1
|
Giuliani KTK, Adams BC, Healy HG, Kassianos AJ. Regulated cell death in chronic kidney disease: current evidence and future clinical perspectives. Front Cell Dev Biol 2024; 12:1497460. [PMID: 39544363 PMCID: PMC11560912 DOI: 10.3389/fcell.2024.1497460] [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: 09/17/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024] Open
Abstract
Chronic kidney disease (CKD) is the progressive loss of kidney function/structure over a period of at least 3 months. It is characterised histologically by the triad of cell loss, inflammation and fibrosis. This literature review focuses on the forms of cell death that trigger downstream inflammation and fibrosis, collectively called regulated cell death (RCD) pathways. Discrete forms of RCD have emerged as central mediators of CKD pathology. In particular, pathways of regulated necrosis - including mitochondrial permeability transition pore (mPTP)-mediated necrosis, necroptosis, ferroptosis and pyroptosis - have been shown to mediate kidney pathology directly or through the release of danger signals that trigger a pro-inflammatory response, further amplifying tissue injury in a cellular process called necroinflammation. Despite accumulating evidence in pre-clinical models, no clinical studies have yet targeted these RCD modes in human CKD. The review summarizes recent advances in our understanding of RCD pathways in CKD, looks at inter-relations between the pathways (with the emphasis on propagation of death signals) and the evidence for therapeutic targeting of molecules in the RCD pathways to prevent or treat CKD.
Collapse
Affiliation(s)
- Kurt T. K. Giuliani
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Benjamin C. Adams
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Helen G. Healy
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Andrew J. Kassianos
- Conjoint Internal Medicine Laboratory, Chemical Pathology, Pathology Queensland, Brisbane, QLD, Australia
- Kidney Health Service, Royal Brisbane and Women’s Hospital, Brisbane, QLD, Australia
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| |
Collapse
|
2
|
Wu L, Wang XJ, Luo X, Zhang J, Zhao X, Chen Q. Diabetic peripheral neuropathy based on Schwann cell injury: mechanisms of cell death regulation and therapeutic perspectives. Front Endocrinol (Lausanne) 2024; 15:1427679. [PMID: 39193373 PMCID: PMC11348392 DOI: 10.3389/fendo.2024.1427679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 07/24/2024] [Indexed: 08/29/2024] Open
Abstract
Diabetic peripheral neuropathy (DPN) is a complication of diabetes mellitus that lacks specific treatment, its high prevalence and disabling neuropathic pain greatly affects patients' physical and mental health. Schwann cells (SCs) are the major glial cells of the peripheral nervous system, which play an important role in various inflammatory and metabolic neuropathies by providing nutritional support, wrapping axons and promoting repair and regeneration. Increasingly, high glucose (HG) has been found to promote the progression of DPN pathogenesis by targeting SCs death regulation, thus revealing the specific molecular process of programmed cell death (PCD) in which SCs are disrupted is an important link to gain insight into the pathogenesis of DPN. This paper is the first to review the recent progress of HG studies on apoptosis, autophagy, pyroptosis, ferroptosis and necroptosis pathways in SCs, and points out the crosstalk between various PCDs and the related therapeutic perspectives, with the aim of providing new perspectives for a deeper understanding of the mechanisms of DPN and the exploration of effective therapeutic targets.
Collapse
Affiliation(s)
- Lijiao Wu
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiang Jin Wang
- School of Sports Medicine and Health, Chengdu Sports University, Chengdu, China
| | - Xi Luo
- Jiangsu Key Laboratory for Functional Substance of Chinese Medicine, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingqi Zhang
- School of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xinyi Zhao
- College of lntegrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Hunan, China
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
3
|
Rudokas MW, McKay M, Toksoy Z, Eisen JN, Bögner M, Young LH, Akar FG. Mitochondrial network remodeling of the diabetic heart: implications to ischemia related cardiac dysfunction. Cardiovasc Diabetol 2024; 23:261. [PMID: 39026280 PMCID: PMC11264840 DOI: 10.1186/s12933-024-02357-1] [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: 01/17/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024] Open
Abstract
Mitochondria play a central role in cellular energy metabolism, and their dysfunction is increasingly recognized as a critical factor in the pathogenesis of diabetes-related cardiac pathophysiology, including vulnerability to ischemic events that culminate in myocardial infarction on the one hand and ventricular arrhythmias on the other. In diabetes, hyperglycemia and altered metabolic substrates lead to excessive production of reactive oxygen species (ROS) by mitochondria, initiating a cascade of oxidative stress that damages mitochondrial DNA, proteins, and lipids. This mitochondrial injury compromises the efficiency of oxidative phosphorylation, leading to impaired ATP production. The resulting energy deficit and oxidative damage contribute to functional abnormalities in cardiac cells, placing the heart at an increased risk of electromechanical dysfunction and irreversible cell death in response to ischemic insults. While cardiac mitochondria are often considered to be relatively autonomous entities in their capacity to produce energy and ROS, their highly dynamic nature within an elaborate network of closely-coupled organelles that occupies 30-40% of the cardiomyocyte volume is fundamental to their ability to exert intricate regulation over global cardiac function. In this article, we review evidence linking the dynamic properties of the mitochondrial network to overall cardiac function and its response to injury. We then highlight select studies linking mitochondrial ultrastructural alterations driven by changes in mitochondrial fission, fusion and mitophagy in promoting cardiac ischemic injury to the diabetic heart.
Collapse
Affiliation(s)
- Michael W Rudokas
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Margaret McKay
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Biomedical Engineering, Yale University Schools of Engineering and Applied Sciences, New Haven, CT, USA
| | - Zeren Toksoy
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Julia N Eisen
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Markus Bögner
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Lawrence H Young
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Fadi G Akar
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale School of Medicine, New Haven, CT, USA.
- Department of Biomedical Engineering, Yale University Schools of Engineering and Applied Sciences, New Haven, CT, USA.
- Department of Biomedical Engineering, Electro-biology and Arrhythmia Therapeutics Laboratory, Yale University Schools of Medicine, Engineering and Applied Sciences, 300 George Street, 793 - 748C, New Haven, CT, 06511, USA.
| |
Collapse
|
4
|
Mendoza A, Patel P, Robichaux D, Ramirez D, Karch J. Inhibition of the mPTP and Lipid Peroxidation Is Additively Protective Against I/R Injury. Circ Res 2024; 134:1292-1305. [PMID: 38618716 PMCID: PMC11081482 DOI: 10.1161/circresaha.123.323882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/16/2024]
Abstract
BACKGROUND During myocardial ischemia/reperfusion (I/R) injury, high levels of matrix Ca2+ and reactive oxygen species (ROS) induce the opening of the mitochondrial permeability transition pore (mPTP), which causes mitochondrial dysfunction and ultimately necrotic death. However, the mechanisms of how these triggers individually or cooperatively open the pore have yet to be determined. METHODS Here, we use a combination of isolated mitochondrial assays and in vivo I/R surgery in mice. We challenged isolated liver and heart mitochondria with Ca2+, ROS, and Fe2+ to induce mitochondrial swelling. Using inhibitors of the mPTP (cyclosporine A or ADP) lipid peroxidation (ferrostatin-1, MitoQ), we determined how the triggers elicit mitochondrial damage. Additionally, we used the combination of inhibitors during I/R injury in mice to determine if dual inhibition of these pathways is additivity protective. RESULTS In the absence of Ca2+, we determined that ROS fails to trigger mPTP opening. Instead, high levels of ROS induce mitochondrial dysfunction and rupture independently of the mPTP through lipid peroxidation. As expected, Ca2+ in the absence of ROS induces mPTP-dependent mitochondrial swelling. Subtoxic levels of ROS and Ca2+ synergize to induce mPTP opening. Furthermore, this synergistic form of Ca2+- and ROS-induced mPTP opening persists in the absence of CypD (cyclophilin D), suggesting the existence of a CypD-independent mechanism for ROS sensitization of the mPTP. These ex vivo findings suggest that mitochondrial dysfunction may be achieved by multiple means during I/R injury. We determined that dual inhibition of the mPTP and lipid peroxidation is significantly more protective against I/R injury than individually targeting either pathway alone. CONCLUSIONS In the present study, we have investigated the relationship between Ca2+ and ROS, and how they individually or synergistically induce mitochondrial swelling. Our findings suggest that Ca2+ mediates mitochondrial damage through the opening of the mPTP, although ROS mediates its damaging effects through lipid peroxidation. However, subtoxic levels both Ca2+ and ROS can induce mPTP-mediated mitochondrial damage. Targeting both of these triggers to preserve mitochondria viability unveils a highly effective therapeutic approach for mitigating I/R injury.
Collapse
Affiliation(s)
- Arielys Mendoza
- Department of Integrative Physiology (A.M., P.P., D.R., D.R., J.K.), Baylor College of Medicine, Houston TX
| | - Pooja Patel
- Department of Integrative Physiology (A.M., P.P., D.R., D.R., J.K.), Baylor College of Medicine, Houston TX
| | - Dexter Robichaux
- Department of Integrative Physiology (A.M., P.P., D.R., D.R., J.K.), Baylor College of Medicine, Houston TX
| | - Daniel Ramirez
- Department of Integrative Physiology (A.M., P.P., D.R., D.R., J.K.), Baylor College of Medicine, Houston TX
| | - Jason Karch
- Department of Integrative Physiology (A.M., P.P., D.R., D.R., J.K.), Baylor College of Medicine, Houston TX
- the Cardiovascular Research Institute (J.K.), Baylor College of Medicine, Houston TX
| |
Collapse
|
5
|
Huang C, Zhang H, Yang Y, Liu H, Chen J, Wang Y, Liang L, Hu H, Liu Y. Synthesis, characterization, molecular docking, RNA-sequence and anticancer efficacy evaluation in vitro of ruthenium(II) complexes on B16 cells. J Inorg Biochem 2023; 247:112329. [PMID: 37478780 DOI: 10.1016/j.jinorgbio.2023.112329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 07/10/2023] [Accepted: 07/11/2023] [Indexed: 07/23/2023]
Abstract
In recent years, the studies of the ruthenium(II) complexes on anticancer activity have been paid great attention, many Ru(II) complexes possess high anticancer efficiency. In this paper, three ligands CPIP (2-(4-chlorophenyl)-1H-imidazo[4,5-f][1,10]phenanthroline), DCPIP (2-(3,4-dichlorophenyl)-1H-imidazo[4,5-f][1,10]phenanthroline), TCPIP (2-(2,3,5-trichlorophenyl)-1H-imidazo[4,5-f][1,10]phenanthroline) and their three ruthenium (II) complexes [Ru(dip)2(CPIP)](PF6)2 (1, dip = 4,7-diphenyl-1,10-phenanthroline), [Ru(dip)2(DCPIP)](PF6)2 (2) and [Ru(dip)2(TCPIP)](PF6)2 (3) were synthesized and characterized. 3-(4,5-dimethylthiazole-2-yl)-2,5-biphenyl tetrazolium bromide (MTT) assay was used to investigate in vitro cytotoxicity of complexes against various cancer cells. The results showed that complexes 1-3 exhibited pronounced cytotoxic effect on B16 cells with low IC50 values of 7.2 ± 0.1, 11.7 ± 0.6 and 1.2 ± 0.2 μM, respectively. The 3D model demonstrated that the complexes can validly prevent the cell proliferation. Apoptosis determined using Annexin V-FITC/PI double staining revealed that complexes 1-3 can effectively induce apoptosis in B16 cells. The intracellular localization of 1-3 in the mitochondria, the levels of intracellular reactive oxygen species (ROS), the opening of mitochondrial permeability transition pore as well as the decline of mitochondrial membrane potential were investigated, which demonstrated that the complexes 1-3 led to apoptosis via a ROS-mediated mitochondrial dysfunction pathway. The RNA-sequence indicated that the complexes upregulate the expression of 74 genes and downregulate the expression of 81 genes. The molecular docking showed that the complexes interact with the proteins through hydrogen bond, π-cation and π-π interaction. The results show that ruthenium(II) complexes 1, 2 and 3 can block tumor cell growth and induce cell death through autophagy and ROS-mediated mitochondrial dysfunction pathways.
Collapse
Affiliation(s)
- Chunxia Huang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Huiwen Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yan Yang
- Department of Pharmacy, Guangdong Second Provincial General Hospital, Guangzhou 510317, PR China.
| | - Haimei Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Jing Chen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yi Wang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Lijuan Liang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Huiyan Hu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China
| | - Yunjun Liu
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, PR China; Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topic Precise Drug Delivery System, Guangdong Pharmaceutical University, Guangzhou 510006, PR China.
| |
Collapse
|
6
|
Punetha M, Saini S, Chaudhary S, Bala R, Sharma M, Kumar P, Kumar D, Yadav PS. Mitochondria-targeted antioxidant MitoQ ameliorates ROS production and improves cell viability in cryopreserved buffalo fibroblasts. Tissue Cell 2023; 82:102067. [PMID: 36958101 DOI: 10.1016/j.tice.2023.102067] [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: 09/15/2022] [Revised: 02/15/2023] [Accepted: 03/09/2023] [Indexed: 03/13/2023]
Abstract
Cryopreservation commonly decreases the cellular functionality and post-thaw viability of cells. Reactive oxygen species (ROS) generated during cryopreservation degrade mitochondrial activity and promote the release of cytochrome C which activates caspases required for apoptosis. Antioxidants have the potential to improve the recovery efficiency of cells by reducing ROS production and maintaining mitochondrial membrane potential (MMP). The present study was conducted to explore the role of MitoQ, a derivative of coenzyme Q10 on cryopreserved fibroblasts derived from buffalo skin. To achieve our goal, buffalo skin fibroblasts were treated with varying concentrations of MitoQ (0, 0.1, 0.5, 1, 2, and 10 μM) for 24, 48, and 72 h. The MMP, ROS generation, cell viability was measured by flow cytometry. Furthermore, expression of genes related to mitochondrial oxidative stress (NRF2, GPX, and SOD), apoptosis (BAK and caspase 3) and cell proliferation (AKT) were also assessed. The results showed that over a period of 72 h lower concentrations of MitoQ (0.1-0.5 μM) decrease the ROS production, improves MMP and cell viability whilst the high concentration of MitoQ (2-10 μM) increased the oxidative damage to the cells. Taken together, our study provide important insights into the novel role of MitoQ in cryopreserved buffalo skin fibroblasts. In conclusion, we demonstrated the dose-dependent functional role of MitoQ on cryopreserved fibroblasts for improving post-thaw cell viability and cellular function.
Collapse
Affiliation(s)
- Meeti Punetha
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, Haryana 125001, India
| | - Sheetal Saini
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, Haryana 125001, India
| | - Suman Chaudhary
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, Haryana 125001, India
| | - Renu Bala
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, Haryana 125001, India
| | - Maninder Sharma
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, Haryana 125001, India
| | - Pradeep Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, Haryana 125001, India
| | - Dharmendra Kumar
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, Haryana 125001, India.
| | - P S Yadav
- Animal Physiology and Reproduction Division, ICAR-Central Institute for Research on Buffaloes, Hisar, Haryana 125001, India.
| |
Collapse
|
7
|
Feng Y, Imam Aliagan A, Tombo N, Bopassa JC. Mitofilin Heterozygote Mice Display an Increase in Myocardial Injury and Inflammation after Ischemia/Reperfusion. Antioxidants (Basel) 2023; 12:921. [PMID: 37107296 PMCID: PMC10135852 DOI: 10.3390/antiox12040921] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/17/2023] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Mitochondrial inner membrane protein (Mitofilin/Mic60) is part of a big complex that constituent the mitochondrial inner membrane organizing system (MINOS), which plays a critical role in maintaining mitochondrial architecture and function. We recently showed that Mitofilin physically binds to Cyclophilin D, and disruption of this interaction promotes the opening of mitochondrial permeability transition pore (mPTP) and determines the extent of I/R injury. Here, we investigated whether Mitofilin knockout in the mouse enhances myocardial injury and inflammation after I/R injury. We found that full-body deletion (homozygote) of Mitofilin induces a lethal effect in the offspring and that a single allele expression of Mitofilin is sufficient to rescue the mouse phenotype in normal conditions. Using non-ischemic hearts from wild-type (WT) and Mitofilin+/- (HET) mice, we report that the mitochondria structure and calcium retention capacity (CRC) required to induce the opening of mPTP were similar in both groups. However, the levels of mitochondrial dynamics proteins involved in both fusion/fission, including MFN2, DRP1, and OPA1, were slightly reduced in Mitofilin+/- mice compared to WT. After I/R, the CRC and cardiac functional recovery were reduced while the mitochondria structure was more damaged, and myocardial infarct size was increased in Mitofilin+/- mice compared to WT. Mitofilin+/- mice exhibited an increase in the mtDNA release in the cytosol and ROS production, as well as dysregulated SLC25As (3, 5, 11, and 22) solute carrier function, compared to WT. In addition, Mitofilin+/- mice displayed an increase in the transcript of pro-inflammatory markers, including IL-6, ICAM, and TNF-α. These results suggest that Mitofilin knockdown induces mitochondrial cristae damage that promotes dysregulation of SLC25As solute carriers, leading to an increase in ROS production and reduction in CRC after I/R. These effects are associated with an increase in the mtDNA release into the cytosol, where it activates signaling cascades leading to nuclear transcription of pro-inflammatory cytokines that aggravate I/R injury.
Collapse
Affiliation(s)
| | | | | | - Jean C. Bopassa
- Department of Cellular and Integrative Physiology, School of Medicine, The University of Texas Health Science Center at San Antonio, 7703 Floyd Curl Dr., San Antonio, TX 78229, USA
| |
Collapse
|
8
|
Zhao L, Cheng J, Liu D, Gong H, Bai D, Sun W. Potentilla anserina polysaccharide alleviates cadmium-induced oxidative stress and apoptosis of H9c2 cells by regulating the MG53-mediated RISK pathway. Chin J Nat Med 2023; 21:279-291. [PMID: 37120246 DOI: 10.1016/s1875-5364(23)60436-4] [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: 09/23/2022] [Indexed: 05/01/2023]
Abstract
Oxidative stress plays a crucial role in cadmium (Cd)-induced myocardial injury. Mitsugumin 53 (MG53) and its mediated reperfusion injury salvage kinase (RISK) pathway have been demonstrated to be closely related to myocardial oxidative damage. Potentilla anserina L. polysaccharide (PAP) is a polysaccharide with antioxidant capacity, which exerts protective effect on Cd-induced damage. However, it remains unknown whether PAP can prevent and treat Cd-induced cardiomyocyte damages. The present study was desgined to explore the effect of PAP on Cd-induced damage in H9c2 cells based on MG53 and the mediated RISK pathway. For in vitro evaluation, cell viability and apoptosis rate were analyzed by CCK-8 assay and flow cytometry, respectively. Furthermore, oxidative stress was assessed by 2',7'-dichlorodihydrofluorescein diacetate (DCFH-DA) staining and using superoxide dismutase (SOD), catalase (CAT), and glutathione/oxidized glutathione (GSH/GSSG) kits. The mitochondrial function was measured by JC-10 staining and ATP detection assay. Western blot was performed to detect the expression of proteins related to MG53, the RISK pathway, and apoptosis. The results indicated that Cd increased the levels of reactive oxygen species (ROS) in H9c2 cells. Cd decreased the activities of SOD and CAT and the ratio of GSH/GSSG, resulting in decreases in cell viability and increases in apoptosis. Interestingly, PAP reversed Cd-induced oxidative stress and cell apoptosis. Meanwhile, Cd reduced the expression of MG53 in H9c2 cells and inhibited the RISK pathway, which was mediated by decreasing the ratio of p-AktSer473/Akt, p-GSK3βSer9/GSK3β and p-ERK1/2/ERK1/2. In addition, Cd impaired mitochondrial function, which involved a reduction in ATP content and mitochondrial membrane potential (MMP), and an increase in the ratio of Bax/Bcl-2, cytoplasmic cytochrome c/mitochondrial cytochrome c, and Cleaved-Caspase 3/Pro-Caspase 3. Importantly, PAP alleviated Cd-induced MG53 reduction, activated the RISK pathway, and reduced mitochondrial damage. Interestingly, knockdown of MG53 or inhibition of the RISK pathway attenuated the protective effect of PAP in Cd-induced H9c2 cells. In sum, PAP reduces Cd-induced damage in H9c2 cells, which is mediated by increasing MG53 expression and activating the RISK pathway.
Collapse
Affiliation(s)
- Lixia Zhao
- Institute of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; School of Nursing, Gansu University of Chinese Medicine, Lanzhou 730000, China; Key Laboratory of Dunhuang Medicine, Ministry of Education, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Ju Cheng
- Institute of Genetics, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Di Liu
- Key laboratory of Evidence Science Techniques Research and Application of Gansu Province, Gansu University of Political Science and Law, Lanzhou 730000, China
| | - Hongxia Gong
- School of Basic Medical Sciences, Gansu University of Chinese Medicine, Lanzhou 730000, China
| | - Decheng Bai
- Institute of Integrated Traditional Chinese and Western Medicine, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Wei Sun
- Department of Cardiac Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
9
|
Yu Y, Chen M, Guo Q, Shen L, Liu X, Pan J, Zhang Y, Xu T, Zhang D, Wei G. Human umbilical cord mesenchymal stem cell exosome-derived miR-874-3p targeting RIPK1/PGAM5 attenuates kidney tubular epithelial cell damage. Cell Mol Biol Lett 2023; 28:12. [PMID: 36750776 PMCID: PMC9903493 DOI: 10.1186/s11658-023-00425-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/20/2023] [Indexed: 02/09/2023] Open
Abstract
BACKGROUND Kidney insults due to various pathogenic factors, such as trauma, infection, and inflammation, can cause tubular epithelial cell injury and death, leading to acute kidney injury and the transformation of acute kidney injury to chronic kidney disease. There is no definitive treatment available. In previous studies, human umbilical cord mesenchymal stem cells have been shown to promote kidney injury. In this preclinical study, we investigate the role and mechanism of human umbilical cord mesenchymal stem cell exosomes (HucMSC-Exos) on the repair of renal tubular epithelial cells after injury. METHODS C57BL/6 mice underwent unilateral ureteral obstruction, and epithelial cell injury was induced in HK-2 cells by cisplatin. HucMSC-Exos were assessed in vivo and in vitro. The extent of renal cell injury, activation of necroptosis pathway, and mitochondrial quality-control-related factors were determined in different groups. We also analyzed the possible regulatory effector molecules in HucMSC-Exos by transcriptomics. RESULTS HucMSC-Exo inhibited necroptosis after renal tubular epithelial cell injury and promoted the dephosphorylation of the S637 site of the Drp1 gene by reducing the expression of PGAM5. This subsequently inhibited mitochondrial fission and maintained mitochondrial functional homeostasis, mitigating renal injury and promoting repair. In addition, HucMSC-Exo displayed a regulatory role by targeting RIPK1 through miR-874-3p. CONCLUSION The collective findings of the present study demonstrate that HucMSC-Exos can regulate necroptosis through miR-874-3p to attenuate renal tubular epithelial cell injury and enhance repair, providing new therapeutic modalities and ideas for the treatment of AKI and the process of AKI to CKD transformation to mitigate renal damage.
Collapse
Affiliation(s)
- Yihang Yu
- grid.488412.3Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 China ,grid.419897.a0000 0004 0369 313XChongqing Key Laboratory of Children Urogenital Department and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014 China ,grid.488412.3National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014 China
| | - Meiling Chen
- grid.488412.3Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 China ,grid.419897.a0000 0004 0369 313XChongqing Key Laboratory of Children Urogenital Department and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014 China ,grid.488412.3National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014 China
| | - Qitong Guo
- grid.488412.3Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 China ,grid.419897.a0000 0004 0369 313XChongqing Key Laboratory of Children Urogenital Department and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014 China ,grid.488412.3National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014 China
| | - Lianju Shen
- grid.488412.3Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 China ,grid.419897.a0000 0004 0369 313XChongqing Key Laboratory of Children Urogenital Department and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014 China ,grid.488412.3National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014 China
| | - Xing Liu
- grid.488412.3Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 China ,grid.419897.a0000 0004 0369 313XChongqing Key Laboratory of Children Urogenital Department and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014 China ,grid.488412.3National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014 China
| | - Jianbo Pan
- grid.203458.80000 0000 8653 0555Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, 400016 China
| | - Yuanyuan Zhang
- grid.241167.70000 0001 2185 3318Wake Forest Institute for Regenerative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27101 USA
| | - Tao Xu
- grid.12527.330000 0001 0662 3178Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing, 100084 China
| | - Deying Zhang
- Department of Urology, Children's Hospital of Chongqing Medical University, Chongqing, 400014, China. .,Chongqing Key Laboratory of Children Urogenital Department and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014, China. .,National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014, China.
| | - Guanghui Wei
- grid.488412.3Department of Urology, Children’s Hospital of Chongqing Medical University, Chongqing, 400014 China ,grid.419897.a0000 0004 0369 313XChongqing Key Laboratory of Children Urogenital Department and Tissue Engineering, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing, 400014 China ,grid.488412.3National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing, 400014 China
| |
Collapse
|
10
|
Yalamanchili K, Afzal N, Boyman L, Mannella CA, Lederer WJ, Jafri MS. Understanding the Dynamics of the Transient and Permanent Opening Events of the Mitochondrial Permeability Transition Pore with a Novel Stochastic Model. MEMBRANES 2022; 12:494. [PMID: 35629820 PMCID: PMC9146742 DOI: 10.3390/membranes12050494] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 04/18/2022] [Accepted: 04/29/2022] [Indexed: 02/04/2023]
Abstract
The mitochondrial permeability transition pore (mPTP) is a non-selective pore in the inner mitochondrial membrane (IMM) which causes depolarization when it opens under conditions of oxidative stress and high concentrations of Ca2+. In this study, a stochastic computational model was developed to better understand the dynamics of mPTP opening and closing associated with elevated reactive oxygen species (ROS) in cardiomyocytes. The data modeled are from "photon stress" experiments in which the fluorescent dye TMRM (tetramethylrhodamine methyl ester) is both the source of ROS (induced by laser light) and sensor of the electrical potential difference across the IMM. Monte Carlo methods were applied to describe opening and closing of the pore along with the Hill Equation to account for the effect of ROS levels on the transition probabilities. The amplitude distribution of transient mPTP opening events, the number of transient mPTP opening events per minute in a cell, the time it takes for recovery after transient depolarizations in the mitochondria, and the change in TMRM fluorescence during the transition from transient to permanent mPTP opening events were analyzed. The model suggests that mPTP transient open times have an exponential distribution that are reflected in TMRM fluorescence. A second multiple pore model in which individual channels have no permanent open state suggests that 5-10 mPTP per mitochondria would be needed for sustained mitochondrial depolarization at elevated ROS with at least 1 mPTP in the transient open state.
Collapse
Affiliation(s)
- Keertana Yalamanchili
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA; (K.Y.); (N.A.)
- Thomas Jefferson High School for Science and Technology, Alexandria, VA 22312, USA
| | - Nasrin Afzal
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA; (K.Y.); (N.A.)
| | - Liron Boyman
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (L.B.); (C.A.M.); (W.J.L.)
| | - Carmen A. Mannella
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (L.B.); (C.A.M.); (W.J.L.)
| | - W. Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (L.B.); (C.A.M.); (W.J.L.)
| | - M. Saleet Jafri
- Thomas Jefferson High School for Science and Technology, Alexandria, VA 22312, USA
| |
Collapse
|
11
|
Rodrigues-Ferreira C, Lopes JA, Carneiro PF, Dos Santos Lessa C, Galina A, Vieyra A. Bone Marrow Mononuclear Cells Restore Normal Mitochondrial Ca 2+ Handling and Ca 2+-Induced Depolarization of the Internal Mitochondrial Membrane by Inhibiting the Permeability Transition Pore After Ischemia/Reperfusion. Cell Transplant 2022; 31:9636897221085883. [PMID: 35343271 PMCID: PMC8958683 DOI: 10.1177/09636897221085883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Acute kidney injury due to ischemia followed by reperfusion (IR) is a severe clinical condition with high death rates. IR affects the proximal tubule segments due to their predominantly oxidative metabolism and profoundly altered mitochondrial functions. We previously described the impact of IR on oxygen consumption, the generation of membrane potential (ΔΨ), and formation of reactive oxygen species, together with inflammatory and structural alterations. We also demonstrated the benefits of bone marrow mononuclear cells (BMMC) administration in these alterations. The objective of the present study has been to investigate the effect of IR and the influence of BMMC on the mechanisms of Ca2+ handling in mitochondria of the proximal tubule cells. IR inhibited the rapid accumulation of Ca2+ (Ca2+ green fluorescence assays) and induced the opening of the cyclosporine A-sensitive permeability transition pore (PTP), alterations prevented by BMMC. IR accelerated Ca2+-induced decrease of ΔΨ (Safranin O fluorescence assays), as evidenced by decreased requirement for Ca2+ load and t1/2 for complete depolarization. Addition of BMMC and ADP recovered the normal depolarization profile, suggesting that stabilization of the adenine nucleotide translocase (ANT) in a conformation that inhibits PTP opening offers a partial defense mechanism against IR injury. Moreover, as ANT forms a complex with the voltage-dependent anion channel (VDAC) in the outer mitochondrial membrane, it is possible that this complex is also a target for IR injury—thus favoring Ca2+ release, as well as the supramolecular structure that BMMC protects. These beneficial effects are accompanied by a stimulus of the citric acid cycle—which feed the mitochondrial complexes with the electrons removed from different substrates—as the result of accentuated stimulus of citrate synthase activity by BMMC.
Collapse
Affiliation(s)
- Clara Rodrigues-Ferreira
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Roberto Alcântara Gomes Institute of Biology, Rio de Janeiro State University, Rio de Janeiro, Brazil
| | - Jarlene Alécia Lopes
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Priscila Fonseca Carneiro
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Cristiane Dos Santos Lessa
- Leopoldo de Meis Institute of Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Antonio Galina
- Leopoldo de Meis Institute of Biochemistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Adalberto Vieyra
- Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Center for Structural Biology and Bioimaging, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Graduate Program of Translational Biomedicine, Grande Rio University, Duque de Caxias, Brazil
| |
Collapse
|
12
|
TASK-1 regulates mitochondrial function under hypoxia. Biochem Biophys Res Commun 2021; 578:163-169. [PMID: 34571371 DOI: 10.1016/j.bbrc.2021.09.032] [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] [Received: 09/11/2021] [Accepted: 09/16/2021] [Indexed: 11/23/2022]
Abstract
TASK-1, TWIK-related acid-sensitive potassium channel 1, is a member of the two-pore- domain potassium channel family. It is constitutively active at resting potentials and strongly expressed in the heart. However, little is known about the role of TASK-1 channels in hypoxia. A cellular model of hypoxia and reoxygenation from rat heart-derived H9c2 cells or TASK-1 deficient HEK293T cells was employed to explore the role of TASK-1 channels in cytoprotection against hypoxia. The cell viability assay revealed that TASK-1 expression increased the number of viable cells subjected to 2 h of hypoxia followed by 2 h of reoxygenation (H/R). To dissect the protective role of TASK-1 on mitochondrial function, mitochondrial membrane potential (MMP) was assessed by tetramethylrhodamine fluorescence. It was demonstrated that MMP was significantly decreased by H/R, but it was maintained by TASK-1 expression or pretreatment with cyclosporin A, an inhibitor of mitochondrial permeability transition pore (mPTP). The effect of cyclosporin A on MMP was not further altered by TASK-1 expression. Moreover, TASK-1 expression significantly blocked cytochrome c release induced by H/R. While a small fraction of endogenous TASK-1 was found to colocalize with the mitochondrial marker MitoTracker in H9c2 cells, H/R did not alter the extent of colocalization of TASK-1 with MitoTracker. The total TASK-1 protein level was not significantly affected by H/R. In summary, we provided the evidence that TASK-1 channels confer cytoprotection against hypoxia-reoxygenation injury, possibly by their capacity of maintaining the mitochondrial membrane potential via inhibiting MPTP opening.
Collapse
|
13
|
Morciano G, Naumova N, Koprowski P, Valente S, Sardão VA, Potes Y, Rimessi A, Wieckowski MR, Oliveira PJ. The mitochondrial permeability transition pore: an evolving concept critical for cell life and death. Biol Rev Camb Philos Soc 2021; 96:2489-2521. [PMID: 34155777 DOI: 10.1111/brv.12764] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023]
Abstract
In this review, we summarize current knowledge of perhaps one of the most intriguing phenomena in cell biology: the mitochondrial permeability transition pore (mPTP). This phenomenon, which was initially observed as a sudden loss of inner mitochondrial membrane impermeability caused by excessive calcium, has been studied for almost 50 years, and still no definitive answer has been provided regarding its mechanisms. From its initial consideration as an in vitro artifact to the current notion that the mPTP is a phenomenon with physiological and pathological implications, a long road has been travelled. We here summarize the role of mitochondria in cytosolic calcium control and the evolving concepts regarding the mitochondrial permeability transition (mPT) and the mPTP. We show how the evolving mPTP models and mechanisms, which involve many proposed mitochondrial protein components, have arisen from methodological advances and more complex biological models. We describe how scientific progress and methodological advances have allowed milestone discoveries on mPTP regulation and composition and its recognition as a valid target for drug development and a critical component of mitochondrial biology.
Collapse
Affiliation(s)
- Giampaolo Morciano
- Maria Cecilia Hospital, GVM Care & Research, Via Corriera 1, Cotignola, Ravenna, 48033, Italy.,Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Via Fossato di Mortara 70, Ferrara, 44121, Italy
| | - Natalia Naumova
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua Medical School, Via Giustiniani 2, Padova, 35128, Italy
| | - Piotr Koprowski
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, Warsaw, 02-093, Poland
| | - Sara Valente
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, 3060-197, Portugal
| | - Vilma A Sardão
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, 3060-197, Portugal
| | - Yaiza Potes
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, Warsaw, 02-093, Poland
| | - Alessandro Rimessi
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Via Fossato di Mortara 70, Ferrara, 44121, Italy
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, Warsaw, 02-093, Poland
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, 3060-197, Portugal
| |
Collapse
|
14
|
Shen L, Gan Q, Yang Y, Reis C, Zhang Z, Xu S, Zhang T, Sun C. Mitophagy in Cerebral Ischemia and Ischemia/Reperfusion Injury. Front Aging Neurosci 2021; 13:687246. [PMID: 34168551 PMCID: PMC8217453 DOI: 10.3389/fnagi.2021.687246] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 05/10/2021] [Indexed: 02/03/2023] Open
Abstract
Ischemic stroke is a severe cerebrovascular disease with high mortality and morbidity. In recent years, reperfusion treatments based on thrombolytic and thrombectomy are major managements for ischemic stroke patients, and the recanalization time window has been extended to over 24 h. However, with the extension of the time window, the risk of ischemia/reperfusion (I/R) injury following reperfusion therapy becomes a big challenge for patient outcomes. I/R injury leads to neuronal death due to the imbalance in metabolic supply and demand, which is usually related to mitochondrial dysfunction. Mitophagy is a type of selective autophagy referring to the process of specific autophagic elimination of damaged or dysfunctional mitochondria to prevent the generation of excessive reactive oxygen species (ROS) and the subsequent cell death. Recent advances have implicated the protective role of mitophagy in cerebral ischemia is mainly associated with its neuroprotective effects in I/R injury. This review discusses the involvement of mitochondria dynamics and mitophagy in the pathophysiology of ischemic stroke and I/R injury in particular, focusing on the therapeutic potential of mitophagy regulation and the possibility of using mitophagy-related interventions as an adjunctive approach for neuroprotective time window extension after ischemic stroke.
Collapse
Affiliation(s)
- Luoan Shen
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining, China
| | - Qinyi Gan
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining, China
| | - Youcheng Yang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining, China
| | - Cesar Reis
- VA Loma Linda Healthcare System, Loma Linda University, Loma Linda, CA, United States
| | - Zheng Zhang
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining, China
| | - Shanshan Xu
- Institute for Advanced Study, Shenzhen University, Shenzhen, China
| | - Tongyu Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chengmei Sun
- Zhejiang University-University of Edinburgh Institute, School of Medicine, Zhejiang University, Haining, China.,Institute for Advanced Study, Shenzhen University, Shenzhen, China
| |
Collapse
|
15
|
Misrani A, Tabassum S, Yang L. Mitochondrial Dysfunction and Oxidative Stress in Alzheimer's Disease. Front Aging Neurosci 2021; 13:617588. [PMID: 33679375 PMCID: PMC7930231 DOI: 10.3389/fnagi.2021.617588] [Citation(s) in RCA: 306] [Impact Index Per Article: 76.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
Mitochondria play a pivotal role in bioenergetics and respiratory functions, which are essential for the numerous biochemical processes underpinning cell viability. Mitochondrial morphology changes rapidly in response to external insults and changes in metabolic status via fission and fusion processes (so-called mitochondrial dynamics) that maintain mitochondrial quality and homeostasis. Damaged mitochondria are removed by a process known as mitophagy, which involves their degradation by a specific autophagosomal pathway. Over the last few years, remarkable efforts have been made to investigate the impact on the pathogenesis of Alzheimer’s disease (AD) of various forms of mitochondrial dysfunction, such as excessive reactive oxygen species (ROS) production, mitochondrial Ca2+ dyshomeostasis, loss of ATP, and defects in mitochondrial dynamics and transport, and mitophagy. Recent research suggests that restoration of mitochondrial function by physical exercise, an antioxidant diet, or therapeutic approaches can delay the onset and slow the progression of AD. In this review, we focus on recent progress that highlights the crucial role of alterations in mitochondrial function and oxidative stress in the pathogenesis of AD, emphasizing a framework of existing and potential therapeutic approaches.
Collapse
Affiliation(s)
- Afzal Misrani
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Sidra Tabassum
- School of Life Sciences, Guangzhou University, Guangzhou, China
| | - Li Yang
- School of Life Sciences, Guangzhou University, Guangzhou, China
| |
Collapse
|
16
|
Urbani A, Prosdocimi E, Carrer A, Checchetto V, Szabò I. Mitochondrial Ion Channels of the Inner Membrane and Their Regulation in Cell Death Signaling. Front Cell Dev Biol 2021; 8:620081. [PMID: 33585458 PMCID: PMC7874202 DOI: 10.3389/fcell.2020.620081] [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: 10/21/2020] [Accepted: 12/07/2020] [Indexed: 12/11/2022] Open
Abstract
Mitochondria are bioenergetic organelles with a plethora of fundamental functions ranging from metabolism and ATP production to modulation of signaling events leading to cell survival or cell death. Ion channels located in the outer and inner mitochondrial membranes critically control mitochondrial function and, as a consequence, also cell fate. Opening or closure of mitochondrial ion channels allow the fine-tuning of mitochondrial membrane potential, ROS production, and function of the respiratory chain complexes. In this review, we critically discuss the intracellular regulatory factors that affect channel activity in the inner membrane of mitochondria and, indirectly, contribute to cell death. These factors include various ligands, kinases, second messengers, and lipids. Comprehension of mitochondrial ion channels regulation in cell death pathways might reveal new therapeutic targets in mitochondria-linked pathologies like cancer, ischemia, reperfusion injury, and neurological disorders.
Collapse
Affiliation(s)
- Andrea Urbani
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- Department of Biology, University of Padova, Padua, Italy
| | | | - Andrea Carrer
- Department of Biomedical Sciences, University of Padova, Padua, Italy
- Department of Biology, University of Padova, Padua, Italy
| | | | - Ildikò Szabò
- Department of Biology, University of Padova, Padua, Italy
| |
Collapse
|
17
|
Jiang L, Yin X, Chen YH, Chen Y, Jiang W, Zheng H, Huang FQ, Liu B, Zhou W, Qi LW, Li J. Proteomic analysis reveals ginsenoside Rb1 attenuates myocardial ischemia/reperfusion injury through inhibiting ROS production from mitochondrial complex I. Am J Cancer Res 2021; 11:1703-1720. [PMID: 33408776 PMCID: PMC7778584 DOI: 10.7150/thno.43895] [Citation(s) in RCA: 123] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 11/10/2020] [Indexed: 11/13/2022] Open
Abstract
Rationale: Reactive oxygen species (ROS) burst from mitochondrial complex I is considered the critical cause of ischemia/reperfusion (I/R) injury. Ginsenoside Rb1 has been reported to protect the heart against I/R injury; however, the underlying mechanism remains unclear. This work aimed to investigate if ginsenoside Rb1 attenuates cardiac I/R injury by inhibiting ROS production from mitochondrial complex I. Methods: In in vivo experiments, mice were given ginsenoside Rb1 and then subjected to I/R injury. Mitochondrial ROS levels in the heart were determined using the mitochondrial-targeted probe MitoB. Mitochondrial proteins were used for TMT-based quantitative proteomic analysis. In in vitro experiments, adult mouse cardiomyocytes were pretreated with ginsenoside Rb1 and then subjected to hypoxia and reoxygenation insult. Mitochondrial ROS, NADH dehydrogenase activity, and conformational changes of mitochondrial complex I were analyzed. Results: Ginsenoside Rb1 decreased mitochondrial ROS production, reduced myocardial infarct size, preserved cardiac function, and limited cardiac fibrosis. Proteomic analysis showed that subunits of NADH dehydrogenase in mitochondrial complex I might be the effector proteins regulated by ginsenoside Rb1. Ginsenoside Rb1 inhibited complex I- but not complex II- or IV-dependent O2 consumption and enzyme activity. The inhibitory effects of ginsenoside Rb1 on mitochondrial I-dependent respiration and reperfusion-induced ROS production were rescued by bypassing complex I using yeast NADH dehydrogenase. Molecular docking and surface plasmon resonance experiments indicated that ginsenoside Rb1 reduced NADH dehydrogenase activity, probably via binding to the ND3 subunit to trap mitochondrial complex I in a deactive form upon reperfusion. Conclusion: Inhibition of mitochondrial complex I-mediated ROS burst elucidated the probable underlying mechanism of ginsenoside Rb1 in alleviating cardiac I/R injury.
Collapse
|
18
|
Dong Y, Zhao J, Zhu Q, Liu H, Wang J, Lu W. Melatonin inhibits the apoptosis of rooster Leydig cells by suppressing oxidative stress via AKT-Nrf2 pathway activation. Free Radic Biol Med 2020; 160:1-12. [PMID: 32758663 DOI: 10.1016/j.freeradbiomed.2020.06.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 05/31/2020] [Accepted: 06/09/2020] [Indexed: 12/11/2022]
Abstract
Oxidative stress has been described as a key driver of Leydig cell apoptosis. Melatonin has antioxidative and antiapoptotic effects, but the potential effects and mechanism of melatonin on oxidative stress and apoptosis in rooster Leydig cells remain unclear. Our results showed that melatonin biosynthetic enzymes and melatonin receptors were expressed in rooster Leydig cells and their expression were locally inhibited as rooster sexual maturation. We found that melatonin inhibited H2O2-induced apoptosis of rooster Leydig cell by activating the melatonin receptors Mel-1a and Mel-1b. Additionally, melatonin protects mitochondria from damage by reducing the level of oxidative stress in Leydig cells. Melatonin relieved H2O2-induced oxidative stress by significantly reducing intracellular ROS, MDA and 8-OHdG levels and increasing SOD and GSH-Px activities. Simultaneously, melatonin significantly reduced H2O2-induced depolarization of ΔΨm and decreased the release of Cytochrome C and Ca2+. We also observed that melatonin activated the Nrf2 pathway, while Nrf2 silencing abrogated the anti-oxidative and anti-apoptotic effects of melatonin in rooster Leydig cells. Furthermore, melatonin promoted the phosphorylation of AKT, while AKT inhibitor suppressed the Nrf2 pathway activated by melatonin and alleviated the inhibitory effects of melatonin on apoptosis and oxidative stress. In conclusion, melatonin could inhibit apoptosis in rooster Leydig cells by suppressing oxidative stress via activation of the AKT-Nrf2 pathway.
Collapse
Affiliation(s)
- Yangyunyi Dong
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jing Zhao
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Qingyu Zhu
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Hongyu Liu
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China
| | - Jun Wang
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| | - Wenfa Lu
- Joint Laboratory of Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Key Laboratory of Animal Production, Product Quality and Security, Ministry of Education, Jilin Agricultural University, Jilin Changchun, 130118, China; Jilin Province Engineering Laboratory for Ruminant Reproductive Biotechnology and Healthy Production, College of Animal Science and Technology, Jilin Agricultural University, Changchun, 130118, China.
| |
Collapse
|
19
|
Zhang XX, Wu XS, Mi SH, Fang SJ, Liu S, Xin Y, Zhao QM. Neuregulin-1 promotes mitochondrial biogenesis, attenuates mitochondrial dysfunction, and prevents hypoxia/reoxygenation injury in neonatal cardiomyocytes. Cell Biochem Funct 2020; 38:549-557. [PMID: 32037595 DOI: 10.1002/cbf.3503] [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: 09/14/2019] [Revised: 12/02/2019] [Accepted: 12/22/2019] [Indexed: 01/24/2023]
Abstract
Neuregulin-1 (NRG-1)/erythroblastic leukaemia viral oncogene homologues (ErbB) pathway activation plays a crucial role in regulating the adaptation of the adult heart to physiological and pathological stress. In the present study, we investigate the effect of recombined human NRG-1 (rhNRG-1) on mitochondrial biogenesis, mitochondrial function, and cell survival in neonatal rat cardiac myocytes (NRCMs) exposed to hypoxia/reoxygenation (H/R). The results of this study showed that, in the H/R-exposed NRCMs, mitochondrial biogenesis was impaired, as manifested by the decrease of the expression of peroxisome proliferator-activated receptor gamma coactivator-1 alpha (PGC-1α) and mitochondrial membrane proteins, the inner membrane (Tim23), mitofusin 1 (Mfn1), and mitofusin 2 (Mfn2). RhNRG-1 pretreatment effectively restored the expression of PGC-1α and these membrane proteins, upregulated the expression of the anti-apoptosis proteins Bcl-2 and Bcl-xL, preserved the mitochondrial membrane potential, and attenuated H/R-induced cell apoptosis. Blocking PGC-1 expression with siRNA abolished the beneficial role of rhNRG-1 on mitochondrial function and cell survival. The results of the present study strongly suggest that NRG-1/ErbB activation enhances the adaption of cardiomyocytes to H/R injury via promoted mitochondrial biogenesis and improved mitochondrial homeostasis. SIGNIFICANCE OF THE STUDY: The results of this research revealed for the first time the relationship between neuregulin-1 (NRG-1)/erythroblastic leukaemia viral oncogene homologues (ErbB) activation and mitochondrial biogenesis in neonatal cardiomyocytes and verified the significance of this promoted mitochondrial biogenesis in attenuating hypoxia/reoxygenation injury. This finding may open a new field to further understand the biological role of NRG-1/ErbB signalling pathway in cardiomyocyte.
Collapse
Affiliation(s)
- Xiao-Xia Zhang
- Department of Cardiology, Beijing AnZhen Hospital, Capital Medical University, Beijing, China
| | - Xue-Si Wu
- Department of Cardiology, Beijing AnZhen Hospital, Capital Medical University, Beijing, China
| | - Shu-Hua Mi
- Department of Cardiology, Beijing AnZhen Hospital, Capital Medical University, Beijing, China
| | - Shan-Juan Fang
- Emergency and Critical Care Center, Beijing Anzhen Hospital, Capital Medical University, China
| | - Sa Liu
- Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, The Capital Medical University, Beijing, China
| | - Yi Xin
- Institute of Heart Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, The Capital Medical University, Beijing, China
| | - Quan-Ming Zhao
- Department of Cardiology, Beijing AnZhen Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
20
|
Abstract
Adult cardiomyocytes are postmitotic cells that undergo very limited cell division. Thus, cardiomyocyte death as occurs during myocardial infarction has very detrimental consequences for the heart. Mitochondria have emerged as an important regulator of cardiovascular health and disease. Mitochondria are well established as bioenergetic hubs for generating ATP but have also been shown to regulate cell death pathways. Indeed many of the same signals used to regulate metabolism and ATP production, such as calcium and reactive oxygen species, are also key regulators of mitochondrial cell death pathways. It is widely hypothesized that an increase in calcium and reactive oxygen species activate a large conductance channel in the inner mitochondrial membrane known as the PTP (permeability transition pore) and that opening of this pore leads to necroptosis, a regulated form of necrotic cell death. Strategies to reduce PTP opening either by inhibition of PTP or inhibiting the rise in mitochondrial calcium or reactive oxygen species that activate PTP have been proposed. A major limitation of inhibiting the PTP is the lack of knowledge about the identity of the protein(s) that form the PTP and how they are activated by calcium and reactive oxygen species. This review will critically evaluate the candidates for the pore-forming unit of the PTP and discuss recent data suggesting that assumption that the PTP is formed by a single molecular identity may need to be reconsidered.
Collapse
Affiliation(s)
- Tyler M Bauer
- Laboratory of Cardiac Physiology, National Heart, Lung and Blood Institute, Bethesda, MD
| | - Elizabeth Murphy
- Laboratory of Cardiac Physiology, National Heart, Lung and Blood Institute, Bethesda, MD
| |
Collapse
|
21
|
Nakashima T, Noguchi T, Tahara Y, Nishimura K, Yasuda S, Onozuka D, Iwami T, Yonemoto N, Nagao K, Nonogi H, Ikeda T, Sato N, Tsutsui H. Public-access defibrillation and neurological outcomes in patients with out-of-hospital cardiac arrest in Japan: a population-based cohort study. Lancet 2019; 394:2255-2262. [PMID: 31862250 DOI: 10.1016/s0140-6736(19)32488-2] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2019] [Revised: 08/28/2019] [Accepted: 08/29/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND More than 80% of public-access defibrillation attempts do not result in sustained return of spontaneous circulation in patients who have had an out-of-hospital cardiac arrest (OHCA) and a shockable heart rhythm before arrival of emergency medical service (EMS) personnel. Neurological and survival outcomes in such patients have not been evaluated. We aimed to assess the neurological status and survival outcomes in such patients. METHODS This is a retropective analysis of a cohort study from a prospective, nationwide, population-based registry of 1 299 784 patients who had an OHCA event between Jan 1, 2005, and Dec 31, 2015 in Japan. The primary outcome was favourable neurological outcome (Cerebral Performance Category of 1 or 2) at 30 days after the OHCA and the secondary outcome was survival at 30 days following the OHCA. This study is registered with the University Hospital Medical Information Network Clinical Trials Registry, UMIN000009918. FINDINGS We identified 28 019 patients with bystander-witnessed OHCA and shockable heart rhythm who had received CPR from a bystander. Of these, 2242 (8·0%) patients did not achieve return of spontaneous circulation with CPR plus public-access defibrillation, and 25 087 (89·5%) patients did not achieve return of spontaneous circulation with CPR alone before EMS arrival. The proportion of patients with a favourable neurological outcome was significantly higher in those who received public-access defibrillation than those who did not (845 [37·7%] vs 5676 [22·6%]; adjusted odds ratio [OR] after propensity score-matching, 1·45 [95% CI 1·24-1·69], p<0·0001). The proportion of patients who survived at 30 days after the OHCA was also significantly higher in those who received public-access defibrillation than those who did not (987 [44·0%] vs 7976 [31·8%]; adjusted OR after propensity score-matching, 1·31 [95% CI 1·13-1·52], p<0·0001). INTERPRETATION Our findings support the benefits of public-access defibrillation and greater accessibility and availability of automated external defibrillators in the community. FUNDING None.
Collapse
Affiliation(s)
- Takahiro Nakashima
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Centre, Suita, Japan
| | - Teruo Noguchi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Centre, Suita, Japan
| | - Yoshio Tahara
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Centre, Suita, Japan.
| | - Kunihiro Nishimura
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Centre, Suita, Japan
| | - Satoshi Yasuda
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Centre, Suita, Japan
| | - Daisuke Onozuka
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Centre, Suita, Japan; Department of Health Communication, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Taku Iwami
- Kyoto University Health Service, Kyoto, Japan
| | - Naohiro Yonemoto
- Department of Biostatistics, Kyoto University School of Public Health, Kyoto, Japan
| | - Ken Nagao
- Cardiovascular Centre, Nihon University Hospital, Tokyo, Japan
| | - Hiroshi Nonogi
- Intensive Care Centre, Shizuoka General Hospital, Shizuoka, Japan
| | - Takanori Ikeda
- Department of Cardiovascular Medicine, Toho University Faculty of Medicine, Tokyo, Japan
| | - Naoki Sato
- Cardiology and Intensive Care Unit, Nippon Medical School, Musashi-Kosugi Hospital, Kawasaki, Japan
| | - Hiroyuki Tsutsui
- Department of Cardiovascular Medicine, Kyushu University, Fukuoka, Japan
| |
Collapse
|
22
|
Abrigo J, Marín T, Aguirre F, Tacchi F, Vilos C, Simon F, Arrese M, Cabrera D, Cabello-Verrugio C. N-Acetyl Cysteine Attenuates the Sarcopenia and Muscle Apoptosis Induced by Chronic Liver Disease. Curr Mol Med 2019; 20:60-71. [DOI: 10.2174/1566524019666190917124636] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 08/12/2019] [Accepted: 08/29/2019] [Indexed: 12/18/2022]
Abstract
Background:
Sarcopenia is characterized by the loss of muscle mass and
strength (muscle atrophy) because of aging or chronic diseases, such as chronic liver
disease (CLD). Different mechanisms are involved in skeletal muscle atrophy, including
decreased muscle fibre diameter and myosin heavy chain levels and increased
ubiquitin–proteasome pathway activity, oxidative stress and myonuclear apoptosis. We
recently found that all these mechanisms, except myonuclear apoptosis, which was not
evaluated in the previous study, were involved in muscle atrophy associated with
hepatotoxin 5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)-induced CLD.
Objective:
In the present study, we evaluated the involvement of myonuclear apoptosis
in CLD-associated sarcopenia and the effect of N-acetyl cysteine (NAC) treatment on
muscle strength and apoptosis, using a DDC-supplemented diet-fed mouse model.
Methods:
Four-month-old male C57BL6 mice were fed with a standard or DDCsupplemented
diet for six weeks in the absence or presence of NAC treatment.
Results:
Our results showed that NAC attenuated the decrease in muscle fibre diameter
and muscle strength associated with CLD-induced muscle wasting in gastrocnemius
(GA) muscle of DDC-supplemented diet-fed mice. In addition, in GA muscle of the mice
fed with DDC-supplemented diet-induced CLD showed increased myonuclear apoptosis
compared with the GA muscle of the control diet-fed mice, as evidenced by increased
apoptotic nuclei number, caspase-8 and caspase-9 expression, enzymatic activity of
caspase-3 and BAX/BCL-2 ratio. NAC treatment inhibited all the mechanisms
associated with myonuclear apoptosis in the GA muscle.
Conclusion:
To our knowledge, this is the first study which reports the redox regulation
of muscle strength and myonuclear apoptosis in CLD-induced sarcopenia.
Collapse
Affiliation(s)
- Johanna Abrigo
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Tabita Marín
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Francisco Aguirre
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Franco Tacchi
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Cristian Vilos
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| | - Felipe Simon
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
| | - Marco Arrese
- Departamento de Gastroenterologia, Facultad de Medicina. Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Daniel Cabrera
- Departamento de Gastroenterologia, Facultad de Medicina. Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Claudio Cabello-Verrugio
- Center for the Development of Nanoscience and Nanotechnology (CEDENNA), Universidad de Santiago de Chile, Santiago, Chile
| |
Collapse
|
23
|
Antonucci S, Di Sante M, Sileikyte J, Deveraux J, Bauer T, Bround MJ, Menabò R, Paillard M, Alanova P, Carraro M, Ovize M, Molkentin JD, Cohen M, Forte MA, Bernardi P, Di Lisa F, Murphy E. A novel class of cardioprotective small-molecule PTP inhibitors. Pharmacol Res 2019; 151:104548. [PMID: 31759087 DOI: 10.1016/j.phrs.2019.104548] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 11/12/2019] [Accepted: 11/14/2019] [Indexed: 01/03/2023]
Abstract
Ischemia/reperfusion (I/R) injury is mediated in large part by opening of the mitochondrial permeability transition pore (PTP). Consequently, inhibitors of the PTP hold great promise for the treatment of a variety of cardiovascular disorders. At present, PTP inhibition is obtained only through the use of drugs (e.g. cyclosporine A, CsA) targeting cyclophilin D (CyPD) which is a key modulator, but not a structural component of the PTP. This limitation might explain controversial findings in clinical studies. Therefore, we investigated the protective effects against I/R injury of small-molecule inhibitors of the PTP (63 and TR002) that do not target CyPD. Both compounds exhibited a dose-dependent inhibition of PTP opening in isolated mitochondria and were more potent than CsA. Notably, PTP inhibition was observed also in mitochondria devoid of CyPD. Compounds 63 and TR002 prevented PTP opening and mitochondrial depolarization induced by Ca2+ overload and by reactive oxygen species in neonatal rat ventricular myocytes (NRVMs). Remarkably, both compounds prevented cell death, contractile dysfunction and sarcomeric derangement induced by anoxia/reoxygenation injury in NRVMs at sub-micromolar concentrations, and were more potent than CsA. Cardioprotection was observed also in adult mouse ventricular myocytes and human iPSc-derived cardiomyocytes, as well as ex vivo in perfused hearts. Thus, this study demonstrates that 63 and TR002 represent novel cardioprotective agents that inhibit PTP opening independent of CyPD targeting.
Collapse
Affiliation(s)
| | - Moises Di Sante
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Justina Sileikyte
- Vollum Institute, and Department of Physiology and Pharmacology, Portland, OR, USA
| | - Jordan Deveraux
- Vollum Institute, and Department of Physiology and Pharmacology, Portland, OR, USA
| | - Tyler Bauer
- Systems Biology Center, NHLBI, NIH, Bethesda, MD, USA
| | - Michael J Bround
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA
| | - Roberta Menabò
- Department of Biomedical Sciences, University of Padova, Padova, Italy; National Research Council of Italy (CNR), Padova, Italy
| | - Melanie Paillard
- CarMeN Laboratory, University Claude Bernard Lyon 1, INSA Lyon, Oullins, France
| | - Petra Alanova
- Department of Biomedical Sciences, University of Padova, Padova, Italy; Department of Developmental Cardiology, Institute of Physiology CAS, Prague, Czech Republic
| | - Michela Carraro
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Michel Ovize
- CarMeN Laboratory, University Claude Bernard Lyon 1, INSA Lyon, Oullins, France
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, OH, USA; Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Michael Cohen
- Vollum Institute, and Department of Physiology and Pharmacology, Portland, OR, USA
| | - Michael A Forte
- Vollum Institute, and Department of Physiology and Pharmacology, Portland, OR, USA
| | - Paolo Bernardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy; National Research Council of Italy (CNR), Padova, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Padova, Italy; National Research Council of Italy (CNR), Padova, Italy.
| | | |
Collapse
|
24
|
Myocardial Adaptation in Pseudohypoxia: Signaling and Regulation of mPTP via Mitochondrial Connexin 43 and Cardiolipin. Cells 2019; 8:cells8111449. [PMID: 31744200 PMCID: PMC6912244 DOI: 10.3390/cells8111449] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 11/15/2019] [Indexed: 12/26/2022] Open
Abstract
Therapies intended to mitigate cardiovascular complications cannot be applied in practice without detailed knowledge of molecular mechanisms. Mitochondria, as the end-effector of cardioprotection, represent one of the possible therapeutic approaches. The present review provides an overview of factors affecting the regulation processes of mitochondria at the level of mitochondrial permeability transition pores (mPTP) resulting in comprehensive myocardial protection. The regulation of mPTP seems to be an important part of the mechanisms for maintaining the energy equilibrium of the heart under pathological conditions. Mitochondrial connexin 43 is involved in the regulation process by inhibition of mPTP opening. These individual cardioprotective mechanisms can be interconnected in the process of mitochondrial oxidative phosphorylation resulting in the maintenance of adenosine triphosphate (ATP) production. In this context, the degree of mitochondrial membrane fluidity appears to be a key factor in the preservation of ATP synthase rotation required for ATP formation. Moreover, changes in the composition of the cardiolipin’s structure in the mitochondrial membrane can significantly affect the energy system under unfavorable conditions. This review aims to elucidate functional and structural changes of cardiac mitochondria subjected to preconditioning, with an emphasis on signaling pathways leading to mitochondrial energy maintenance during partial oxygen deprivation.
Collapse
|
25
|
Tiong YL, Ng KY, Koh RY, Ponnudurai G, Chye SM. Melatonin Prevents Oxidative Stress-Induced Mitochondrial Dysfunction and Apoptosis in High Glucose-Treated Schwann Cells via Upregulation of Bcl2, NF-κB, mTOR, Wnt Signalling Pathways. Antioxidants (Basel) 2019; 8:antiox8070198. [PMID: 31247931 PMCID: PMC6680940 DOI: 10.3390/antiox8070198] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 06/07/2019] [Accepted: 06/14/2019] [Indexed: 12/11/2022] Open
Abstract
Neuropathy is a complication that affects more than 50% of long-standing diabetic patients. One of the causes of diabetes neuropathy (DN) is the apoptosis of Schwann cells due to prolonged exposure to high glucose and build-up of oxidative stress. Melatonin is a hormone that has a known antioxidant property. In this study, we investigated the protective effect of melatonin on high glucose-induced Schwann cells' apoptosis. Our results revealed that high glucose promoted apoptosis via mitochondrial-related oxidative stress and downregulated Bcl-2 family proteins in Schwann cells. In this signalling pathway, Bcl-2, Bcl-XL and Mcl-1 proteins were down-regulated while p-BAD and Puma proteins were up-regulated by high glucose treatment. Besides, we also proved that high glucose promoted apoptosis in Schwann cells through decreasing the p-NF-κB in the NF-κB signalling pathway. Key regulators of mTOR signalling pathway such as p-mTOR, Rictor and Raptor were also down-regulated after high glucose treatment. Additionally, high glucose treatment also decreased the Wnt signalling pathway downstream proteins (Wnt 5a/b, p-Lrp6 and Axin). Our results showed that melatonin treatment significantly inhibited high glucose-induced ROS generation, restored mitochondrial membrane potential and inhibited high glucose-induced apoptosis in Schwann cells. Furthermore, melatonin reversed the alterations of protein expression caused by high glucose treatment. Our results concluded that melatonin alleviates high glucose-induced apoptosis in Schwann cells through mitigating mitochondrial-related oxidative stress and the alterations of Bcl-2, NF-κB, mTOR and Wnt signalling pathways.
Collapse
Affiliation(s)
- Yee Lian Tiong
- School of Postgraduate Studies, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Khuen Yen Ng
- School of Pharmacy, Monash University Malaysia, Selangor 47500, Malaysia
| | - Rhun Yian Koh
- School of Health Science, International Medical University, Kuala Lumpur 57000, Malaysia
| | | | - Soi Moi Chye
- School of Health Science, International Medical University, Kuala Lumpur 57000, Malaysia.
| |
Collapse
|
26
|
Bour A, Kruglik SG, Chabanon M, Rangamani P, Puff N, Bonneau S. Lipid Unsaturation Properties Govern the Sensitivity of Membranes to Photoinduced Oxidative Stress. Biophys J 2019; 116:910-920. [PMID: 30777304 DOI: 10.1016/j.bpj.2019.01.033] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/28/2019] [Accepted: 01/29/2019] [Indexed: 12/11/2022] Open
Abstract
Unsaturated lipid oxidation is a fundamental process involved in different aspects of cellular bioenergetics; dysregulation of lipid oxidation is often associated with cell aging and death. To study how lipid oxidation affects membrane biophysics, we used a chlorin photosensitizer to oxidize vesicles of various lipid compositions and degrees of unsaturation in a controlled manner. We observed different shape transitions that can be interpreted as an increase in the area of the targeted membrane followed by a decrease. These area modifications induced by the chemical modification of the membrane upon oxidation were followed in situ by Raman tweezers microspectroscopy. We found that the membrane area increase corresponds to the lipids' peroxidation and is initiated by the delocalization of the targeted double bonds in the tails of the lipids. The subsequent decrease of membrane area can be explained by the formation of cleaved secondary products. As a result of these area changes, we observe vesicle permeabilization after a time lag that is characterized in relation with the level of unsaturation. The evolution of photosensitized vesicle radius was measured and yields an estimation of the mechanical changes of the membrane over oxidation time. The membrane is both weakened and permeabilized by the oxidation. Interestingly, the effect of unsaturation level on the dynamics of vesicles undergoing photooxidation is not trivial and thus carefully discussed. Our findings shed light on the fundamental dynamic mechanisms underlying the oxidation of lipid membranes and highlight the role of unsaturations on their physical and chemical properties.
Collapse
Affiliation(s)
- Aurélien Bour
- Sorbonne Université, Faculté des Sciences et Ingénierie, CNRS, Laboratoire Jean Perrin, Paris, France
| | - Sergei G Kruglik
- Sorbonne Université, Faculté des Sciences et Ingénierie, CNRS, Laboratoire Jean Perrin, Paris, France
| | - Morgan Chabanon
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California
| | - Padmini Rangamani
- Department of Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, California
| | - Nicolas Puff
- Sorbonne Université, Faculté des Sciences et Ingénierie, UFR 925, Paris, France; University Paris Diderot, Sorbonne Paris Cité, CNRS, Laboratoire Matière et Systèmes Complexes, UMR 7057, Paris, France
| | - Stephanie Bonneau
- Sorbonne Université, Faculté des Sciences et Ingénierie, CNRS, Laboratoire Jean Perrin, Paris, France.
| |
Collapse
|
27
|
Chuang CC, Zhou T, Olfert IM, Zuo L. Hypoxic Preconditioning Attenuates Reoxygenation-Induced Skeletal Muscle Dysfunction in Aged Pulmonary TNF-α Overexpressing Mice. Front Physiol 2019; 9:1720. [PMID: 30622474 PMCID: PMC6308319 DOI: 10.3389/fphys.2018.01720] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 11/15/2018] [Indexed: 11/26/2022] Open
Abstract
Aim: Skeletal muscle subjected to hypoxia followed by reoxygenation is susceptible to injury and subsequent muscle function decline. This phenomenon can be observed in the diaphragm during strenuous exercise or in pulmonary diseases such as chronic obstructive pulmonary diseases (COPD). Previous studies have shown that PO2 cycling or hypoxic preconditioning (HPC), as it can also be referred to as, protects muscle function via mechanisms involving reactive oxygen species (ROS). However, this HPC protection has not been fully elucidated in aged pulmonary TNF-α overexpressing (Tg+) mice (a COPD-like model). We hypothesize that HPC can exert protection on the diaphragms of Tg+ mice during reoxygenation through pathways involving ROS/phosphoinositide 3-kinase (PI3K)/protein kinase B (Akt)/extracellular signal regulated kinase (ERK), as well as the downstream activation of mitochondrial ATP-sensitive potassium channel (mitoKATP) and inhibition of mitochondrial permeability transition pore (mPTP). Methods: Isolated Tg+ diaphragm muscle strips were pre-treated with inhibitors for ROS, PI3K, Akt, ERK, or a combination of mitoKATP inhibitor and mPTP opener, respectively, prior to HPC. Another two groups of muscles were treated with either mitoKATP activator or mPTP inhibitor without HPC. Muscles were treated with 30-min hypoxia, followed by 15-min reoxygenation. Data were analyzed by multi-way ANOVA and expressed as means ± SE. Results: Muscle treated with HPC showed improved muscle function during reoxygenation (n = 5, p < 0.01). Inhibition of ROS, PI3K, Akt, or ERK abolished the protective effect of HPC. Simultaneous inhibition of mitoKATP and activation of mPTP also diminished HPC effects. By contrast, either the opening of mitoKATP channel or the closure of mPTP provided a similar protective effect to HPC by alleviating muscle function decline, suggesting that mitochondria play a role in HPC initiation (n = 5; p < 0.05). Conclusion: Hypoxic preconditioning may protect respiratory skeletal muscle function in Tg+ mice during reoxygenation through redox-sensitive signaling cascades and regulations of mitochondrial channels.
Collapse
Affiliation(s)
- Chia-Chen Chuang
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH, United States
| | - Tingyang Zhou
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH, United States
| | - I Mark Olfert
- Division of Exercise Physiology, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States.,Interdisciplinary Biophysics Graduate Program, The Ohio State University, Columbus, OH, United States.,Department of Biology, The University of Maine, Presque Isle, ME, United States
| |
Collapse
|
28
|
The hypoxia-tolerant vertebrate brain: Arresting synaptic activity. Comp Biochem Physiol B Biochem Mol Biol 2018; 224:61-70. [DOI: 10.1016/j.cbpb.2017.11.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 11/23/2017] [Accepted: 11/30/2017] [Indexed: 01/16/2023]
|
29
|
Teixeira J, Oliveira C, Cagide F, Amorim R, Garrido J, Borges F, Oliveira PJ. Discovery of a new mitochondria permeability transition pore (mPTP) inhibitor based on gallic acid. J Enzyme Inhib Med Chem 2018. [PMID: 29513043 PMCID: PMC6010063 DOI: 10.1080/14756366.2018.1442831] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Pharmacological interventions targeting mitochondria present several barriers for a complete efficacy. Therefore, a new mitochondriotropic antioxidant (AntiOxBEN3) based on the dietary antioxidant gallic acid was developed. AntiOxBEN3 accumulated several thousand-fold inside isolated rat liver mitochondria, without causing disruption of the oxidative phosphorylation apparatus, as seen by the unchanged respiratory control ratio, phosphorylation efficiency, and transmembrane electric potential. AntiOxBEN3 showed also limited toxicity on human hepatocarcinoma cells. Moreover, AntiOxBEN3 presented robust iron-chelation and antioxidant properties in both isolated liver mitochondria and cultured rat and human cell lines. Along with its low toxicity profile and high antioxidant activity, AntiOxBEN3 strongly inhibited the calcium-dependent mitochondrial permeability transition pore (mPTP) opening. From our data, AntiOxBEN3 can be considered as a lead compound for the development of a new class of mPTP inhibitors and be used as mPTP de-sensitiser for basic research or clinical applications or emerge as a therapeutic application in mitochondria dysfunction-related disorders.
Collapse
Affiliation(s)
- José Teixeira
- a CIQUP, Department of Chemistry and Biochemistry, Faculty of Sciences , University of Porto , Porto , Portugal.,b Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech , Cantanhede , Portugal
| | - Catarina Oliveira
- a CIQUP, Department of Chemistry and Biochemistry, Faculty of Sciences , University of Porto , Porto , Portugal
| | - Fernando Cagide
- a CIQUP, Department of Chemistry and Biochemistry, Faculty of Sciences , University of Porto , Porto , Portugal
| | - Ricardo Amorim
- a CIQUP, Department of Chemistry and Biochemistry, Faculty of Sciences , University of Porto , Porto , Portugal.,c PhD Programme in Experimental Biology and Biomedicine (PDBEB) , Center for Neuroscience and Cell Biology, University of Coimbra , Coimbra , Portugal.,d III-Institute for Interdisciplinary Research , University of Coimbra , Portugal
| | - Jorge Garrido
- e Department of Chemical Engineering, School of Engineering (ISEP) , Polytechnic Institute of Porto , Porto , Portugal
| | - Fernanda Borges
- a CIQUP, Department of Chemistry and Biochemistry, Faculty of Sciences , University of Porto , Porto , Portugal
| | - Paulo J Oliveira
- b Center for Neuroscience and Cell Biology, University of Coimbra, UC-Biotech , Cantanhede , Portugal
| |
Collapse
|
30
|
O'Connor RS, Guo L, Ghassemi S, Snyder NW, Worth AJ, Weng L, Kam Y, Philipson B, Trefely S, Nunez-Cruz S, Blair IA, June CH, Milone MC. The CPT1a inhibitor, etomoxir induces severe oxidative stress at commonly used concentrations. Sci Rep 2018; 8:6289. [PMID: 29674640 PMCID: PMC5908836 DOI: 10.1038/s41598-018-24676-6] [Citation(s) in RCA: 134] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 04/03/2018] [Indexed: 11/09/2022] Open
Abstract
Etomoxir (ETO) is a widely used small-molecule inhibitor of fatty acid oxidation (FAO) through its irreversible inhibitory effects on the carnitine palmitoyl-transferase 1a (CPT1a). We used this compound to evaluate the role of fatty acid oxidation in rapidly proliferating T cells following costimulation through the CD28 receptor. We show that ETO has a moderate effect on T cell proliferation with no observable effect on memory differentiation, but a marked effect on oxidative metabolism. We show that this oxidative metabolism is primarily dependent upon glutamine rather than FAO. Using an shRNA approach to reduce CPT1a in T cells, we further demonstrate that the inhibition of oxidative metabolism in T cells by ETO is independent of its effects on FAO at concentrations exceeding 5 μM. Concentrations of ETO above 5 μM induce acute production of ROS with associated evidence of severe oxidative stress in proliferating T cells. In aggregate, these data indicate that ETO lacks specificity for CTP1a above 5 μM, and caution should be used when employing this compound for studies in cells due to its non-specific effects on oxidative metabolism and cellular redox.
Collapse
Affiliation(s)
- Roddy S O'Connor
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Lili Guo
- Penn SRP center, Center of Excellence in Environmental Toxicology, and Department of Systems Pharmacology and Translational Therapeutics at the University of Pennsylvania, Philadelphia, PA, USA
| | - Saba Ghassemi
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Andrew J Worth
- Penn SRP center, Center of Excellence in Environmental Toxicology, and Department of Systems Pharmacology and Translational Therapeutics at the University of Pennsylvania, Philadelphia, PA, USA
| | - Liwei Weng
- Penn SRP center, Center of Excellence in Environmental Toxicology, and Department of Systems Pharmacology and Translational Therapeutics at the University of Pennsylvania, Philadelphia, PA, USA
| | | | | | - Sophie Trefely
- A.J. Drexel Autism Institute, Drexel University, Philadelphia, PA, USA
| | - Selene Nunez-Cruz
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Ian A Blair
- Penn SRP center, Center of Excellence in Environmental Toxicology, and Department of Systems Pharmacology and Translational Therapeutics at the University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Milone
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
31
|
Abstract
The evolution of cardiac disease after an acute ischemic event depends on a complex and dynamic network of mechanisms alternating from ischemic damage due to acute coronary occlusion to reperfusion injury due to the adverse effects of coronary revascularization till post-ischemic remodeling. Cardioprotection is a new purpose of the therapeutic interventions in cardiology with the goal to reduce infarct size and thus prevent the progression toward heart failure after an acute ischemic event. In a complex biological system such as the human one, an effective cardioprotective strategy should diachronically target the network of cross-talking pathways underlying the disease progression. Thyroid system is strictly interconnected with heart homeostasis, and recent studies highlighted its role in cardioprotection, in particular through the preservation of mitochondrial function and morphology, the antifibrotic and proangiogenetic effect and also to the potential induction of cell regeneration and growth. The objective of this review was to highlight the cardioprotective role of triiodothyronine in the complexity of post-ischemic disease evolution.
Collapse
|
32
|
Vyas FS, Nelson CP, Dickenson JM. Role of transglutaminase 2 in A 1 adenosine receptor- and β 2-adrenoceptor-mediated pharmacological pre- and post-conditioning against hypoxia-reoxygenation-induced cell death in H9c2 cells. Eur J Pharmacol 2017; 819:144-160. [PMID: 29208472 DOI: 10.1016/j.ejphar.2017.11.049] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 11/20/2017] [Accepted: 11/30/2017] [Indexed: 02/07/2023]
Abstract
Pharmacologically-induced pre- and post-conditioning represent attractive therapeutic strategies to reduce ischaemia/reperfusion injury during cardiac surgery and following myocardial infarction. We have previously reported that transglutaminase 2 (TG2) activity is modulated by the A1 adenosine receptor and β2-adrenoceptor in H9c2 cardiomyoblasts. The primary aim of this study was to determine the role of TG2 in A1 adenosine receptor and β2-adrenoceptor-induced pharmacological pre- and post-conditioning in the H9c2 cells. H9c2 cells were exposed to 8h hypoxia (1% O2) followed by 18h reoxygenation, after which cell viability was assessed by monitoring mitochondrial reduction of MTT, lactate dehydrogenase release and caspase-3 activation. N6-cyclopentyladenosine (CPA; A1 adenosine receptor agonist), formoterol (β2-adrenoceptor agonist) or isoprenaline (non-selective β-adrenoceptor agonist) were added before hypoxia/reoxygenation (pre-conditioning) or at the start of reoxygenation following hypoxia (post-conditioning). Pharmacological pre- and post-conditioning with CPA and isoprenaline significantly reduced hypoxia/reoxygenation-induced cell death. In contrast, formoterol did not elicit protection. Pre-treatment with pertussis toxin (Gi/o-protein inhibitor), DPCPX (A1 adenosine receptor antagonist) or TG2 inhibitors (Z-DON and R283) attenuated the A1 adenosine receptor-induced pharmacological pre- and post-conditioning. Similarly, pertussis toxin, ICI 118,551 (β2-adrenoceptor antagonist) or TG2 inhibition attenuated the isoprenaline-induced cell survival. Knockdown of TG2 using small interfering RNA (siRNA) attenuated CPA and isoprenaline-induced pharmacological pre- and post-conditioning. Finally, proteomic analysis following isoprenaline treatment identified known (e.g. protein S100-A6) and novel (e.g. adenine phosphoribosyltransferase) protein substrates for TG2. These results have shown that A1 adenosine receptor and β2-adrenoceptor-induced protection against simulated hypoxia/reoxygenation occurs in a TG2 and Gi/o-protein dependent manner in H9c2 cardiomyoblasts.
Collapse
Affiliation(s)
- Falguni S Vyas
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK
| | - Carl P Nelson
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK
| | - John M Dickenson
- School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham NG11 8NS, UK.
| |
Collapse
|
33
|
Propofol Prevents Oxidative Stress by Decreasing the Ischemic Accumulation of Succinate in Focal Cerebral Ischemia–Reperfusion Injury. Neurochem Res 2017; 43:420-429. [DOI: 10.1007/s11064-017-2437-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 11/03/2017] [Accepted: 11/13/2017] [Indexed: 12/21/2022]
|
34
|
Dhingra A, Jayas R, Afshar P, Guberman M, Maddaford G, Gerstein J, Lieberman B, Nepon H, Margulets V, Dhingra R, Kirshenbaum LA. Ellagic acid antagonizes Bnip3-mediated mitochondrial injury and necrotic cell death of cardiac myocytes. Free Radic Biol Med 2017; 112:411-422. [PMID: 28838842 DOI: 10.1016/j.freeradbiomed.2017.08.010] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 08/03/2017] [Accepted: 08/13/2017] [Indexed: 01/10/2023]
Abstract
The Bcl-2 protein Bnip3 is crucial for provoking oxidative injury to mitochondria following anthracycline treatment or ischemia-reperfusion injury. Herein, we investigate the effects of the polyphenolic compound ellagic acid (EA) on Bnip3 mediated mitochondrial injury and necrotic cell death in cardiac myocytes. In contrast to vehicle treated cardiomyocytes, Bnip3 was highly enriched in mitochondrial fractions of cardiac myocytes treated with the anthracycline doxorubicin or in cells subjected to hypoxia (HPX). Mitochondrial associated Bnip3 was accompanied by mPTP opening and loss of ∆Ψm. The dynamin related fission protein Drp-1 was phosphorylated (Drp1616) and coincided with excessive mitochondrial fragmentation, mitophagy and necrosis in cardiac myocytes treated with doxorubicin or subjected to hypoxia. Moreover, knock-down of Bnip3 was sufficient to prevent mitochondrial fission and doxorubicin-induced cell death supporting the involvement of Bnip3 in doxorubicin cardiotoxity. Interestingly, mitochondrial associated Bnip3 in cells treated with doxorubicin was markedly reduced by EA. This resulted in significantly less mitochondrial fission and cell death. Notably, EA similarly suppressed mitochondrial injury and cell death induced by hypoxia or Bnip3 over-expression. Herein, we identify a novel signaling axis that operationally links EA and Bnip3 for suppression of cardiac cell death. We provide compelling new evidence that EA suppresses mitochondrial injury and necrotic cell death of cardiac myocytes by functionally abrogating Bnip3 activity. Hence, by suppressing mitochondrial injury induced by Bnip3, EA may provide a therapeutic advantage in reducing oxidative injury and cardiac dysfunction in cancer patients undergoing anthracycline treatment or individuals with ischemic cardiac stress.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antibiotics, Antineoplastic/toxicity
- Apoptosis/drug effects
- Autophagy/drug effects
- Cell Hypoxia/drug effects
- Doxorubicin/antagonists & inhibitors
- Doxorubicin/toxicity
- Dynamins/genetics
- Dynamins/metabolism
- Ellagic Acid/pharmacology
- Membrane Potential, Mitochondrial/drug effects
- Membrane Proteins/antagonists & inhibitors
- Membrane Proteins/genetics
- Membrane Proteins/metabolism
- Mitochondria, Heart/drug effects
- Mitochondria, Heart/metabolism
- Mitochondria, Heart/pathology
- Mitochondrial Dynamics/drug effects
- Mitochondrial Dynamics/genetics
- Mitochondrial Membrane Transport Proteins/genetics
- Mitochondrial Membrane Transport Proteins/metabolism
- Mitochondrial Permeability Transition Pore
- Mitochondrial Proteins/antagonists & inhibitors
- Mitochondrial Proteins/genetics
- Mitochondrial Proteins/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Necrosis/genetics
- Necrosis/metabolism
- Necrosis/pathology
- Phosphorylation/drug effects
- Primary Cell Culture
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- Rats
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- Abhinav Dhingra
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Rahul Jayas
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Pegah Afshar
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Matthew Guberman
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Graham Maddaford
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Johnathan Gerstein
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Brooke Lieberman
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Hilary Nepon
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Victoria Margulets
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Rimpy Dhingra
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6
| | - Lorrie A Kirshenbaum
- The Institute of Cardiovascular Sciences, St. Boniface Hospital Albrechtsen Research Centre, Department of Physiology and Pathophysiology, Canada; College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada R2H 2H6.
| |
Collapse
|
35
|
Andrienko TN, Pasdois P, Pereira GC, Ovens MJ, Halestrap AP. The role of succinate and ROS in reperfusion injury - A critical appraisal. J Mol Cell Cardiol 2017; 110:1-14. [PMID: 28689004 PMCID: PMC5678286 DOI: 10.1016/j.yjmcc.2017.06.016] [Citation(s) in RCA: 110] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Revised: 06/14/2017] [Accepted: 06/30/2017] [Indexed: 12/20/2022]
Abstract
We critically assess the proposal that succinate-fuelled reverse electron flow (REF) drives mitochondrial matrix superoxide production from Complex I early in reperfusion, thus acting as a key mediator of ischemia/reperfusion (IR) injury. Real-time surface fluorescence measurements of NAD(P)H and flavoprotein redox state suggest that conditions are unfavourable for REF during early reperfusion. Furthermore, rapid loss of succinate accumulated during ischemia can be explained by its efflux rather than oxidation. Moreover, succinate accumulation during ischemia is not attenuated by ischemic preconditioning (IP) despite powerful cardioprotection. In addition, measurement of intracellular reactive oxygen species (ROS) during reperfusion using surface fluorescence and mitochondrial aconitase activity detected major increases in ROS only after mitochondrial permeability transition pore (mPTP) opening was first detected. We conclude that mPTP opening is probably triggered initially by factors other than ROS, including increased mitochondrial [Ca2+]. However, IP only attenuates [Ca2+] increases later in reperfusion, again after initial mPTP opening, implying that IP regulates mPTP opening through additional mechanisms. One such is mitochondria-bound hexokinase 2 (HK2) which dissociates from mitochondria during ischemia in control hearts but not those subject to IP. Indeed, there is a strong correlation between the extent of HK2 loss from mitochondria during ischemia and infarct size on subsequent reperfusion. Mechanisms linking HK2 dissociation to mPTP sensitisation remain to be fully established but several related processes have been implicated including VDAC1 oligomerisation, the stability of contact sites between the inner and outer membranes, cristae morphology, Bcl-2 family members and mitochondrial fission proteins such as Drp1.
Collapse
Affiliation(s)
- Tatyana N Andrienko
- School of Biochemistry and The Bristol Heart Institute, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Philippe Pasdois
- School of Biochemistry and The Bristol Heart Institute, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Gonçalo C Pereira
- School of Biochemistry and The Bristol Heart Institute, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Matthew J Ovens
- School of Biochemistry and The Bristol Heart Institute, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK
| | - Andrew P Halestrap
- School of Biochemistry and The Bristol Heart Institute, Medical Sciences Building, University of Bristol, Bristol BS8 1TD, UK.
| |
Collapse
|
36
|
Santa-Helena E, Teixeira S, Castro MRD, Cabrera DDC, D'Oca CDRM, D'Oca MGM, Votto APS, Nery LEM, Gonçalves CAN. Protective role of the novel hybrid 3,5-dipalmitoyl-nifedipine in a cardiomyoblast culture subjected to simulated ischemia/reperfusion. Biomed Pharmacother 2017; 92:356-364. [PMID: 28554131 DOI: 10.1016/j.biopha.2017.05.091] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 05/22/2017] [Accepted: 05/22/2017] [Indexed: 12/21/2022] Open
Abstract
This work investigated the acute effects of the calcium channel blocker nifedipine and its new fatty hybrid derived from palmitic acid, 3,5-dipalmitoyl-nifedipine, compared to endocannabinoid anandamide during the process of inducing ischemia and reperfusion in cardiomyoblast H9c2 heart cells. The cardiomyoblasts were treated in 24 or 96-well plates (according to the test being performed) and maintaining the treatment until the end of hypoxia induction. The molecules were tested at concentrations of 10 and 100μM, cells were treated 24h after assembling the experimental plates and immediately before the I/R. Cell viability, apoptosis and necrosis, and generation of reactive oxygen species were evaluated. Nifedipine and 3,5-dipalmitoyl-nifedipine were used to assess radical scavenging potential and metal chelation. All tested molecules managed to reduce the levels of reactive oxygen species compared to the starvation+vehicle group. In in vitro assays, 3,5-dipalmitoyl-nifedipine showed more antioxidant activity than nifedipine. These results indicate the ability of this molecule to act as a powerful ROS scavenger. Cell viability was highest when cells were induced to I/R by both concentrations of anandamide and the higher concentration of DPN. These treatments also reduced cell death. Therefore, it was demonstrated that the process of hybridization of nifedipine with two palmitic acid chains assigns a greater cardioprotective effect to this molecule, thereby reducing the damage caused by hypoxia and reoxygenation in cardiomyoblast cultures.
Collapse
Affiliation(s)
- Eduarda Santa-Helena
- Graduate Program in Physiological Sciences, Comparative Animal Physiology, Institute of Biological Sciences, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil; Institute of Biological Sciences, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil
| | - Stefanie Teixeira
- Institute of Biological Sciences, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil
| | - Micheli Rosa de Castro
- Graduate Program in Physiological Sciences, Comparative Animal Physiology, Institute of Biological Sciences, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil
| | - Diego da Costa Cabrera
- Kolbe Organic Synthesis Laboratory, School of Chemistry and Food, FURG, Rio Grande, RS, Brazil
| | | | - Marcelo G Montes D'Oca
- Kolbe Organic Synthesis Laboratory, School of Chemistry and Food, FURG, Rio Grande, RS, Brazil
| | - Ana Paula S Votto
- Graduate Program in Physiological Sciences, Comparative Animal Physiology, Institute of Biological Sciences, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil; Institute of Biological Sciences, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil
| | - Luiz Eduardo Maia Nery
- Graduate Program in Physiological Sciences, Comparative Animal Physiology, Institute of Biological Sciences, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil; Institute of Biological Sciences, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil.
| | - Carla Amorim Neves Gonçalves
- Graduate Program in Physiological Sciences, Comparative Animal Physiology, Institute of Biological Sciences, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil; Institute of Biological Sciences, Universidade Federal do Rio Grande, FURG, Rio Grande, RS, Brazil
| |
Collapse
|
37
|
Andrienko T, Pasdois P, Rossbach A, Halestrap AP. Real-Time Fluorescence Measurements of ROS and [Ca2+] in Ischemic / Reperfused Rat Hearts: Detectable Increases Occur only after Mitochondrial Pore Opening and Are Attenuated by Ischemic Preconditioning. PLoS One 2016; 11:e0167300. [PMID: 27907091 PMCID: PMC5131916 DOI: 10.1371/journal.pone.0167300] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 11/12/2016] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial permeability transition pore (mPTP) opening is critical for ischemia / reperfusion (I/R) injury and is associated with increased [Ca2+] and reactive oxygen species (ROS). Here we employ surface fluorescence to establish the temporal sequence of these events in beating perfused hearts subject to global I/R. A bespoke fluorimeter was used to synchronously monitor surface fluorescence and reflectance of Langendorff-perfused rat hearts at multiple wavelengths, with simultaneous measurements of hemodynamic function. Potential interference by motion artefacts and internal filtering was assessed and minimised. Re-oxidation of NAD(P)H and flavoproteins on reperfusion (detected using autofluorescence) was rapid (t0.5 < 15 s) and significantly slower following ischemic preconditioning (IP). This argues against superoxide production from reduced Complex 1 being a critical mediator of initial mPTP opening during early reperfusion. Furthermore, MitoPY1 (a mitochondria-targeted H2O2-sensitive fluorescent probe) and aconitase activity measurements failed to detect matrix ROS increases during early reperfusion. However, two different fluorescent cytosolic ROS probes did detect ROS increases after 2–3 min of reperfusion, which was shown to be after initiation of mPTP opening. Cyclosporin A (CsA) and IP attenuated these responses and reduced infarct size. [Ca2+]i (monitored with Indo-1) increased progressively during ischemia, but dropped rapidly within 90 s of reperfusion when total mitochondrial [Ca2+] was shown to be increased. These early changes in [Ca2+] were not attenuated by IP, but substantial [Ca2+] increases were observed after 2–3 min reperfusion and these were prevented by both IP and CsA. Our data suggest that the major increases in ROS and [Ca2+] detected later in reperfusion are secondary to mPTP opening. If earlier IP-sensitive changes occur that might trigger initial mPTP opening they are below our limit of detection. Rather, we suggest that IP may inhibit initial mPTP opening by alternative mechanisms such as prevention of hexokinase 2 dissociation from mitochondria during ischemia.
Collapse
Affiliation(s)
- Tatyana Andrienko
- School of Biochemistry and Bristol Cardiovascular, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Philippe Pasdois
- School of Biochemistry and Bristol Cardiovascular, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
- INSERM U1045—L'Institut de Rythmologie et Modélisation Cardiaque (LIRYC), Université de Bordeaux, Bordeaux, France
| | - Andreas Rossbach
- School of Biochemistry and Bristol Cardiovascular, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
| | - Andrew P Halestrap
- School of Biochemistry and Bristol Cardiovascular, Biomedical Sciences Building, University of Bristol, Bristol, United Kingdom
- * E-mail:
| |
Collapse
|
38
|
Thyroid hormones improve cardiac function and decrease expression of pro-apoptotic proteins in the heart of rats 14 days after infarction. Apoptosis 2016; 21:184-94. [PMID: 26659365 DOI: 10.1007/s10495-015-1204-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Apoptosis is a key process associated with pathological cardiac remodelling in early-phase post-myocardial infarction. In this context, several studies have demonstrated an anti-apoptotic effect of thyroid hormones (TH). The aim of this study was to evaluate the effects of TH on the expression of proteins associated with the apoptotic process 14 days after infarction. Male Wistar rats (300-350 g) (n = 8/group) were divided into four groups: Sham-operated (SHAM), infarcted (AMI), sham-operated + TH (SHAMT) and infarcted + TH (AMIT). For 12 days, the animals received T3 and T4 [2 and 8 µg/(100 g day)] by gavage. After this, the rats were submitted to haemodynamic and echocardiographic analysis, and then were sacrificed and the heart tissue was collected for molecular analysis. Statistical analyses included two-way ANOVA with Student-Newman-Keuls post test. Ethics Committee number: 23262. TH administration prevented the loss of ventricular wall thickness and improved cardiac function in the infarcted rats 14 days after the injury. AMI rats presented an increase in the pro-apoptotic proteins p53 and JNK. The hormonal treatment prevented this increase in AMIT rats. In addition, TH administration decreased the Bax:Bcl-2 ratio in the infarcted rats. TH administration improved cardiac functional parameters, and decreased the expression of pro-apoptotic proteins 14 days after myocardial infarction.
Collapse
|
39
|
Paradis S, Charles AL, Meyer A, Lejay A, Scholey JW, Chakfé N, Zoll J, Geny B. Chronology of mitochondrial and cellular events during skeletal muscle ischemia-reperfusion. Am J Physiol Cell Physiol 2016; 310:C968-82. [PMID: 27076618 DOI: 10.1152/ajpcell.00356.2015] [Citation(s) in RCA: 90] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Peripheral artery disease (PAD) is a common circulatory disorder of the lower limb arteries that reduces functional capacity and quality of life of patients. Despite relatively effective available treatments, PAD is a serious public health issue associated with significant morbidity and mortality. Ischemia-reperfusion (I/R) cycles during PAD are responsible for insufficient oxygen supply, mitochondriopathy, free radical production, and inflammation and lead to events that contribute to myocyte death and remote organ failure. However, the chronology of mitochondrial and cellular events during the ischemic period and at the moment of reperfusion in skeletal muscle fibers has been poorly reviewed. Thus, after a review of the basal myocyte state and normal mitochondrial biology, we discuss the physiopathology of ischemia and reperfusion at the mitochondrial and cellular levels. First we describe the chronology of the deleterious biochemical and mitochondrial mechanisms activated by I/R. Then we discuss skeletal muscle I/R injury in the muscle environment, mitochondrial dynamics, and inflammation. A better understanding of the chronology of the events underlying I/R will allow us to identify key factors in the development of this pathology and point to suitable new therapies. Emerging data on mitochondrial dynamics should help identify new molecular and therapeutic targets and develop protective strategies against PAD.
Collapse
Affiliation(s)
- Stéphanie Paradis
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France;
| | - Anne-Laure Charles
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| | - Alain Meyer
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| | - Anne Lejay
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France; Department of Vascular Surgery and Kidney Transplantation, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France; and
| | - James W Scholey
- Department of Medicine and Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
| | - Nabil Chakfé
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Vascular Surgery and Kidney Transplantation, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France; and
| | - Joffrey Zoll
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| | - Bernard Geny
- University of Strasbourg, Fédération de Médecine Translationelle, EA 3072, Strasbourg, France; Department of Physiology and Functional Explorations, Thoracic Pathology Unit, Centre Hospitalier Régional Universitaire de Strasbourg, Strasbourg, France
| |
Collapse
|
40
|
Pingitore A, Iervasi G, Forini F. Role of the Thyroid System in the Dynamic Complex Network of Cardioprotection. Eur Cardiol 2016; 11:36-42. [PMID: 30310446 DOI: 10.15420/ecr.2016:9:2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Cardioprotection is a common goal of new therapeutic strategies in patients with coronary artery disease and/or left ventricular dysfunction. Myocardial damage following ischaemia/reperfusion injury lead to left ventricular adverse remodelling through many mechanisms arising from different cell types in different myocardial districts, namely the border and remote zone. Cardioprotection must face this complex, dynamic network of cooperating units. In this scenario, thyroid hormones can represent an effective therapeutic strategy due to the numerous actions and regulating mechanisms carried out at the level of the myocytes, interstitium and the vasculature, as well as to the activation of different pro-survival intracellular pathways involved in cardioprotection.
Collapse
Affiliation(s)
| | - Giorgio Iervasi
- Clinical Physiology Institute, National Research Council (CNR), Pisa, Italy
| | - Francesca Forini
- Clinical Physiology Institute, National Research Council (CNR), Pisa, Italy
| |
Collapse
|
41
|
Morin D, Musman J, Pons S, Berdeaux A, Ghaleh B. Mitochondrial translocator protein (TSPO): From physiology to cardioprotection. Biochem Pharmacol 2015; 105:1-13. [PMID: 26688086 DOI: 10.1016/j.bcp.2015.12.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Accepted: 12/04/2015] [Indexed: 01/08/2023]
Abstract
The mitochondrial translocator protein (TSPO) is a high affinity cholesterol binding protein which is primarily located in the outer mitochondrial membrane where it has been shown to interact with proteins implicated in mitochondrial permeability transition pore (mPTP) formation. TSPO is found in different species and is expressed at high levels in tissues that synthesize steroids but is also present in other peripheral tissues especially in the heart. TSPO has been involved in the import of cholesterol into mitochondria, a key step in steroidogenesis. This constitutes the main established function of the protein which was recently challenged by genetic studies. TSPO has also been associated directly or indirectly with a wide range of cellular functions such as apoptosis, cell proliferation, differentiation, regulation of mitochondrial function or porphyrin transport. In the heart the role of TSPO remains undefined but a growing body of evidence suggests that TSPO plays a critical role in regulating physiological cardiac function and that TSPO ligands may represent interesting drugs to protect the heart under pathological conditions. This article briefly reviews current knowledge regarding TSPO and discusses its role in the cardiovascular system under physiological and pathologic conditions. More particularly, it provides evidence that TSPO can represent an alternative strategy to develop new pharmacological agents to protect the myocardium against ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Didier Morin
- INSERM U955, Équipe 3, Créteil, France; Université Paris-Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France.
| | - Julien Musman
- INSERM U955, Équipe 3, Créteil, France; Université Paris-Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France.
| | - Sandrine Pons
- INSERM U955, Équipe 3, Créteil, France; Université Paris-Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France.
| | - Alain Berdeaux
- INSERM U955, Équipe 3, Créteil, France; Université Paris-Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France.
| | - Bijan Ghaleh
- INSERM U955, Équipe 3, Créteil, France; Université Paris-Est, UMR_S955, DHU A-TVB, UPEC, Créteil, France.
| |
Collapse
|
42
|
Differences in the profile of protection afforded by TRO40303 and mild hypothermia in models of cardiac ischemia/reperfusion injury. Eur J Pharmacol 2015; 760:7-19. [DOI: 10.1016/j.ejphar.2015.04.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Revised: 04/07/2015] [Accepted: 04/08/2015] [Indexed: 11/19/2022]
|
43
|
Tribulus terrestris
(Linn.) Attenuates Cellular Alterations Induced by Ischemia in H9c2 Cells Via Antioxidant Potential. Phytother Res 2015; 29:933-43. [DOI: 10.1002/ptr.5336] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2014] [Revised: 10/17/2014] [Accepted: 03/03/2015] [Indexed: 12/31/2022]
|
44
|
Forini F, Nicolini G, Iervasi G. Mitochondria as key targets of cardioprotection in cardiac ischemic disease: role of thyroid hormone triiodothyronine. Int J Mol Sci 2015; 16:6312-36. [PMID: 25809607 PMCID: PMC4394534 DOI: 10.3390/ijms16036312] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/10/2015] [Accepted: 03/12/2015] [Indexed: 12/30/2022] Open
Abstract
Ischemic heart disease is the major cause of mortality and morbidity worldwide. Early reperfusion after acute myocardial ischemia has reduced short-term mortality, but it is also responsible for additional myocardial damage, which in the long run favors adverse cardiac remodeling and heart failure evolution. A growing body of experimental and clinical evidence show that the mitochondrion is an essential end effector of ischemia/reperfusion injury and a major trigger of cell death in the acute ischemic phase (up to 48–72 h after the insult), the subacute phase (from 72 h to 7–10 days) and chronic stage (from 10–14 days to one month after the insult). As such, in recent years scientific efforts have focused on mitochondria as a target for cardioprotective strategies in ischemic heart disease and cardiomyopathy. The present review discusses recent advances in this field, with special emphasis on the emerging role of the biologically active thyroid hormone triiodothyronine (T3).
Collapse
Affiliation(s)
- Francesca Forini
- CNR Institute of Clinical Physiology, Via G. Moruzzi 1, Pisa 56124, Italy.
| | - Giuseppina Nicolini
- CNR Institute of Clinical Physiology, Via G. Moruzzi 1, Pisa 56124, Italy.
- Tuscany Region G. Monasterio Foundation, Via G. Moruzzi 1, Pisa 56124, Italy.
| | - Giorgio Iervasi
- CNR Institute of Clinical Physiology, Via G. Moruzzi 1, Pisa 56124, Italy.
| |
Collapse
|
45
|
Omar SA, Fok H, Tilgner KD, Nair A, Hunt J, Jiang B, Taylor P, Chowienczyk P, Webb AJ. Paradoxical normoxia-dependent selective actions of inorganic nitrite in human muscular conduit arteries and related selective actions on central blood pressures. Circulation 2015; 131:381-9; discussion 389. [PMID: 25533964 DOI: 10.1161/circulationaha.114.009554] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
BACKGROUND Inorganic nitrite dilates small resistance arterioles via hypoxia-facilitated reduction to vasodilating nitric oxide. The effects of nitrite in human conduit arteries have not been investigated. In contrast to nitrite, organic nitrates are established selective dilators of conduit arteries. METHODS AND RESULTS We examined the effects of local and systemic administration of sodium nitrite on the radial artery (a muscular conduit artery), forearm resistance vessels (forearm blood flow), and systemic hemodynamics in healthy male volunteers (n=43). Intrabrachial sodium nitrite (8.7 μmol/min) increased radial artery diameter by a median of 28.0% (25th and 75th percentiles, 25.7% and 40.1%; P<0.001). Nitrite (0.087-87 μmol/min) displayed conduit artery selectivity similar to that of glyceryl trinitrate (0.013-4.4 nmol/min) over resistance arterioles. Nitrite dose-dependently increased local cGMP production at the dose of 2.6 μmol/min by 1.1 pmol·min(-1)·100 mL(-1) tissue (95% confidence interval, 0.5-1.8). Nitrite-induced radial artery dilation was enhanced by administration of acetazolamide (oral or intra-arterial) and oral raloxifene (P=0.0248, P<0.0001, and P=0.0006, respectively) but was inhibited under hypoxia (P<0.0001) and hyperoxia (P=0.0006) compared with normoxia. Systemic intravenous administration of sodium nitrite (8.7 μmol/min) dilated the radial artery by 10.7% (95% confidence interval, 6.8-14.7) and reduced central systolic blood pressure by 11.6 mm Hg (95% confidence interval, 2.4-20.7), augmentation index, and pulse wave velocity without changing peripheral blood pressure. CONCLUSIONS Nitrite selectively dilates conduit arteries at supraphysiological and near-physiological concentrations via a normoxia-dependent mechanism that is associated with cGMP production and is enhanced by acetazolamide and raloxifene. The selective central blood pressure-lowering effects of nitrite have therapeutic potential to reduce cardiovascular events.
Collapse
Affiliation(s)
- Sami A Omar
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, Department of Clinical Pharmacology, London, UK (S.A.O., H.F., A.N., J.H., B.J., P.C., A.J.W.); Division of Women's Health, Women's Health Academic Centre, King's College London and King's Health Partners, London, UK (K.D.T., P.T.); Department of Anaesthetics (A.N.), and Biomedical Research Centre (S.A.O., H.F., A.N., J.H., B.J., P.C., A.W.), Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | - Henry Fok
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, Department of Clinical Pharmacology, London, UK (S.A.O., H.F., A.N., J.H., B.J., P.C., A.J.W.); Division of Women's Health, Women's Health Academic Centre, King's College London and King's Health Partners, London, UK (K.D.T., P.T.); Department of Anaesthetics (A.N.), and Biomedical Research Centre (S.A.O., H.F., A.N., J.H., B.J., P.C., A.W.), Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | - Katharina D Tilgner
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, Department of Clinical Pharmacology, London, UK (S.A.O., H.F., A.N., J.H., B.J., P.C., A.J.W.); Division of Women's Health, Women's Health Academic Centre, King's College London and King's Health Partners, London, UK (K.D.T., P.T.); Department of Anaesthetics (A.N.), and Biomedical Research Centre (S.A.O., H.F., A.N., J.H., B.J., P.C., A.W.), Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | - Ashok Nair
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, Department of Clinical Pharmacology, London, UK (S.A.O., H.F., A.N., J.H., B.J., P.C., A.J.W.); Division of Women's Health, Women's Health Academic Centre, King's College London and King's Health Partners, London, UK (K.D.T., P.T.); Department of Anaesthetics (A.N.), and Biomedical Research Centre (S.A.O., H.F., A.N., J.H., B.J., P.C., A.W.), Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | - Joanne Hunt
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, Department of Clinical Pharmacology, London, UK (S.A.O., H.F., A.N., J.H., B.J., P.C., A.J.W.); Division of Women's Health, Women's Health Academic Centre, King's College London and King's Health Partners, London, UK (K.D.T., P.T.); Department of Anaesthetics (A.N.), and Biomedical Research Centre (S.A.O., H.F., A.N., J.H., B.J., P.C., A.W.), Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | - Benyu Jiang
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, Department of Clinical Pharmacology, London, UK (S.A.O., H.F., A.N., J.H., B.J., P.C., A.J.W.); Division of Women's Health, Women's Health Academic Centre, King's College London and King's Health Partners, London, UK (K.D.T., P.T.); Department of Anaesthetics (A.N.), and Biomedical Research Centre (S.A.O., H.F., A.N., J.H., B.J., P.C., A.W.), Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | - Paul Taylor
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, Department of Clinical Pharmacology, London, UK (S.A.O., H.F., A.N., J.H., B.J., P.C., A.J.W.); Division of Women's Health, Women's Health Academic Centre, King's College London and King's Health Partners, London, UK (K.D.T., P.T.); Department of Anaesthetics (A.N.), and Biomedical Research Centre (S.A.O., H.F., A.N., J.H., B.J., P.C., A.W.), Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | - Phil Chowienczyk
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, Department of Clinical Pharmacology, London, UK (S.A.O., H.F., A.N., J.H., B.J., P.C., A.J.W.); Division of Women's Health, Women's Health Academic Centre, King's College London and King's Health Partners, London, UK (K.D.T., P.T.); Department of Anaesthetics (A.N.), and Biomedical Research Centre (S.A.O., H.F., A.N., J.H., B.J., P.C., A.W.), Guy's & St. Thomas' NHS Foundation Trust, London, UK
| | - Andrew J Webb
- From the King's College London British Heart Foundation Centre, Cardiovascular Division, Department of Clinical Pharmacology, London, UK (S.A.O., H.F., A.N., J.H., B.J., P.C., A.J.W.); Division of Women's Health, Women's Health Academic Centre, King's College London and King's Health Partners, London, UK (K.D.T., P.T.); Department of Anaesthetics (A.N.), and Biomedical Research Centre (S.A.O., H.F., A.N., J.H., B.J., P.C., A.W.), Guy's & St. Thomas' NHS Foundation Trust, London, UK.
| |
Collapse
|
46
|
Lim DC, Brady DC, Po P, Chuang LP, Marcondes L, Kim EY, Keenan BT, Guo X, Maislin G, Galante RJ, Pack AI. Simulating obstructive sleep apnea patients' oxygenation characteristics into a mouse model of cyclical intermittent hypoxia. J Appl Physiol (1985) 2014; 118:544-57. [PMID: 25429097 DOI: 10.1152/japplphysiol.00629.2014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mouse models of cyclical intermittent hypoxia (CIH) are used to study the consequences of both hypoxia and oxidative stress in obstructive sleep apnea (OSA). Whether or not a mouse model of CIH that simulates OSA patients' oxygenation characteristics would translate into improved patient care remains unanswered. First we identified oxygenation characteristics using the desaturation and resaturation time in 47 OSA subjects from the Molecular Signatures of Obstructive Sleep Apnea Cohort (MSOSA). We observe that a cycle of intermittent hypoxia is not sinusoidal; specifically, desaturation time increases in an almost linear relationship to the degree of hypoxia (nadir), whereas resaturation time is somewhat constant (∼15 s), irrespective of the nadir. Second, we modified the Hycon mouse model of CIH to accommodate a 15-s resaturation time. Using this modified CIH model, we explored whether a short resaturation schedule (15 s), which includes the characteristics of OSA patients, had a different effect on levels of oxidative stress (i.e., urinary 8,12-iso-iPF2α-VI levels) compared with sham and a long resaturation schedule (90 s), a schedule that is not uncommon in rodent models of CIH. Results suggest that shorter resaturation time may result in a higher level of 8,12-iso-iPF2α-VI compared with long resaturation or sham conditions. Therefore, simulating the rodent model of CIH to reflect this and other OSA patients' oxygenation characteristics may be worthy of consideration to better understand the effects of hypoxia, oxidative stress, and their interactions.
Collapse
Affiliation(s)
- Diane C Lim
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania;
| | - Daniel C Brady
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Pengse Po
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Li Pang Chuang
- Department of Thoracic Medicine and Department of Sleep Center, Chang Gung Memorial Hospital, Taipei, Taiwan and Graduate Institute of Clinical Medical Sciences, Chang Gung University, Tauyan, Taiwan; and
| | | | - Emily Y Kim
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Brendan T Keenan
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Xiaofeng Guo
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Greg Maislin
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Raymond J Galante
- Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Allan I Pack
- Division of Sleep Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania; Center for Sleep and Circadian Neurobiology, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
47
|
Barau C, Ghaleh B, Berdeaux A, Morin D. Cytochrome P450 and myocardial ischemia: potential pharmacological implication for cardioprotection. Fundam Clin Pharmacol 2014; 29:1-9. [DOI: 10.1111/fcp.12087] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 05/20/2014] [Accepted: 06/13/2014] [Indexed: 12/18/2022]
Affiliation(s)
- Caroline Barau
- Inserm, U955, Equipe 03; F-94000 Créteil France
- UMR_S955, UPEC; Université Paris-Est; F-94000 Créteil France
| | - Bijan Ghaleh
- Inserm, U955, Equipe 03; F-94000 Créteil France
- UMR_S955, UPEC; Université Paris-Est; F-94000 Créteil France
| | - Alain Berdeaux
- Inserm, U955, Equipe 03; F-94000 Créteil France
- UMR_S955, UPEC; Université Paris-Est; F-94000 Créteil France
| | - Didier Morin
- Inserm, U955, Equipe 03; F-94000 Créteil France
- UMR_S955, UPEC; Université Paris-Est; F-94000 Créteil France
| |
Collapse
|
48
|
Hu Y, Li L, Yin W, Shen L, You B, Gao H. Protective effect of proanthocyanidins on anoxia-reoxygenation injury of myocardial cells mediated by the PI3K/Akt/GSK-3β pathway and mitochondrial ATP-sensitive potassium channel. Mol Med Rep 2014; 10:2051-8. [PMID: 25109283 DOI: 10.3892/mmr.2014.2459] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 04/09/2014] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to examine the protective effect of proanthocyanidins anoxia-reoxygenation injury of myocardial cells and its association with phosphatidylinositol-3-kinase/Akt and glycogen synthase kinase (PI3K/Akt/GSK)-3β and ATP-sensitive potassium channels. Neonatal rat myocardial cells were cultured and an anoxia-reoxygenation model was established following pretreatment with various drugs. The experiment was divided into five groups according to an experimental scheme. An MTT assay was used to examine the cell survival, and reactive oxygen species (ROS) levels and apoptosis were detected by flow cytometry. Myocardial apoptosis was also examined by terminal deoxynucleotidyl transferase dUTP nick end labeling staining and western blot analysis was employed to detect the expression levels of caspase-3, p-Akt and p-glycogen synthase kinase (GSK)-3β. The results revealed that myocardial cells in the anoxia-reoxygenation group (A/R) exhibited reduced survival rates, increased ROS levels and enhanced caspase-3 expression, as compared with the control group (CN; P<0.05). However, the increase in p-Akt and p-GSK-3β expression was not significantly different. In the proanthocyanidin pretreatment group (PC) the myocardial cell survival rate was increased, ROS levels were reduced, caspase-3 expression was decreased and p-Akt and p-GSK-3β expression levels were significantly increased as compared with the A/R group (P<0.05). Blockade of the PIK3/Akt channel by LY294002 eliminated the protective effects of proanthocyanidins and induced a significant decrease in p-Akt protein and p-GSK-3β expression levels as compared with the PC group. The inhibitor of mitochondrial ATP-sensitive potassium (mitoKATP) channels, 5-HD, also significantly suppressed the protective effects of proanthocyanidins, but had no evident impact on p-Akt and p-GSK-3β expression as compared with the PC group. In conclusion, pretreatment with proanthocyanidins had a protective effect on rat myocardial cell anoxia/reoxygenation injury. This effect is associated with the activation of the PI3K/Akt/GSK-3β signaling pathway and the opening of mitoKATP channels, which may have important roles downstream of PI3K.
Collapse
Affiliation(s)
- Yanyan Hu
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Li Li
- Department of Medicine, Shandong University, Jinan, Shandong 250012, P.R. China
| | - Wenbin Yin
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Lin Shen
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Beian You
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| | - Haiqing Gao
- Department of Geriatrics, Qilu Hospital of Shandong University, Jinan, Shandong 250012, P.R. China
| |
Collapse
|
49
|
Abstract
Decreased oxygen availability impairs cellular energy production and, without a coordinated and matched decrease in energy consumption, cellular and whole organism death rapidly ensues. Of particular interest are mechanisms that protect brain from low oxygen injury, as this organ is not only the most sensitive to hypoxia, but must also remain active and functional during low oxygen stress. As a result of natural selective pressures, some species have evolved molecular and physiological mechanisms to tolerate prolonged hypoxia with no apparent detriment. Among these mechanisms are a handful of responses that are essential for hypoxia tolerance, including (i) sensors that detect changes in oxygen availability and initiate protective responses; (ii) mechanisms of energy conservation; (iii) maintenance of basic brain function; and (iv) avoidance of catastrophic cell death cascades. As the study of hypoxia-tolerant brain progresses, it is becoming increasingly apparent that mitochondria play a central role in regulating all of these critical mechanisms. Furthermore, modulation of mitochondrial function to mimic endogenous neuroprotective mechanisms found in hypoxia-tolerant species confers protection against otherwise lethal hypoxic stresses in hypoxia-intolerant organs and organisms. Therefore, lessons gleaned from the investigation of endogenous mechanisms of hypoxia tolerance in hypoxia-tolerant organisms may provide insight into clinical pathologies related to low oxygen stress.
Collapse
Affiliation(s)
- Matthew E. Pamenter
- Department of Zoology, The University of British Columbia, #4200-6270 University Boulevard, Vancouver, BC V6T 1Z4, Canada
| |
Collapse
|
50
|
De Marchi E, Bonora M, Giorgi C, Pinton P. The mitochondrial permeability transition pore is a dispensable element for mitochondrial calcium efflux. Cell Calcium 2014; 56:1-13. [PMID: 24755650 PMCID: PMC4074345 DOI: 10.1016/j.ceca.2014.03.004] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/15/2014] [Accepted: 03/21/2014] [Indexed: 02/06/2023]
Abstract
The mitochondrial permeability transition pore (mPTP) has long been known to have a role in mitochondrial calcium (Ca(2+)) homeostasis under pathological conditions as a mediator of the mitochondrial permeability transition and the activation of the consequent cell death mechanism. However, its role in the context of mitochondrial Ca(2+) homeostasis is not yet clear. Several studies that were based on PPIF inhibition or knock out suggested that mPTP is involved in the Ca(2+) efflux mechanism, while other observations have revealed the opposite result. The c subunit of the mitochondrial F1/FO ATP synthase has been recently found to be a fundamental component of the mPTP. In this work, we focused on the contribution of the mPTP in the Ca(2+) efflux mechanism by modulating the expression of the c subunit. We observed that forcing mPTP opening or closing did not impair mitochondrial Ca(2+) efflux. Therefore, our results strongly suggest that the mPTP does not participate in mitochondrial Ca(2+) homeostasis in a physiological context in HeLa cells.
Collapse
Affiliation(s)
- Elena De Marchi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Massimo Bonora
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Carlotta Giorgi
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paolo Pinton
- Department of Morphology, Surgery and Experimental Medicine, Section of Pathology, Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy.
| |
Collapse
|