, but not STIM1, reduces H9c2 cell apoptosis following I/R injury, and similar results were obtained in primary neonatal cardiomyocytes. This anti-apoptotic effect could be attributed to the inhibited activation of mitochondrial apoptosis pathway. Moreover, STIM2 knockdown reduces ER calcium release and simultaneously alleviates mitochondrial calcium overload in H9c2 cells following I/R injury. Furthermore, STIM2 knockdown decreases mitochondrial injury and preserves mitochondrial function following I/R injury. Collectively, these results suggest that the protective role of STIM2 knockdown against I/R injury in cardiomyocytes is associated with the reduced mitochondrial calcium overload and preserved mitochondrial function. Hence, our study may provide a novel insight into the regulation of mitochondrial-mediated cardiomyocyte apoptosis following I/R injury.
Collapse
|
43
|
Abstract
Liposomes have the potential to be used for drug delivery. Meanwhile, liposome size may affect their accumulation in the target tissue. We investigated the myocardial accumulation of 2 populations of liposomes (∼70 and 110 nm diameter) during ischemia and their effect on ischemia/reperfusion injury. Isolated rat hearts were subjected to 30 minutes of low-flow ischemia with the liposomes, followed by 30 minutes of liposome-free reperfusion. The liposomes were loaded with the fluorescent dye Nile Red to assess their accumulation in the myocardium. The cardiac functional recovery during reperfusion was evaluated using force–velocity characteristics and coronary flow (CF). Reperfusion injury was evaluated by lactate dehydrogenase release. In addition, CF and contractility were assessed in hearts perfused normally with 70 nm liposomes. There was a 6- and 4-fold greater accumulation of the small liposomes in the myocardium and mitochondria, respectively, compared with the large liposomes. Importantly, even without any incorporated drugs, both populations of liposomes improved functional recovery and reduced lactate dehydrogenase release. However, the smaller liposomes showed significantly higher protective and vasodilatory effects during reperfusion than the larger particles. These liposomes also increased CF and contractility during normal perfusion. We suggest that the protective properties of the liposomes could be related to their membrane-stabilizing effect.
Collapse
|
44
|
Bodo S, Campagne C, Thin TH, Higginson DS, Vargas HA, Hua G, Fuller JD, Ackerstaff E, Russell J, Zhang Z, Klingler S, Cho H, Kaag MG, Mazaheri Y, Rimner A, Manova-Todorova K, Epel B, Zatcky J, Cleary CR, Rao SS, Yamada Y, Zelefsky MJ, Halpern HJ, Koutcher JA, Cordon-Cardo C, Greco C, Haimovitz-Friedman A, Sala E, Powell SN, Kolesnick R, Fuks Z. Single-dose radiotherapy disables tumor cell homologous recombination via ischemia/reperfusion injury. J Clin Invest 2019; 129:786-801. [PMID: 30480549 PMCID: PMC6355243 DOI: 10.1172/jci97631] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 11/20/2018] [Indexed: 12/20/2022] Open
Abstract
Tumor cure with conventional fractionated radiotherapy is 65%, dependent on tumor cell-autonomous gradual buildup of DNA double-strand break (DSB) misrepair. Here we report that single-dose radiotherapy (SDRT), a disruptive technique that ablates more than 90% of human cancers, operates a distinct dual-target mechanism, linking acid sphingomyelinase-mediated (ASMase-mediated) microvascular perfusion defects to DNA unrepair in tumor cells to confer tumor cell lethality. ASMase-mediated microcirculatory vasoconstriction after SDRT conferred an ischemic stress response within parenchymal tumor cells, with ROS triggering the evolutionarily conserved SUMO stress response, specifically depleting chromatin-associated free SUMO3. Whereas SUMO3, but not SUMO2, was indispensable for homology-directed repair (HDR) of DSBs, HDR loss of function after SDRT yielded DSB unrepair, chromosomal aberrations, and tumor clonogen demise. Vasoconstriction blockade with the endothelin-1 inhibitor BQ-123, or ROS scavenging after SDRT using peroxiredoxin-6 overexpression or the SOD mimetic tempol, prevented chromatin SUMO3 depletion, HDR loss of function, and SDRT tumor ablation. We also provide evidence of mouse-to-human translation of this biology in a randomized clinical trial, showing that 24 Gy SDRT, but not 3×9 Gy fractionation, coupled early tumor ischemia/reperfusion to human cancer ablation. The SDRT biology provides opportunities for mechanism-based selective tumor radiosensitization via accessing of SDRT/ASMase signaling, as current studies indicate that this pathway is tractable to pharmacologic intervention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Katia Manova-Todorova
- Laboratory of Molecular Cytology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Boris Epel
- Department of Radiation and Cellular Oncology, Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, Illinois, USA
| | | | | | | | | | | | - Howard J. Halpern
- Department of Radiation and Cellular Oncology, Center for EPR Imaging In Vivo Physiology, The University of Chicago, Chicago, Illinois, USA
| | | | - Carlos Cordon-Cardo
- Department of Pathology, Mount Sinai School of Medicine, New York, New York, USA
| | | | | | | | | | | | - Zvi Fuks
- Department of Radiation Oncology
- Champalimaud Centre, Lisbon, Portugal
| |
Collapse
|
45
|
Gorbunov AS, Maslov LN, Jaggi AS, Singh N, De Petrocellis L, Boshchenko AA, Roohbakhsh A, Bezuglov VV, Oeltgen PR. Physiological and Pathological Role of TRPV1, TRPV2 and TRPV4 Channels in Heart. Curr Cardiol Rev 2019; 15:244-251. [PMID: 30848206 PMCID: PMC8142357 DOI: 10.2174/1573403x15666190307112326] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 02/15/2019] [Accepted: 02/26/2019] [Indexed: 12/14/2022] Open
Abstract
Transient receptor potential vanilloid channel 2 (TRPV2) is required for normal cardiac contractility. The stimulation of TRPV1 in isolated cardiomyocytes can aggravate the effect of hypoxia/ reoxygenation (H/R) on H9C2 cells. The knockout of the TRPV1 gene promotes increased tolerance of the isolated perfused heart to the impact of ischemia/reperfusion (I/R). However, activation of TRPV1 increases the resistance of the heart to I/R due to calcitonin gene-related peptide (CGRP) release from afferent nerve endings. It has been established that TRPV1 and TRPV2 are involved in the pathogenesis of myocardial infarction and, in all likelihood, ensure the cardiac tolerance to the ischemia/reperfusion. It has also been documented that the activation of TRPV4 negatively affects the stability of cardiomyocytes to the H/R. The blockade of TRPV4 can be considered as a new approach to the prevention of I/R injury of the heart. Studies also indicate that TRPV1 is involved in the pathogenesis of cardiac hypertrophy and that TRPV2 channels participate in the pathogenesis of dilated cardiomyopathy. Excessive expression of TRPV2 leads to chronic Ca2+- overload of cardiomyocytes, which may contribute to the development of cardiomyopathy.
Collapse
Affiliation(s)
| | - Leonid N. Maslov
- Address correspondence to this author at the Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center of the Russian Academy of Science, Kyevskaya 111A, 634012 Tomsk, Russia; Tel. +7 3822 262174; E-mail:
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Mitoproteomics: Tackling Mitochondrial Dysfunction in Human Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:1435934. [PMID: 30533169 PMCID: PMC6250043 DOI: 10.1155/2018/1435934] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 08/29/2018] [Indexed: 12/11/2022]
Abstract
Mitochondria are highly dynamic and regulated organelles that historically have been defined based on their crucial role in cell metabolism. However, they are implicated in a variety of other important functions, making mitochondrial dysfunction an important axis in several pathological contexts. Despite that conventional biochemical and molecular biology approaches have provided significant insight into mitochondrial functionality, innovative techniques that provide a global view of the mitochondrion are still necessary. Proteomics fulfils this need by enabling accurate, systems-wide quantitative analysis of protein abundance. More importantly, redox proteomics approaches offer unique opportunities to tackle oxidative stress, a phenomenon that is intimately linked to aging, cardiovascular disease, and cancer. In addition, cutting-edge proteomics approaches reveal how proteins exert their functions in complex interaction networks where even subtle alterations stemming from early pathological states can be monitored. Here, we describe the proteomics approaches that will help to deepen the role of mitochondria in health and disease by assessing not only changes to mitochondrial protein composition but also alterations to their redox state and how protein interaction networks regulate mitochondrial function and dynamics. This review is aimed at showing the reader how the application of proteomics approaches during the last 20 years has revealed crucial mitochondrial roles in the context of aging, neurodegenerative disorders, metabolic disease, and cancer.
Collapse
|
47
|
Basheer WA, Fu Y, Shimura D, Xiao S, Agvanian S, Hernandez DM, Hitzeman TC, Hong T, Shaw RM. Stress response protein GJA1-20k promotes mitochondrial biogenesis, metabolic quiescence, and cardioprotection against ischemia/reperfusion injury. JCI Insight 2018; 3:121900. [PMID: 30333316 DOI: 10.1172/jci.insight.121900] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/11/2018] [Indexed: 01/23/2023] Open
Abstract
Connexin 43 (Cx43), a product of the GJA1 gene, is a gap junction protein facilitating intercellular communication between cardiomyocytes. Cx43 protects the heart from ischemic injury by mechanisms that are not well understood. GJA1 mRNA can undergo alternative translation, generating smaller isoforms in the heart, with GJA1-20k being the most abundant. Here, we report that ischemic and ischemia/reperfusion (I/R) injuries upregulate endogenous GJA1-20k protein in the heart, which targets to cardiac mitochondria and associates with the outer mitochondrial membrane. Exploring the functional consequence of increased GJA1-20k, we found that AAV9-mediated gene transfer of GJA1-20k in mouse hearts increases mitochondrial biogenesis while reducing mitochondrial membrane potential, respiration, and ROS production. By doing so, GJA1-20k promotes a protective mitochondrial phenotype, as seen with ischemic preconditioning (IPC), which also increases endogenous GJA1-20k in heart lysates and mitochondrial fractions. As a result, AAV9-GJA1-20k pretreatment reduces myocardial infarct size in mouse hearts subjected to in vivo ischemic injury or ex vivo I/R injury, similar to an IPC-induced cardioprotective effect. In conclusion, GJA1-20k is an endogenous stress response protein that induces mitochondrial biogenesis and metabolic hibernation, preconditioning the heart against I/R insults. Introduction of exogenous GJA1-20k is a putative therapeutic strategy for patients undergoing anticipated ischemic injury.
Collapse
Affiliation(s)
- Wassim A Basheer
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Ying Fu
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Daisuke Shimura
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Shaohua Xiao
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Sosse Agvanian
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Diana M Hernandez
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tara C Hitzeman
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - TingTing Hong
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, California
| | - Robin M Shaw
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA.,Department of Medicine, Cedars-Sinai Medical Center and UCLA, Los Angeles, California
| |
Collapse
|
48
|
Tavares JGP, Errante PR, Govato TCP, Vasques ÊR, Ferraz RRN, Taha MO, Menezes-Rodrigues FS, Caricati-Neto A. Cardioprotective effect of preconditioning is more efficient than postconditioning in rats submitted to cardiac ischemia and reperfusion1. Acta Cir Bras 2018; 33:588-596. [PMID: 30110060 DOI: 10.1590/s0102-865020180070000004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/24/2018] [Indexed: 01/19/2023] Open
Abstract
PURPOSE To investigate the cardioprotective effects of ischemic preconditioning (preIC) and postconditioning (postIC) in animal model of cardiac ischemia/reperfusion. METHODS Adult rats were submitted to protocol of cardiac ischemia/reperfusion (I/R) and randomized into three experimental groups: cardiac I/R (n=33), preCI + cardiac I/R (n=7) and postCI + cardiac I/R (n=8). After this I/R protocol, the incidence of ventricular arrhythmia (VA), atrioventricular block (AVB) and lethality (LET) was evaluated using the electrocardiogram (ECG) analysis. RESULTS After reestablishment of coronary blood flow, we observed variations of the ECG trace with increased incidence of ventricular arrhythmia (VA) (85%), atrioventricular block (AVB) (79%), and increase of lethality (70%) in cardiac I/R group. The comparison between I/R + preIC group with I/R group demonstrated significant reduction in VA incidence to 28%, AVB to 0% and lethality to 14%. The comparison of I/R + postIC group with I/R group was observed significance reduction in AVB incidence to 25% and lethality to 25%. CONCLUSION The preconditioning strategies produce cardioprotection more efficient that postconditioning against myocardial dysfunctions and lethality by cardiac ischemia and reperfusion.
Collapse
Affiliation(s)
- José Gustavo Padrão Tavares
- Fellow PhD degree, Postgraduate Program in Pharmacology, Universidade Federal de São Paulo (UNIFESP), Brazil. Conception and design of the study, analysis and interpretation of data, manuscript writing
| | - Paolo Ruggero Errante
- Fellow PhD degree, Postgraduate Program in Pharmacology, UNIFESP, Sao Paulo-SP, Brazil. Analysis and interpretation of data, manuscript writing
| | - Tânia Carmem Peñaranda Govato
- Assistant Professor, Department of Pharmacology, Faculdade de Medicina do ABC (FMABC), Santo Andre-SP, Brazil. Statistical analysis
| | - Ênio Rodrigues Vasques
- Fellow PhD degree, Department of Gastroenterology, Faculty of Medicine, Universidade de São Paulo (USP), Brazil. Interpretation of electrocardiogram
| | - Renato Ribeiro Nogueira Ferraz
- Full Professor, Program in Management of Health System (PMPA-GSS), Universidade Nove de Julho (UNINOVE), Sao Paulo-SP, Brazil. Critical revision
| | - Murched Omar Taha
- Associate Professor, Department of Surgery, UNIFESP, Sao Paulo-SP, Brazil. Technical procedures
| | | | - Afonso Caricati-Neto
- Associate Professor, Department of Pharmacology, UNIFESP, Sao Paulo-SP, Brazil. Conception and design of the study, critical revision
| |
Collapse
|
49
|
He X, Zeng H, Chen JX. Emerging role of SIRT3 in endothelial metabolism, angiogenesis, and cardiovascular disease. J Cell Physiol 2018; 234:2252-2265. [PMID: 30132870 DOI: 10.1002/jcp.27200] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Accepted: 07/12/2018] [Indexed: 12/11/2022]
Abstract
Sirtuin 3 (SIRT3) a mitochondrial enzyme that plays an important role in energy homeostasis, cardiac remodeling, and heart failure (HF). The expression of SIRT3 declines with advanced age, cardiovascular, and metabolic diseases. Accumulating evidence suggests that SIRT3 plays a critical role in protecting the heart from cardiac hypertrophy, cardiac dysfunction associated with HF, and in the protection of cardiac cells from stress-mediated cell death. Clinical studies have demonstrated that HF with preserved ejection fraction (HFpEF) in patients present with abnormalities in coronary microcirculation related to endothelial dysfunction and coronary microvascular rarefaction. Although SIRT3-mediated regulation of mitochondrial homeostasis and heart function has been intensively investigated, the effect of SIRT3 on endothelial cell (EC) glycolytic metabolism and microvascular function has not been well studied. ECs utilize glycolysis for generating ATP rather than oxidative phosphorylation to maintain their normal functions and promote angiogenesis and EC-cardiomyocyte interactions. Emerging evidence indicates that SIRT3 is involved in the regulation of endothelial metabolism and angiogenesis and thus affects the development of cardiovascular diseases associated with aging. This review will discuss the current knowledge of SIRT3 and its functional role on endothelial metabolism, cardiac function, and cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaochen He
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Heng Zeng
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| | - Jian-Xiong Chen
- Department of Pharmacology and Toxicology, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
50
|
Soliman AF, Anees LM, Ibrahim DM. Cardioprotective effect of zingerone against oxidative stress, inflammation, and apoptosis induced by cisplatin or gamma radiation in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2018; 391:819-832. [PMID: 29736620 DOI: 10.1007/s00210-018-1506-4] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/17/2018] [Indexed: 01/24/2023]
Abstract
Despite their clinical benefits in cancer treatment, the deleterious effects on heart following chemo/radiotherapy are of increasing importance. Zingerone, a natural polyphenol, possesses multiple biological activities, such as antioxidant and anti-inflammatory. Thus, the current study was designed to assess the potential cardioprotective effects of zingerone against cisplatin or γ-radiation. Zingerone was given by intragastric intubation (25 mg/kg) daily for three successive weeks prior to the induction of cardiotoxicity using a single dose of cisplatin (20 mg/kg, i.p.) or a whole body γ-irradiation at a single dose of 6 Gy. Zingerone pre-treatment significantly reduced the abnormalities in heart histology and the increase in the cardiotoxicity indices, serum lactate dehydrogenase, and creatine kinase-MB activities, as well as plasma cardiac troponin T and B-natriuretic peptide, induced by cisplatin or γ-radiation. Further, zingerone, except for superoxide dismutase, notably ameliorated the state of oxidative stress as evidenced by a significant decrease in malondialdehyde level accompanied with a significant increase in the reduced glutathione content and catalase activity. Additionally, zingerone mitigated the increase in the inflammatory markers including serum level of tumor necrosis factor-alpha, cardiac myeloperoxidase activity, and cyclooxygenase-2 protein expression. Moreover, zingerone alleviated the elevation of caspase-3 gene expression and the prominent nuclear DNA fragmentation and attenuated the decrease in mitochondrial complexes' activities. This study sheds the light on a probable protective role of zingerone as an antioxidant, anti-inflammatory, and antiapoptotic agent against cisplatin- or γ-radiation-induced cardiotoxicity and holds a potential in regard to therapeutic intervention for chemo/radiotherapy mediated cardiac damage.
Collapse
Affiliation(s)
- Ahmed F Soliman
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Lobna M Anees
- Health Radiation Research Department, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Doaa M Ibrahim
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| |
Collapse
|