1
|
Zhong H, Zhu L, Kong X, Zhang K, Tang L, Zhang H, Zhang B, Gou X. Characterization and comparative transcriptomic analysis of high-altitude adaptation in Tibetan chicken using RNA-sequencing. Poult Sci 2025; 104:104749. [PMID: 39793236 PMCID: PMC11761939 DOI: 10.1016/j.psj.2024.104749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 12/19/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Tibetan chicken, an indigenous breed, inhabit highland regions and are crucial livestock for local Tibetans. Compared with other chicken breeds that have migrated from lowlands to highlands, Tibetan chicken exhibits superior physiological adaptations to high-altitude environments. However, the genetic mechanisms underlying these adaptations remain unclear. Herein, we generated high-quality RNA-sequencing data from the heart samples of 19 adult lowland and Tibetan chickens subjected to hypoxic and normoxic conditions for 5 weeks. We explored the relationships between chicken populations and the environment using different expression gene detection and weighted gene co-expression network analysis (WGCNA). The Tibetan chicken genome was analyzed for positive selection pressures associated with high-altitude adaptation, to reveal 63 candidate genes primarily involved in heart development and apoptotic signaling pathways, including PARK7, which was then validated using quantitative real-time polymerase chain reaction (qRT-PCR). Further hypoxia-induced cardiomyocyte model tests showed the cardioprotective function of PARK7 by cell viability assay, indicating the cardioprotective function of PARK7 in chickens under low-oxygen conditions. Our findings provided new insights into the genetic basis of high-altitude adaptation in Tibetan chickens and offered values for breeding programs aimed at enhancing the resilience of livestock in challenging environments.
Collapse
Affiliation(s)
- Haian Zhong
- School of Animal Science and Technology, Foshan University, Foshan 528231, PR China; National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Li Zhu
- School of Animal Science and Technology, Foshan University, Foshan 528231, PR China; College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, PR China
| | - Xiaoyan Kong
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, PR China
| | - Kang Zhang
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Lin Tang
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, PR China
| | - Hao Zhang
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Bo Zhang
- National Engineering Laboratory for Animal Breeding, Beijing Key Laboratory for Animal Genetic Improvement, College of Animal Science and Technology, China Agricultural University, Beijing 100193, PR China
| | - Xiao Gou
- School of Animal Science and Technology, Foshan University, Foshan 528231, PR China.
| |
Collapse
|
2
|
Zhou JB, Wei TP, Wu D, Zhou F, Wang RX. DJ-1 as a Novel Therapeutic Target for Mitigating Myocardial Ischemia-Reperfusion Injury. Cardiovasc Ther 2024; 2024:6615720. [PMID: 39742003 PMCID: PMC11661871 DOI: 10.1155/cdr/6615720] [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: 09/18/2024] [Accepted: 11/28/2024] [Indexed: 01/03/2025] Open
Abstract
Ischemic heart disease (IHD) remains one of the most prominent causes of mortality and morbidity globally, and the risk of ischemia-reperfusion injury is becoming more severe and constant. This underscores the need to develop new methods to protect the heart from damage. DJ-1 is a multifunctional intracellular protein encoded by the PARK7 gene that plays roles in processes including the control of autophagy, the preservation of mitochondrial integrity, the prevention of apoptosis, and the elimination of oxidative stress. DJ-1 has recently been the focus of growing interest as a target molecule relevant to treating myocardial ischemia-reperfusion injury due to its protective properties and its role in cellular response mechanisms. Consistently, DJ-1-related interventions, such as its exogenous administration or the use of pharmacological agents, have been demonstrated to help protect the myocardium from ischemia-reperfusion injury and associated adverse outcomes. This review provides an overview of DJ-1 and its therapeutic relevance in the myocardium in the setting of ischemia and reperfusion.
Collapse
Affiliation(s)
- Jia-Bin Zhou
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center Nanjing Medical University, Wuxi 214023, China
| | - Tian-Peng Wei
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center Nanjing Medical University, Wuxi 214023, China
| | - Dan Wu
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center Nanjing Medical University, Wuxi 214023, China
| | - Feng Zhou
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center Nanjing Medical University, Wuxi 214023, China
| | - Ru-Xing Wang
- Department of Cardiology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center Nanjing Medical University, Wuxi 214023, China
| |
Collapse
|
3
|
Salis Torres A, Lee JE, Caporali A, Semple RK, Horrocks MH, MacRae VE. Mitochondrial Dysfunction as a Potential Mechanism Mediating Cardiac Comorbidities in Parkinson's Disease. Int J Mol Sci 2024; 25:10973. [PMID: 39456761 PMCID: PMC11507255 DOI: 10.3390/ijms252010973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/02/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Individuals diagnosed with Parkinson's disease (PD) often exhibit heightened susceptibility to cardiac dysfunction, reflecting a complex interaction between these conditions. The involvement of mitochondrial dysfunction in the development and progression of cardiac dysfunction and PD suggests a plausible commonality in some aspects of their molecular pathogenesis, potentially contributing to the prevalence of cardiac issues in PD. Mitochondria, crucial organelles responsible for energy production and cellular regulation, play important roles in tissues with high energetic demands, such as neurons and cardiac cells. Mitochondrial dysfunction can occur in different and non-mutually exclusive ways; however, some mechanisms include alterations in mitochondrial dynamics, compromised bioenergetics, biogenesis deficits, oxidative stress, impaired mitophagy, and disrupted calcium balance. It is plausible that these factors contribute to the increased prevalence of cardiac dysfunction in PD, suggesting mitochondrial health as a potential target for therapeutic intervention. This review provides an overview of the physiological mechanisms underlying mitochondrial quality control systems. It summarises the diverse roles of mitochondria in brain and heart function, highlighting shared pathways potentially exhibiting dysfunction and driving cardiac comorbidities in PD. By highlighting strategies to mitigate dysfunction associated with mitochondrial impairment in cardiac and neural tissues, our review aims to provide new perspectives on therapeutic approaches.
Collapse
Affiliation(s)
- Agustina Salis Torres
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK;
| | - Ji-Eun Lee
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
- IRR Chemistry Hub, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh EH16 4UU, UK
| | - Andrea Caporali
- Centre for Cardiovascular Science, Queen’s Medical Research Institute (QMRI), The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; (A.C.); (R.K.S.)
| | - Robert K. Semple
- Centre for Cardiovascular Science, Queen’s Medical Research Institute (QMRI), The University of Edinburgh, 47 Little France Crescent, Edinburgh EH16 4TJ, UK; (A.C.); (R.K.S.)
| | - Mathew H. Horrocks
- EaStCHEM School of Chemistry, University of Edinburgh, Edinburgh EH9 3FJ, UK;
- MRC Human Genetics Unit, Institute for Genetics and Molecular Medicine, The University of Edinburgh, Edinburgh EH4 2XU, UK
| | - Vicky E. MacRae
- The Roslin Institute and R(D)SVS, University of Edinburgh, Easter Bush, Midlothian EH25 9RH, UK; (A.S.T.); (J.-E.L.)
| |
Collapse
|
4
|
Ji YW, Wen XY, Tang HP, Jin ZS, Su WT, Zhou L, Xia ZY, Xia ZY, Lei SQ. DJ-1: Potential target for treatment of myocardial ischemia-reperfusion injury. Biomed Pharmacother 2024; 179:117383. [PMID: 39232383 DOI: 10.1016/j.biopha.2024.117383] [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: 06/21/2024] [Revised: 08/29/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024] Open
Abstract
Ischemic heart disease (IHD) is a significant global health concern, resulting in high rates of mortality and disability among patients. Although coronary blood flow reperfusion is a key treatment for IHD, it often leads to acute myocardial ischemia-reperfusion injury (IRI). Current intervention strategies have limitations in providing adequate protection for the ischemic myocardium. DJ-1, originally known as a Parkinson's disease related protein, is a highly conserved cytoprotective protein. It is involved in enhancing mitochondrial function, scavenging reactive oxygen species (ROS), regulating autophagy, inhibiting apoptosis, modulating anaerobic metabolism, and exerting anti-inflammatory effects. DJ-1 is also required for protective strategies, such as ischemic preconditioning, ischemic postconditioning, remote ischemic preconditioning and pharmacological conditioning. Therefore, DJ-1 emerges as a potential target for the treatment of myocardial IRI. Our comprehensive review delves into its protective mechanisms in myocardial IRI and the structural foundations underlying its functions.
Collapse
Affiliation(s)
- Yan-Wei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin-Yu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - He-Peng Tang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhen-Shuai Jin
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wa-Ting Su
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lu Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zheng-Yuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Shao-Qing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
5
|
Pfister K, Young V, Frankel B, Silva Barbosa A, Burton J, Bons J, Zhang B, Chiba T, Uhlean R, Goetzman E, Schilling B, Sims-Lucas S. Succinylation of Park7 activates a protective metabolic response to acute kidney injury. Am J Physiol Renal Physiol 2024; 327:F128-F136. [PMID: 38695076 DOI: 10.1152/ajprenal.00062.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 04/15/2024] [Accepted: 04/29/2024] [Indexed: 05/04/2024] Open
Abstract
Acute kidney injury (AKI) is extremely prevalent among hospitalizations and presents a significant risk for the development of chronic kidney disease and increased mortality. Ischemia caused by shock, trauma, and transplant are common causes of AKI. To attenuate ischemic AKI therapeutically, we need a better understanding of the physiological and cellular mechanisms underlying damage. Instances of ischemia are most damaging in proximal tubule epithelial cells (PTECs) where hypoxic signaling cascades, and perhaps more rapidly, posttranslational modifications (PTMs), act in concert to change cellular metabolism. Here, we focus on the effects of the understudied PTM, lysine succinylation. We have previously shown a protective effect of protein hypersuccinylation on PTECs after depletion of the desuccinylase sirtuin5. General trends in the results suggested that hypersuccinylation led to upregulation of peroxisomal activity and was protective against kidney injury. Included in the list of changes was the Parkinson's-related deglycase Park7. There is little known about any links between peroxisome activity and Park7. In this study, we show in vitro and in vivo that Park7 has a crucial role in protection from AKI and upregulated peroxisome activity. These data in combination with published results of Park7's protective role in cardiovascular damage and chronic kidney disease lead us to hypothesize that succinylation of Park7 may ameliorate oxidative damage resulting from AKI and prevent disease progression. This novel mechanism provides a potential therapeutic mechanism that can be targeted.NEW & NOTEWORTHY Succinylation is an understudied posttranslational modification that has been shown to increase peroxisomal activity. Furthermore, increased peroxisomal activity has been shown to reduce oxidative stress and protect proximal tubules after acute kidney injury. Analysis of mass spectrometry succinylomic and proteomic data reveals a novel role for Parkinson's related Park7 in mediating Nrf2 antioxidant response after kidney injury. This novel protection pathway provides new insights for kidney injury prevention and development of novel therapeutics.
Collapse
Affiliation(s)
- Katherine Pfister
- Department of Pediatrics, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Victoria Young
- Department of Pediatrics, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Brendon Frankel
- Department of Pediatrics, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Anne Silva Barbosa
- Department of Pediatrics, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Jordan Burton
- Buck Institute for Research on Aging, Novato, California, United States
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, California, United States
| | - Bob Zhang
- Department of Pediatrics, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Takuto Chiba
- Department of Pediatrics, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Rebecca Uhlean
- Department of Pediatrics, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Eric Goetzman
- Department of Pediatrics, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Birgit Schilling
- Buck Institute for Research on Aging, Novato, California, United States
| | - Sunder Sims-Lucas
- Department of Pediatrics, University of Pittsburgh School of Medicine, University of Pittsburgh Medical Center Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| |
Collapse
|
6
|
Najjar JA, Calvert JW. Effects of protein glycation and protective mechanisms against glycative stress. Curr Opin Pharmacol 2024; 76:102464. [PMID: 38796877 PMCID: PMC11229435 DOI: 10.1016/j.coph.2024.102464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/15/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024]
Abstract
Glycation is a posttranslational modification of proteins that contributes to the vast array of biological information that can be conveyed via a singular proteome. Understanding the role of advanced glycation end-products (AGEs) in human health and pathophysiology can be difficult, as the physiological effects of AGEs have been associated with multiple biological processes and disease state development, including acute myocardial ischemia-reperfusion injury, heart failure, and atherosclerosis, as well as tumor cell migration. The critical role of the glyoxalase system in the detoxification of methylglyoxal and other AGEs has been well established. Recently, evidence has emerged that DJ-1 displays antiglycative activity and may contribute to another mechanism of protection against protein glycation outside of the glyoxalase system. Identification of potential substrates of DJ-1 and determination of the pathways in which DJ-1 operates, is needed to fully understand the role of this protein in modulating biological homeostasis and the development of disease.
Collapse
Affiliation(s)
- Jade A Najjar
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, USA
| | - John W Calvert
- Division of Cardiothoracic Surgery, Department of Surgery, Emory University School of Medicine, USA.
| |
Collapse
|
7
|
Mota GAF, de Souza SLB, Vileigas DF, da Silva VL, Sant'Ana PG, Costa LCDS, Padovani CR, Zanatti Bazan SG, Buzalaf MAR, Santos LDD, Okoshi MP, Gatto M, Cicogna AC. Myocardial proteome changes in aortic stenosis rats subjected to long-term aerobic exercise. J Cell Physiol 2024; 239:e31199. [PMID: 38291668 DOI: 10.1002/jcp.31199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 01/08/2024] [Accepted: 01/12/2024] [Indexed: 02/01/2024]
Abstract
The effects of exercise training (ET) on the heart of aortic stenosis (AS) rats are controversial and the mechanisms involved in alterations induced by ET have been poorly clarified. In this study, we analyzed the myocardial proteome to identify proteins modulated by moderate-intensity aerobic ET in rats with chronic supravalvular AS. Wistar rats were divided into four groups: sedentary control (C-Sed), exercised control (C-Ex), sedentary aortic stenosis (AS-Sed), and exercised AS (AS-Ex). ET consisted of five treadmill running sessions per week for 16 weeks. Statistical analysis was performed by ANOVA or Kruskal-Wallis and Goodman tests. Results were discussed at a significance level of 5%. At the end of the experiment, AS-Ex rats had higher functional capacity, lower blood lactate concentration, and better cardiac structural and left ventricular (LV) functional parameters than the AS-Sed. Myocardial proteome analysis showed that AS-Sed had higher relative protein abundance related to the glycolytic pathway, oxidative stress, and inflammation, and lower relative protein abundance related to beta-oxidation than C-Sed. AS-Ex had higher abundance of one protein related to mitochondrial biogenesis and lower relative protein abundance associated with oxidative stress and inflammation than AS-Sed. Proteomic data were validated for proteins related to lipid and glycolytic metabolism. Chronic pressure overload changes the abundance of myocardial proteins that are mainly involved in lipid and glycolytic energy metabolism in rats. Moderate-intensity aerobic training attenuates changes in proteins related to oxidative stress and inflammation and increases the COX4I1 protein, related to mitochondrial biogenesis. Protein changes are combined with improved functional capacity, cardiac remodeling, and LV function in AS rats.
Collapse
Affiliation(s)
- Gustavo Augusto Ferreira Mota
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | - Sérgio Luiz Borges de Souza
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | | | - Vitor Loureiro da Silva
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | - Paula Grippa Sant'Ana
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | - Licia Carla da Silva Costa
- Department of Biochemistry and Tissue Biology, Institute of Biology, University of Campinas, UNICAMP, Campinas, São Paulo, Brazil
| | - Carlos Roberto Padovani
- Department of Biostatistics, Institute of Bioscience, São Paulo State University, Botucatu, São Paulo, Brazil
| | - Silméia Garcia Zanatti Bazan
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | | | | | - Marina Politi Okoshi
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | - Mariana Gatto
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| | - Antonio Carlos Cicogna
- Department of Internal Medicine, Botucatu Medical School, São Paulo State University, UNESP, São Paulo, São Paulo, Brazil
| |
Collapse
|
8
|
Zhang W, Jiang H, Wu G, Huang P, Wang H, An H, Liu S, Zhang W. The pathogenesis and potential therapeutic targets in sepsis. MedComm (Beijing) 2023; 4:e418. [PMID: 38020710 PMCID: PMC10661353 DOI: 10.1002/mco2.418] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 10/01/2023] [Accepted: 10/12/2023] [Indexed: 12/01/2023] Open
Abstract
Sepsis is defined as "a life-threatening organ dysfunction caused by dysregulated host systemic inflammatory and immune response to infection." At present, sepsis continues to pose a grave healthcare concern worldwide. Despite the use of supportive measures in treating traditional sepsis, such as intravenous fluids, vasoactive substances, and oxygen plus antibiotics to eradicate harmful pathogens, there is an ongoing increase in both the morbidity and mortality associated with sepsis during clinical interventions. Therefore, it is urgent to design specific pharmacologic agents for the treatment of sepsis and convert them into a novel targeted treatment strategy. Herein, we provide an overview of the molecular mechanisms that may be involved in sepsis, such as the inflammatory response, immune dysfunction, complement deactivation, mitochondrial damage, and endoplasmic reticulum stress. Additionally, we highlight important targets involved in sepsis-related regulatory mechanisms, including GSDMD, HMGB1, STING, and SQSTM1, among others. We summarize the latest advancements in potential therapeutic drugs that specifically target these signaling pathways and paramount targets, covering both preclinical studies and clinical trials. In addition, this review provides a detailed description of the crosstalk and function between signaling pathways and vital targets, which provides more opportunities for the clinical development of new treatments for sepsis.
Collapse
Affiliation(s)
- Wendan Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Honghong Jiang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Faculty of PediatricsNational Engineering Laboratory for Birth defects prevention and control of key technologyBeijing Key Laboratory of Pediatric Organ Failurethe Chinese PLA General HospitalBeijingChina
| | - Gaosong Wu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Pengli Huang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Haonan Wang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Huazhasng An
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational MedicineThe First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan HospitalJinanShandongChina
| | - Sanhong Liu
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
| | - Weidong Zhang
- Shanghai Frontiers Science Center of TCM Chemical BiologyInstitute of Interdisciplinary Integrative Medicine ResearchShanghai University of Traditional Chinese MedicineShanghaiChina
- Department of PhytochemistrySchool of PharmacySecond Military Medical UniversityShanghaiChina
- The Research Center for Traditional Chinese MedicineShanghai Institute of Infectious Diseases and BiosecurityShanghai University of Traditional Chinese MedicineShanghaiChina
- Institute of Medicinal Plant DevelopmentChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
9
|
Lin J, Zheng X, Xiong Z, Xiang Q, Zhao Y, Jiang S, Sun Z, Fan D, Sun C, Li W. DJ-1-mediated p62 degradation delays intervertebral disc degeneration by inhibiting apoptosis of nucleus pulposus cells. Apoptosis 2023; 28:1357-1371. [PMID: 37300741 DOI: 10.1007/s10495-023-01862-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2023] [Indexed: 06/12/2023]
Abstract
Intervertebral disc degeneration (IDD) is the most important pathological basis of degenerative spinal diseases, for which effective interventions are still lacking. Oxidative stress is considered to be one of the leading pathological mechanisms contributing to IDD. However, the exact role of DJ-1 as an essential member of the antioxidant defense system in IDD is still unclear. Therefore, the aim of this study was to investigate the role played by DJ-1 in IDD and to reveal its potential molecular mechanisms. Western blot and immunohistochemical staining assays were performed to detect the expression of DJ-1 in degenerative nucleus pulposus cells (NPCs). After overexpression of DJ-1 in NPCs by lentiviral transfection, DCFH-DA and MitoSOX fluorescent probes were used to evaluate the levels of reactive oxygen species (ROS); while western blot, TUNEL staining, and Caspase-3 activity were used to assess apoptosis. Immunofluorescence staining was used to demonstrate the relationship between DJ-1 and p62. After inhibition of lysosomal degradation function with chloroquine, p62 degradation and apoptosis in DJ-1 overexpressing NPCs were further examined. In vivo, we assessed the therapeutic effect of upregulated DJ-1 on IDD by X-ray, MRI and Safranin O-Fast green staining. The protein expression of DJ-1 was significantly decreased in degenerated NPCs, accompanied by increased apoptosis. However, overexpression of DJ-1 significantly inhibited the elevated ROS levels and apoptosis in NPCs under oxidative stress. Mechanistically, our results showed that upregulation of DJ-1 promoted p62 degradation via the autophagic lysosomal pathway and that the protective effect of DJ-1 on NPCs under oxidative stress was partially mediated by promoting lysosomal pathway degradation of p62. Moreover, intradiscal injection of adeno-associated virus for overexpression of DJ-1 mitigated the progression of IDD in rats. This study reveals that DJ-1 maintains the homeostasis of NPCs by promoting the degradation of p62 through the autophagic lysosomal pathway, suggesting that DJ-1 is a promising new target for IDD intervention.
Collapse
Affiliation(s)
- Jialiang Lin
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Peking University Health Science Center, Beijing, China
| | - Xuanqi Zheng
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Peking University Health Science Center, Beijing, China
| | - Zhencheng Xiong
- Trauma Medical Center, Department of Orthopedics Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Qian Xiang
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Peking University Health Science Center, Beijing, China
| | - Yongzhao Zhao
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
- Peking University Health Science Center, Beijing, China
| | - Shuai Jiang
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Zhuoran Sun
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Dongwei Fan
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Chuiguo Sun
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China
| | - Weishi Li
- Department of Orthopaedics, Peking University Third Hospital, Haidian District, 49 North Garden Road, Beijing, 100191, China.
- Beijing Key Laboratory of Spinal Disease Research, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Ministry of Education, Beijing, China.
| |
Collapse
|
10
|
Sandrelli F, Bisaglia M. Molecular and Physiological Determinants of Amyotrophic Lateral Sclerosis: What the DJ-1 Protein Teaches Us. Int J Mol Sci 2023; 24:ijms24087674. [PMID: 37108835 PMCID: PMC10144135 DOI: 10.3390/ijms24087674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/12/2023] [Accepted: 04/19/2023] [Indexed: 04/29/2023] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is an adult-onset disease which causes the progressive degeneration of cortical and spinal motoneurons, leading to death a few years after the first symptom onset. ALS is mainly a sporadic disorder, and its causative mechanisms are mostly unclear. About 5-10% of cases have a genetic inheritance, and the study of ALS-associated genes has been fundamental in defining the pathological pathways likely also involved in the sporadic forms of the disease. Mutations affecting the DJ-1 gene appear to explain a subset of familial ALS forms. DJ-1 is involved in multiple molecular mechanisms, acting primarily as a protective agent against oxidative stress. Here, we focus on the involvement of DJ-1 in interconnected cellular functions related to mitochondrial homeostasis, reactive oxygen species (ROS) levels, energy metabolism, and hypoxia response, in both physiological and pathological conditions. We discuss the possibility that impairments in one of these pathways may affect the others, contributing to a pathological background in which additional environmental or genetic factors may act in favor of the onset and/or progression of ALS. These pathways may represent potential therapeutic targets to reduce the likelihood of developing ALS and/or slow disease progression.
Collapse
Affiliation(s)
| | - Marco Bisaglia
- Department of Biology, University of Padova, 35131 Padova, Italy
- Study Center for Neurodegeneration (CESNE), 35100 Padova, Italy
| |
Collapse
|
11
|
Tsoporis JN, Amatullah H, Gupta S, Izhar S, Ektesabi AM, Vaswani CM, Desjardins JF, Kabir G, Teixera Monteiro AP, Varkouhi AK, Kavantzas N, Salpeas V, Rizos I, Marshall JC, Parker TG, Leong-Poi H, Dos Santos CC. DJ-1 Deficiency Protects against Sepsis-Induced Myocardial Depression. Antioxidants (Basel) 2023; 12:antiox12030561. [PMID: 36978809 PMCID: PMC10045744 DOI: 10.3390/antiox12030561] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/07/2023] [Accepted: 02/20/2023] [Indexed: 03/30/2023] Open
Abstract
Oxidative stress is considered one of the early underlying contributors of sepsis-induced myocardial depression. DJ-1, also known as PARK7, has a well-established role as an antioxidant. We have previously shown, in a clinically relevant model of polymicrobial sepsis, DJ-1 deficiency improved survival and bacterial clearance by decreasing ROS production. In the present study, we investigated the role of DJ-1 in sepsis-induced myocardial depression. Here we compared wildtype (WT) with DJ-1 deficient mice at 24 and 48 h after cecal ligation and puncture (CLP). In WT mice, DJ-1 was increased in the myocardium post-CLP. DJ-1 deficient mice, despite enhanced inflammatory and oxidative responses, had an attenuated hypertrophic phenotype, less apoptosis, improved mitochondrial function, and autophagy, that was associated with preservation of myocardial function and improved survival compared to WT mice post-CLP. Collectively, these results identify DJ-1 as a regulator of myocardial function and as such, makes it an attractive therapeutic target in the treatment of early sepsis-induced myocardial depression.
Collapse
Affiliation(s)
- James N Tsoporis
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Hajera Amatullah
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sahil Gupta
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Shehla Izhar
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Amin M Ektesabi
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chirag M Vaswani
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Jean-Francois Desjardins
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Golam Kabir
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Ana Paula Teixera Monteiro
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Amir K Varkouhi
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Nikolaos Kavantzas
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Vasileios Salpeas
- 1st Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Ioannis Rizos
- 2nd Department of Cardiology, Attikon University Hospital, 12462 Athens, Greece
| | - John C Marshall
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Thomas G Parker
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Howard Leong-Poi
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
| | - Claudia C Dos Santos
- The Keenan Research Centre of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Unity Health Toronto, 30 Bond Street, Toronto, ON M5B 1W8, Canada
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Institute of Medical Sciences, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
12
|
Persulfidation of DJ-1: Mechanism and Consequences. Biomolecules 2022; 13:biom13010027. [PMID: 36671412 PMCID: PMC9856005 DOI: 10.3390/biom13010027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
DJ-1 (also called PARK7) is a ubiquitously expressed protein involved in the etiology of Parkinson disease and cancers. At least one of its three cysteine residues is functionally essential, and its oxidation state determines the specific function of the enzyme. DJ-1 was recently reported to be persulfidated in mammalian cell lines, but the implications of this post-translational modification have not yet been analyzed. Here, we report that recombinant DJ-1 is reversibly persulfidated at cysteine 106 by reaction with various sulfane donors and subsequently inhibited. Strikingly, this reaction is orders of magnitude faster than C106 oxidation by H2O2, and persulfidated DJ-1 behaves differently than sulfinylated DJ-1. Both these PTMs most likely play a dedicated role in DJ-1 signaling or protective pathways.
Collapse
|
13
|
Fasting increases susceptibility to acute myocardial ischaemia/reperfusion injury through a sirtuin-3 mediated increase in fatty acid oxidation. Sci Rep 2022; 12:20551. [PMID: 36446868 PMCID: PMC9708654 DOI: 10.1038/s41598-022-23847-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 11/07/2022] [Indexed: 11/30/2022] Open
Abstract
Fasting increases susceptibility to acute myocardial ischaemia/reperfusion injury (IRI) but the mechanisms are unknown. Here, we investigate the role of the mitochondrial NAD+-dependent deacetylase, Sirtuin-3 (SIRT3), which has been shown to influence fatty acid oxidation and cardiac outcomes, as a potential mediator of this effect. Fasting was shown to shift metabolism from glucose towards fatty acid oxidation. This change in metabolic fuel substrate utilisation increased myocardial infarct size in wild-type (WT), but not SIRT3 heterozygous knock-out (KO) mice. Further analysis revealed SIRT3 KO mice were better adapted to starvation through an improved cardiac efficiency, thus protecting them from acute myocardial IRI. Mitochondria from SIRT3 KO mice were hyperacetylated compared to WT mice which may regulate key metabolic processes controlling glucose and fatty acid utilisation in the heart. Fasting and the associated metabolic switch to fatty acid respiration worsens outcomes in WT hearts, whilst hearts from SIRT3 KO mice are better adapted to oxidising fatty acids, thereby protecting them from acute myocardial IRI.
Collapse
|
14
|
How Well Do Rodent Models of Parkinson's Disease Recapitulate Early Non-Motor Phenotypes? A Systematic Review. Biomedicines 2022; 10:biomedicines10123026. [PMID: 36551782 PMCID: PMC9775565 DOI: 10.3390/biomedicines10123026] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
The prodromal phase of Parkinson's disease (PD) is characterised by many non-motor symptoms, and these have recently been posited to be predictive of later diagnosis. Genetic rodent models can develop non-motor phenotypes, providing tools to identify mechanisms underlying the early development of PD. However, it is not yet clear how reproducible non-motor phenotypes are amongst genetic PD rodent models, whether phenotypes are age-dependent, and the translatability of these phenotypes has yet to be explored. A systematic literature search was conducted on studies using genetic PD rodent models to investigate non-motor phenotypes; cognition, anxiety/depressive-like behaviour, gastrointestinal (GI) function, olfaction, circadian rhythm, cardiovascular and urinary function. In total, 51 genetic models of PD across 150 studies were identified. We found outcomes of most phenotypes were inconclusive due to inadequate studies, assessment at different ages, or variation in experimental and environmental factors. GI dysfunction was the most reproducible phenotype across all genetic rodent models. The mouse model harbouring mutant A53T, and the wild-type hα-syn overexpression (OE) model recapitulated the majority of phenotypes, albeit did not reliably produce concurrent motor deficits and nigral cell loss. Furthermore, animal models displayed different phenotypic profiles, reflecting the distinct genetic risk factors and heterogeneity of disease mechanisms. Currently, the inconsistent phenotypes within rodent models pose a challenge in the translatability and usefulness for further biomechanistic investigations. This review highlights opportunities to improve phenotype reproducibility with an emphasis on phenotypic assay choice and robust experimental design.
Collapse
|
15
|
Gallinat A, Rakovic A, Klein C, Badimon L. DJ-1 regulates mitochondrial gene expression during ischemia and reperfusion. Free Radic Biol Med 2022; 193:430-436. [PMID: 36341940 DOI: 10.1016/j.freeradbiomed.2022.10.315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/10/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
The early-onset Parkinson's disease protein DJ-1 is a multifunctional protein that plays a protective role against ischemia and reperfusion (I/R) injury and oxidative stress. Despite lacking a canonical RNA-binding domain DJ-1 exhibits RNA-binding activity and multiple transcripts have been identified. However, no functional characterization has been provided to date. Here, we have investigated the DJ-1-interacting transcripts, as well as the role of DJ-1 RNA-binding activity during ischemia and reperfusion. Among the identified DJ-1-interacting transcripts, we have distinguished a significant enrichment of mRNAs encoding mitochondrial proteins. The effects of DJ-1 depletion on mitochondrial protein expression and mitochondrial morphology were investigated using a CRISPR/Cas9 generated DJ-1 knockout (DJ-1KO) cell model. DJ-1 depletion resulted in increased MTND2 protein expression in resting cells; however, after exposure to I/R, MTND2 levels were significantly reduced with respect to wild type cells. Increased mitochondrial fission was consistently found in DJ-1KO cells after I/R exposure. MTND2 transcript binding to DJ-1 was increased during ischemia. Our results indicate that the RNA-binding activity of DJ-1 shield mitochondrial transcripts from oxidative damage.
Collapse
Affiliation(s)
- Alex Gallinat
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | | | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Lina Badimon
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CIBERCV-Instituto de Salud Carlos III, Madrid, Spain; UAB-Chair Cardiovascular Research, Barcelona, Spain.
| |
Collapse
|
16
|
Effects of Lycopene Attenuating Injuries in Ischemia and Reperfusion. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9309327. [PMID: 36246396 PMCID: PMC9568330 DOI: 10.1155/2022/9309327] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 07/25/2022] [Accepted: 09/15/2022] [Indexed: 11/18/2022]
Abstract
Tissue and organ ischemia can lead to cell trauma, tissue necrosis, irreversible damage, and death. While intended to reverse ischemia, reperfusion can further aggravate an ischemic injury (ischemia-reperfusion injury, I/R injury) through a range of pathologic processes. An I/R injury to one organ can also harm other organs, leading to systemic multiorgan failure. A type of carotenoid, lycopene, has been shown to treat and prevent many diseases (e.g., rheumatoid arthritis, cancer, diabetes, osteoporosis, male infertility, neurodegenerative diseases, and cardiovascular disease), making it a hot research topic in health care. Some recent researches have suggested that lycopene can evidently ameliorate ischemic and I/R injuries to many organs, but few clinical studies are available. Therefore, it is essential to review the effects of lycopene on ischemic and I/R injuries to different organs, which may help further research into its potential clinical applications.
Collapse
|
17
|
Gallinat A, Mendieta G, Vilahur G, Padró T, Badimon L. DJ-1 administration exerts cardioprotection in a mouse model of acute myocardial infarction. Front Pharmacol 2022; 13:1002755. [PMID: 36210822 PMCID: PMC9539284 DOI: 10.3389/fphar.2022.1002755] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/09/2022] [Indexed: 01/09/2023] Open
Abstract
Cardiovascular diseases, and particularly acute myocardial infarction (MI), are the most common causes of death worldwide. Infarct size is the major predictor of clinical outcomes in MI. The Parkinson’s disease associated protein, DJ-1 (also known as PARK7), is a multifunctional protein with chaperone, redox sensing and mitochondrial homeostasis activities. Previously, we provided the evidence for a central role of endogenous DJ-1 in the cardioprotection of post-conditioning. In the present study, we tested the hypothesis that systemic administration of recombinant DJ-1 exerts cardioprotective effects in a mouse model of MI and also explored the associated transcriptional response. We report a significant treatment-induced reduction in infarct size, leukocyte infiltration, apoptosis and oxidative stress. Effects potentially mediated by G-protein-coupled receptor signaling and modulation of the immune response. Collectively, our results indicate a protective role for the exogenously administrated DJ-1 upon MI, and provide the first line of evidence for an extracellular activity of DJ-1 regulating cardiac injury in vivo.
Collapse
Affiliation(s)
- Alex Gallinat
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
- Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Guiomar Mendieta
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Gemma Vilahur
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
- CIBERCV-Instituto de Salud Carlos III, Madrid, Spain
| | - Teresa Padró
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
- CIBERCV-Instituto de Salud Carlos III, Madrid, Spain
| | - Lina Badimon
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain
- CIBERCV-Instituto de Salud Carlos III, Madrid, Spain
- Cardiovascular Research Chair, UAB, Barcelona, Spain
- *Correspondence: Lina Badimon,
| |
Collapse
|
18
|
Gallinat A, Badimon L. DJ-1 interacts with the ectopic ATP-synthase in endothelial cells during acute ischemia and reperfusion. Sci Rep 2022; 12:12753. [PMID: 35882968 PMCID: PMC9325725 DOI: 10.1038/s41598-022-16998-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/19/2022] [Indexed: 01/28/2023] Open
Abstract
Endothelial cells (ECs) play a central role in ischemia. ATP-Synthase is now recognized to be ectopically expressed in the cell surface of many cell types, with putative roles described in angiogenesis, proliferation, and intracellular pH regulation. DJ-1 is a multifunctional protein, involved in cell protection against ischemia, ischemia–reperfusion (I/R), and oxidative stress, that regulates mitochondrial ATP-synthase. Here we focused on the characterization of the endothelial dynamics of DJ-1, and its implication in the regulation of the ectopic ATP-synthase (ecATP-S) activity, during acute ischemia and I/R in ECs. We found that DJ-1 is secreted from ECs, by a mechanism enhanced in ischemia and I/R. A cleaved form of DJ-1 (DJ-1∆C) was found only in the secretome of ischemic cells. The ecATP-S activity increased following acute ischemia in ECs, coinciding with DJ-1 and DJ-1∆C secretion. The inhibition of DJ-1 expression inhibited the ecATP-S response to ischemia by ∼ 50%, and its exogenous administration maximized the effect, together with an enhanced Akt phosphorylation and angiotube-formation potential at reperfusion. Immunoprecipitation studies showed direct interaction between DJ-1 and the ecATP-S. Altogether suggesting that DJ-1 is actively cleaved and released from ischemic ECs and plays an important role in the regulation of the ecATP-S activity during acute ischemia and reperfusion.
Collapse
Affiliation(s)
- Alex Gallinat
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni María Claret, 167, 08025, Barcelona, Spain.,Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni María Claret, 167, 08025, Barcelona, Spain. .,CIBERCV-Instituto de Salud Carlos III, Madrid, Spain. .,UAB-Chair Cardiovascular Research, Barcelona, Spain.
| |
Collapse
|
19
|
Pan G, Yang S, Han X, Wang X, Kou L, Xie J, Li C. Parkinson's disease protein 7 protected against oxidative stress of myocardial infarction direct through p47phox and nicotinamide adenine dinucleotide phosphate oxidase 4. Hum Exp Toxicol 2022; 41:9603271221124099. [PMID: 36042578 DOI: 10.1177/09603271221124099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In the present study, we aimed to investigate the role and mechanism of Parkinson's disease protein 7 (Park7) in myocardial infarction (MI). The Park7 expression in the serum and tissues was down-regulated in mice with MI. Recombinant Park7 protein protected against MI-induced injury and reduced oxidative stress in mice model. Conversely, knockout Park7 increased injury of MI and promoted oxidative stress in MI mice model. In embryonic rat cardiac myoblasts H9c2 cells, over-expression of Park7 reduced reactive oxygen species (ROS)-induced oxidative stress, while down-regulation of Park7 increased ROS-induced oxidative stress. Park7 combined nicotinamide adenine dinucleotide phosphate (NADPH) oxidase cytoplasmic subunit p47phox protein had direct effect on inducing NADPH activator. The inhibition of p47phox reduced the effects of Park7 in ROS production of H2O2-treated H9c2 cells. The regulation of NADPH participated in the effects of Park7 on ROS production of in both MI mice model and H2O2-treated H9c2 cells. Our data demonstrated that Park7 protects against oxidative stress in MI model direct through p47phox and NADPH oxidase 4.
Collapse
Affiliation(s)
- Guozhong Pan
- Department of Cardiology, Dongzhimen Hospital, 248912Beijing University of Chinese Medicine, Beijing, China
| | - Shiwei Yang
- Department of Cardiology, Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaowan Han
- Department of Cardiology, Dongzhimen Hospital, 248912Beijing University of Chinese Medicine, Beijing, China
| | - Xian Wang
- Department of Cardiology, Dongzhimen Hospital, 248912Beijing University of Chinese Medicine, Beijing, China
| | - Lanjun Kou
- Department of Cardiology, Dongzhimen Hospital, 248912Beijing University of Chinese Medicine, Beijing, China
| | - Jing Xie
- Department of Cardiology, Dongzhimen Hospital, 248912Beijing University of Chinese Medicine, Beijing, China
| | - Chunyan Li
- Department of Cardiology, Dongzhimen Hospital, 248912Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
20
|
Cuenca-Bermejo L, Almela P, Navarro-Zaragoza J, Fernández Villalba E, González-Cuello AM, Laorden ML, Herrero MT. Cardiac Changes in Parkinson's Disease: Lessons from Clinical and Experimental Evidence. Int J Mol Sci 2021; 22:13488. [PMID: 34948285 PMCID: PMC8705692 DOI: 10.3390/ijms222413488] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 01/18/2023] Open
Abstract
Dysautonomia is a common non-motor symptom in Parkinson's disease (PD). Most dysautonomic symptoms appear due to alterations in the peripheral nerves of the autonomic nervous system, including both the sympathetic and parasympathetic nervous systems. The degeneration of sympathetic nerve fibers and neurons leads to cardiovascular dysfunction, which is highly prevalent in PD patients. Cardiac alterations such as orthostatic hypotension, heart rate variability, modifications in cardiogram parameters and baroreflex dysfunction can appear in both the early and late stages of PD, worsening as the disease progresses. In PD patients it is generally found that parasympathetic activity is decreased, while sympathetic activity is increased. This situation gives rise to an imbalance of both tonicities which might, in turn, promote a higher risk of cardiac damage through tachycardia and vasoconstriction. Cardiovascular abnormalities can also appear as a side effect of PD treatment: L-DOPA can decrease blood pressure and aggravate orthostatic hypotension as a result of a negative inotropic effect on the heart. This unwanted side effect limits the therapeutic use of L-DOPA in geriatric patients with PD and can contribute to the number of hospital admissions. Therefore, it is essential to define the cardiac features related to PD for the monitorization of the heart condition in parkinsonian individuals. This information can allow the application of intervention strategies to improve the course of the disease and the proposition of new alternatives for its treatment to eliminate or reverse the motor and non-motor symptoms, especially in geriatric patients.
Collapse
Affiliation(s)
- Lorena Cuenca-Bermejo
- Clinical and Experimental Neuroscience Group/Biomedical Research Institute of Murcia (NiCE-IMIB)/Institute for Aging Research, School of Medicine, University of Murcia, 30100 Murcia, Spain; (L.C.-B.); (A.-M.G.-C.)
| | - Pilar Almela
- Department of Pharmacology, School of Medicine, Biomedical Research Institute of Murcia (IMIB), University of Murcia, 30100 Murcia, Spain; (P.A.); (J.N.-Z.); (M.-L.L.)
| | - Javier Navarro-Zaragoza
- Department of Pharmacology, School of Medicine, Biomedical Research Institute of Murcia (IMIB), University of Murcia, 30100 Murcia, Spain; (P.A.); (J.N.-Z.); (M.-L.L.)
| | - Emiliano Fernández Villalba
- Clinical and Experimental Neuroscience Group/Biomedical Research Institute of Murcia (NiCE-IMIB)/Institute for Aging Research, School of Medicine, University of Murcia, 30100 Murcia, Spain; (L.C.-B.); (A.-M.G.-C.)
| | - Ana-María González-Cuello
- Clinical and Experimental Neuroscience Group/Biomedical Research Institute of Murcia (NiCE-IMIB)/Institute for Aging Research, School of Medicine, University of Murcia, 30100 Murcia, Spain; (L.C.-B.); (A.-M.G.-C.)
| | - María-Luisa Laorden
- Department of Pharmacology, School of Medicine, Biomedical Research Institute of Murcia (IMIB), University of Murcia, 30100 Murcia, Spain; (P.A.); (J.N.-Z.); (M.-L.L.)
| | - María-Trinidad Herrero
- Clinical and Experimental Neuroscience Group/Biomedical Research Institute of Murcia (NiCE-IMIB)/Institute for Aging Research, School of Medicine, University of Murcia, 30100 Murcia, Spain; (L.C.-B.); (A.-M.G.-C.)
| |
Collapse
|
21
|
Pantner Y, Polavarapu R, Chin LS, Li L, Shimizu Y, Calvert JW. DJ-1 attenuates the glycation of mitochondrial complex I and complex III in the post-ischemic heart. Sci Rep 2021; 11:19408. [PMID: 34593886 PMCID: PMC8484662 DOI: 10.1038/s41598-021-98722-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 09/09/2021] [Indexed: 02/02/2023] Open
Abstract
DJ-1 is a ubiquitously expressed protein that protects cells from stress through its conversion into an active protease. Recent work found that the active form of DJ-1 was induced in the ischemic heart as an endogenous mechanism to attenuate glycative stress-the non-enzymatic glycosylation of proteins. However, specific proteins protected from glycative stress by DJ-1 are not known. Given that mitochondrial electron transport proteins have a propensity for being targets of glycative stress, we investigated if DJ-1 regulates the glycation of Complex I and Complex III after myocardial ischemia-reperfusion (I/R) injury. Initial studies found that DJ-1 localized to the mitochondria and increased its interaction with Complex I and Complex III 3 days after the onset of myocardial I/R injury. Next, we investigated the role DJ-1 plays in modulating glycative stress in the mitochondria. Analysis revealed that compared to wild-type control mice, mitochondria from DJ-1 deficient (DJ-1 KO) hearts showed increased levels of glycative stress following I/R. Additionally, Complex I and Complex III glycation were found to be at higher levels in DJ-1 KO hearts. This corresponded with reduced complex activities, as well as reduced mitochondrial oxygen consumption ant ATP synthesis in the presence of pyruvate and malate. To further determine if DJ-1 influenced the glycation of the complexes, an adenoviral approach was used to over-express the active form of DJ-1(AAV9-DJ1ΔC). Under I/R conditions, the glycation of Complex I and Complex III were attenuated in hearts treated with AAV9-DJ1ΔC. This was accompanied by improvements in complex activities, oxygen consumption, and ATP production. Together, this data suggests that cardiac DJ-1 maintains Complex I and Complex III efficiency and mitochondrial function during the recovery from I/R injury. In elucidating a specific mechanism for DJ-1's role in the post-ischemic heart, these data break new ground for potential therapeutic strategies using DJ-1 as a target.
Collapse
Affiliation(s)
- Yvanna Pantner
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA, 30322, USA
| | - Rohini Polavarapu
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA, 30322, USA
| | - Lih-Shen Chin
- Department Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Lian Li
- Department Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - John W Calvert
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| |
Collapse
|
22
|
Cytoprotective Mechanisms of DJ-1: Implications in Cardiac Pathophysiology. Molecules 2021; 26:molecules26133795. [PMID: 34206441 PMCID: PMC8270312 DOI: 10.3390/molecules26133795] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 11/17/2022] Open
Abstract
DJ-1 was originally identified as an oncogene product while mutations of the gene encoding DJ-1/PARK7 were later associated with a recessive form of Parkinson's disease. Its ubiquitous expression and diversity of function suggest that DJ-1 is also involved in mechanisms outside the central nervous system. In the last decade, the contribution of DJ-1 to the protection from ischemia-reperfusion injury has been recognized and its involvement in the pathophysiology of cardiovascular disease is attracting increasing attention. This review describes the current and gaps in our knowledge of DJ-1, focusing on its role in regulating cardiovascular function. In parallel, we present original data showing an association between increased DJ-1 expression and antiapoptotic and anti-inflammatory markers following cardiac and vascular surgical procedures. Future studies should address DJ-1's role as a plausible novel therapeutic target for cardiovascular disease.
Collapse
|
23
|
Chen R, Li W, Qiu Z, Zhou Q, Zhang Y, Li WY, Ding K, Meng QT, Xia ZY. Ischemic Postconditioning-Mediated DJ-1 Activation Mitigate Intestinal Mucosa Injury Induced by Myocardial Ischemia Reperfusion in Rats Through Keap1/Nrf2 Pathway. Front Mol Biosci 2021; 8:655619. [PMID: 33996908 PMCID: PMC8119885 DOI: 10.3389/fmolb.2021.655619] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Accepted: 04/12/2021] [Indexed: 01/23/2023] Open
Abstract
Intestinal mucosal barrier dysfunction induced by myocardial ischemia reperfusion (IR) injury often leads to adverse cardiovascular outcomes after myocardial infarction. Early detection and prevention of remote intestinal injury following myocardial IR may help to estimate and improve prognosis after acute myocardial infarction (AMI). This study investigated the protective effect of myocardial ischemic postconditioning (IPo) on intestinal barrier injury induced by myocardial IR and the underlying cellular signaling mechanisms with a focus on the DJ-1. Adult SD rats were subjected to unilateral myocardial IR with or without ischemic postconditioning. After 30 min of ischemia and 120 min of reperfusion, heart tissue, intestine, and blood were collected for subsequent examination. The outcome measures were (i) intestinal histopathology, (ii) intestinal barrier function and inflammatory responses, (iii) apoptosis and oxidative stress, and (iv) cellular signaling changes. IPo significantly attenuated intestinal injury induced by myocardial IR. Furthermore, IPo significantly increased DJ-1, nuclear Nrf2, NQO1, and HO-1 expression in the intestine and inhibited IR-induced apoptosis and oxidative stress. The protective effect of IPo was abolished by the knockdown of DJ-1. Conversely, the overexpression of DJ-1 provided a protective effect similar to that of IPo. Our data indicate that IPo protects the intestine against myocardial IR, which is likely mediated by the upregulation of DJ-1/Nrf2 pathway.
Collapse
Affiliation(s)
- Rong Chen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wei Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhen Qiu
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qin Zhou
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuan Zhang
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Wen-Yuan Li
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ke Ding
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing-Tao Meng
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhong-Yuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
24
|
Sharma P, Wang X, Ming CLC, Vettori L, Figtree G, Boyle A, Gentile C. Considerations for the Bioengineering of Advanced Cardiac In Vitro Models of Myocardial Infarction. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2003765. [PMID: 33464713 DOI: 10.1002/smll.202003765] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 09/03/2020] [Indexed: 06/12/2023]
Abstract
Despite the latest advances in cardiovascular biology and medicine, myocardial infarction (MI) remains one of the major causes of deaths worldwide. While reperfusion of the myocardium is critical to limit the ischemic damage typical of a MI event, it causes detrimental morphological and functional changes known as "reperfusion injury." This complex scenario is poorly represented in currently available models of ischemia/reperfusion injury, leading to a poor translation of findings from the bench to the bedside. However, more recent bioengineered in vitro models of the human heart represent more clinically relevant tools to prevent and treat MI in patients. These include 3D cultures of cardiac cells, the use of patient-derived stem cells, and 3D bioprinting technology. This review aims at highlighting the major features typical of a heart attack while comparing current in vitro, ex vivo, and in vivo models. This information has the potential to further guide in developing novel advanced in vitro cardiac models of ischemia/reperfusion injury. It may pave the way for the generation of advanced pathophysiological cardiac models with the potential to develop personalized therapies.
Collapse
Affiliation(s)
- Poonam Sharma
- Faculty of Medicine and Health, University of Newcastle, Newcastle, NSW, 2308, Australia
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, 3004, Australia
| | - Clara Liu Chung Ming
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Laura Vettori
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| | - Gemma Figtree
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
| | - Andrew Boyle
- Faculty of Medicine and Health, University of Newcastle, Newcastle, NSW, 2308, Australia
| | - Carmine Gentile
- School of Medicine and Public Health, University of Sydney, Sydney, NSW, 2000, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, 2065, Australia
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Building 11, Level 10, Room 115, 81 Broadway, Ultimo, NSW, 2007, Australia
| |
Collapse
|
25
|
De Lazzari F, Prag HA, Gruszczyk AV, Whitworth AJ, Bisaglia M. DJ-1: A promising therapeutic candidate for ischemia-reperfusion injury. Redox Biol 2021; 41:101884. [PMID: 33561740 PMCID: PMC7872972 DOI: 10.1016/j.redox.2021.101884] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/13/2021] [Accepted: 01/25/2021] [Indexed: 12/31/2022] Open
Abstract
DJ-1 is a multifaceted protein with pleiotropic functions that has been implicated in multiple diseases, ranging from neurodegeneration to cancer and ischemia-reperfusion injury. Ischemia is a complex pathological state arising when tissues and organs do not receive adequate levels of oxygen and nutrients. When the blood flow is restored, significant damage occurs over and above that of ischemia alone and is termed ischemia-reperfusion injury. Despite great efforts in the scientific community to ameliorate this pathology, its complex nature has rendered it challenging to obtain satisfactory treatments that translate to the clinic. In this review, we will describe the recent findings on the participation of the protein DJ-1 in the pathophysiology of ischemia-reperfusion injury, firstly introducing the features and functions of DJ-1 and, successively highlighting the therapeutic potential of the protein. DJ-1 has been shown to confer protection in ischemia-reperfusion injury models. DJ-1 protection relies on the activation of antioxidant signaling pathways. DJ-1 regulates mitochondrial homeostasis during ischemia and reperfusion. DJ-1 seems to modulate ion homeostasis during ischemia and reperfusion. DJ-1 may represent a promising therapeutic target for ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Federica De Lazzari
- Physiology, Genetics and Behaviour Unit, Department of Biology, University of Padova, 35131, Padova, Italy
| | - Hiran A Prag
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Anja V Gruszczyk
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Alexander J Whitworth
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Marco Bisaglia
- Physiology, Genetics and Behaviour Unit, Department of Biology, University of Padova, 35131, Padova, Italy.
| |
Collapse
|
26
|
Xiang Y, Fu L, Xiang HX, Zheng L, Tan ZX, Wang LX, Cao W, Xu DX, Zhao H. Correlations among Pulmonary DJ-1, VDR and Nrf-2 in patients with Chronic Obstructive Pulmonary Disease: A Case-control Study. Int J Med Sci 2021; 18:2449-2456. [PMID: 33967623 PMCID: PMC8100631 DOI: 10.7150/ijms.58452] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/02/2021] [Indexed: 02/06/2023] Open
Abstract
Parkinson protein 7 (PARK7)/DJ-1 (DJ-1) is a redox sensitive molecular and stabilizer of nuclear factor erythroid 2-related factor 2 (Nrf-2). Nrf-2 regulates the downstream antioxidant defense system and exerts a significant function in patients with chronic obstructive pulmonary disease (COPD). Vitamin D receptor (VDR) is the nuclear receptor that regulates the downstream target genes. This study aimed to analyze the associations among pulmonary function, DJ-1, VDR and Nrf-2 in COPD patients. Serum was collected from 180 COPD patients and control subjects. Thirty-five lung tissues were obtained. DJ-1 was measured using ELISA and western blotting. Nrf-2 and VDR were detected by immunohistochemistry. Serum and pulmonary DJ-1 levels were lower in COPD patients than those in control subjects. Pulmonary VDR-positive nuclei were reduced in COPD patients. Nrf-2-positive nuclei were reduced in lung tissues of COPD patients. On the contrary, Nrf-2-related downstream target proteins were elevated in COPD patients. Further correlation analysis indicated that forced expiratory volume in 1 second (FEV1) was positively associated with pulmonary DJ-1, VDR and Nrf-2 in patients with COPD. In addition, there were positive correlations among DJ-1, VDR and Nrf-2 in lung tissues of COPD patients. In conclusion, DJ-1, VDR and Nrf-2 were decreased in COPD patients compared with control subjects. The reduction of DJ-1 and VDR associating with Nrf-2 downregulation may be involved in the process of COPD.
Collapse
Affiliation(s)
- Ying Xiang
- Respiratory and critical care medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.,Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Lin Fu
- Respiratory and critical care medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.,Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Hui-Xian Xiang
- Respiratory and critical care medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ling Zheng
- Respiratory and critical care medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Zhu-Xia Tan
- Respiratory and critical care medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.,Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Li-Xiang Wang
- Respiratory and critical care medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Wei Cao
- Respiratory and critical care medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| | - Hui Zhao
- Respiratory and critical care medicine, Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.,Department of Toxicology, Anhui Medical University, Hefei, 230032, China
| |
Collapse
|
27
|
Mitochondrial Translocation of DJ-1 Is Mediated by Grp75: Implication in Cardioprotection of Resveratrol Against Hypoxia/Reoxygenation-Induced Oxidative Stress. J Cardiovasc Pharmacol 2020; 75:305-313. [PMID: 32040033 DOI: 10.1097/fjc.0000000000000805] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Resveratrol (Res) was recently reported to ameliorate hypoxia/reoxygenation (H/R)-caused oxidative stress in H9c2 cardiomyocytes through promoting the mitochondrial translocation of DJ-1 protein and subsequently preserving the activity of mitochondrial complex I. However, it is noteworthy that DJ-1 possesses no mitochondria-targeting sequence. Therefore, how Res induces DJ-1 mitochondrial translocation is an important and interesting question for further exploration. Glucose-regulated protein 75 (Grp75), whose N-terminus contains a 51-amino acid long mitochondrial-targeting signal peptide, is a cytoprotective chaperone that partakes in mitochondrial import of several proteins. Here, the contribution of Grp75 to mitochondrial import of DJ-1 by Res was investigated in a cellular model of H/R. Our results showed that Res upregulated the expression of DJ-1 protein, enhanced the interaction of DJ-1 and Grp75, and promoted DJ-1 translocation to mitochondria from cytosol in H9c2 cardiomyocytes undergoing H/R. Importantly, knockdown of Grp75 markedly reduced the interaction of DJ-1 with Grp75 and subsequent DJ-1 mitochondrial translocation induced by Res. Furthermore, Res pretreatment promoted the association of DJ-1 with ND1 and NDUFA4 subunits of complex I, preserved the activity of complex I, decreased mitochondria-derived reactive oxygen species production, and eventually ameliorated H/R-caused oxidative stress damage. Intriguingly, these effects were largely prevented also by small interfering RNA targeting Grp75. Overall, these results suggested that Grp75 interacts with DJ-1 to facilitate its translocation from cytosol to mitochondria, which is required for Res-mediated preservation of mitochondria complex I and cardioprotection from H/R-caused oxidative stress injury.
Collapse
|
28
|
Hernandez‐Resendiz S, Prunier F, Girao H, Dorn G, Hausenloy DJ. Targeting mitochondrial fusion and fission proteins for cardioprotection. J Cell Mol Med 2020; 24:6571-6585. [PMID: 32406208 PMCID: PMC7299693 DOI: 10.1111/jcmm.15384] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 01/05/2023] Open
Abstract
New treatments are needed to protect the myocardium against the detrimental effects of acute ischaemia/reperfusion (IR) injury following an acute myocardial infarction (AMI), in order to limit myocardial infarct (MI) size, preserve cardiac function and prevent the onset of heart failure (HF). Given the critical role of mitochondria in energy production for cardiac contractile function, prevention of mitochondrial dysfunction during acute myocardial IRI may provide novel cardioprotective strategies. In this regard, the mitochondrial fusion and fissions proteins, which regulate changes in mitochondrial morphology, are known to impact on mitochondrial quality control by modulating mitochondrial biogenesis, mitophagy and the mitochondrial unfolded protein response. In this article, we review how targeting these inter-related processes may provide novel treatment targets and new therapeutic strategies for reducing MI size, preventing the onset of HF following AMI.
Collapse
Affiliation(s)
- Sauri Hernandez‐Resendiz
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular & Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Centro de Biotecnologia‐FEMSATecnologico de MonterreyNuevo LeonMexico
| | - Fabrice Prunier
- Institut MITOVASCCNRS UMR 6015 INSERM U1083University Hospital Center of AngersUniversity of AngersAngersFrance
| | - Henrique Girao
- Faculty of MedicineCoimbra Institute for Clinical and Biomedical Research (iCBR)University of CoimbraPortugal
- Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Clinical Academic Centre of Coimbra (CACC)CoimbraPortugal
| | - Gerald Dorn
- Department of Internal MedicineCenter for PharmacogenomicsWashington University School of MedicineSt. LouisMOUSA
| | - Derek J. Hausenloy
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular & Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Yong Loo Lin School of MedicineNational University SingaporeSingaporeSingapore
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
- Cardiovascular Research CenterCollege of Medical and Health SciencesAsia UniversityTaichungTaiwan
| | | |
Collapse
|
29
|
Wang J, Zhang H, Du A, Li Y. DJ-1 alleviates anoxia and hypoglycemia injury in cardiac microvascular via AKT and GSH. Mol Cell Probes 2020; 53:101600. [PMID: 32445781 DOI: 10.1016/j.mcp.2020.101600] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 05/14/2020] [Accepted: 05/14/2020] [Indexed: 10/24/2022]
Abstract
Cardiac microvascular damage, which is often caused by anoxia and hypoglycemia, is associated with the development of cardiac injury. DJ-1 encodes a peptidase C56 protein family related protein, is has been linked to oxidative stress in various cells such as neurons, COPD epithelial cells, and macrophages. However, the effect of DJ-1 towards oxidative stress caused by anoxia and hypoglycemia of cardiac microvascular endothelial cells (CMEC) remains unclear. In this study, we investigated the role and underlying molecular mechanism of DJ-1 in CMEC with anoxia/hypoglycemic (A/H) injury. We found that the mRNA and the protein expression of DJ-1 in CMEC with A/H injury were significantly downregulated. DJ-1 overexpression by pcDNA.3.1-DJ-1 transfection elevated cell viability while it inhibited LDH leakage, cell apoptosis, caspase-3 activity, ROS level, and MDA contents, while knockdown of DJ-1 has the opposite results. In addition, tube formation was increased in DJ-1 overexpression, while it was decreased in DJ-1 knockdown CMEC with A/H injury. In addition, our results indicated that DJ-1 can regulate glutathione (GSH) levels by modulating AKT activity in CMEC with A/H injury. The downregulation of AKT and GSH may remove the protective role of DJ-1 against A/H injury in CMEC. Taken together, this study showed that DJ-1 upregulation protected CMEC against A/H injury via the AKT/GSH signaling pathway.
Collapse
Affiliation(s)
- Jia Wang
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, 110001, China; Department of Nursing, The First Affiliated Hospital of China Medical University, Liaoning, 110001, China
| | - Haishan Zhang
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, 110001, China
| | - Aolin Du
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, 110001, China
| | - Yang Li
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Liaoning, 110001, China.
| |
Collapse
|
30
|
Shimizu Y, Nicholson CK, Polavarapu R, Pantner Y, Husain A, Naqvi N, Chin L, Li L, Calvert JW. Role of DJ-1 in Modulating Glycative Stress in Heart Failure. J Am Heart Assoc 2020; 9:e014691. [PMID: 32067589 PMCID: PMC7070196 DOI: 10.1161/jaha.119.014691] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background DJ‐1 is a ubiquitously expressed protein typically associated with the development of early onset Parkinson disease. Recent data suggest that it also plays a role in the cellular response to stress. Here, we sought to determine the role DJ‐1 plays in the development of heart failure. Methods and Results Initial studies found that DJ‐1 deficient mice (DJ‐1 knockout; male; 8–10 weeks of age) exhibited more severe left ventricular cavity dilatation, cardiac dysfunction, hypertrophy, and fibrosis in the setting of ischemia‐reperfusion–induced heart failure when compared with wild‐type littermates. In contrast, the overexpression of the active form of DJ‐1 using a viral vector approach resulted in significant improvements in the severity of heart failure when compared with mice treated with a control virus. Subsequent studies aimed at evaluating the underlying protective mechanisms found that cardiac DJ‐1 reduces the accumulation of advanced glycation end products and activation of the receptor for advanced glycation end products—thus, reducing glycative stress. Conclusions These results indicate that DJ‐1 is an endogenous cytoprotective protein that protects against the development of ischemia‐reperfusion–induced heart failure by reducing glycative stress. Our findings also demonstrate the feasibility of using a gene therapy approach to deliver the active form of DJ‐1 to the heart as a therapeutic strategy to protect against the consequences of ischemic injury, which is a major cause of death in western populations.
Collapse
Affiliation(s)
- Yuuki Shimizu
- Division of Cardiothoracic SurgeryDepartment of SurgeryCarlyle Fraser Heart CenterEmory University School of MedicineAtlantaGA
- Department of CardiologyNagoya University Graduate School of MedicineNagoyaJapan
| | - Chad K. Nicholson
- Division of Cardiothoracic SurgeryDepartment of SurgeryCarlyle Fraser Heart CenterEmory University School of MedicineAtlantaGA
| | - Rohini Polavarapu
- Division of Cardiothoracic SurgeryDepartment of SurgeryCarlyle Fraser Heart CenterEmory University School of MedicineAtlantaGA
| | - Yvanna Pantner
- Division of Cardiothoracic SurgeryDepartment of SurgeryCarlyle Fraser Heart CenterEmory University School of MedicineAtlantaGA
| | - Ahsan Husain
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| | - Nawazish Naqvi
- Division of CardiologyDepartment of MedicineEmory University School of MedicineAtlantaGA
| | - Lih‐Shen Chin
- Department PharmacologyEmory University School of MedicineAtlantaGA
| | - Lian Li
- Department PharmacologyEmory University School of MedicineAtlantaGA
| | - John W. Calvert
- Division of Cardiothoracic SurgeryDepartment of SurgeryCarlyle Fraser Heart CenterEmory University School of MedicineAtlantaGA
| |
Collapse
|
31
|
Molecular Mechanism Underlying Hypoxic Preconditioning-Promoted Mitochondrial Translocation of DJ-1 in Hypoxia/Reoxygenation H9c2 Cells. Molecules 2019; 25:molecules25010071. [PMID: 31878239 PMCID: PMC6983240 DOI: 10.3390/molecules25010071] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 12/17/2019] [Accepted: 12/20/2019] [Indexed: 01/06/2023] Open
Abstract
DJ-1 was recently reported to be involved in the cardioprotection of hypoxic preconditioning (HPC) against hypoxia/reoxygenation (H/R)-induced oxidative stress damage, by preserving mitochondrial complex I activity and, subsequently, inhibiting mitochondrial reactive oxygen species (ROS) generation. However, the molecular mechanism by which HPC enables mitochondrial translocation of DJ-1, which has no mitochondria-targeting sequence, to preserve mitochondrial complex I, is largely unknown. In this study, co-immunoprecipitation data showed that DJ-1 was associated with glucose-regulated protein 75 (Grp75), and this association was significantly enhanced after HPC. Immunofluorescence imaging and Western blot analysis showed that HPC substantially enhanced the translocation of DJ-1 from cytosol to mitochondria in H9c2 cells subjected to H/R, which was mimicked by DJ-1 overexpression induced by pFlag-DJ-1 transfection. Importantly, knockdown of Grp75 markedly reduced the mitochondrial translocation of DJ-1 induced by HPC and pFlag-DJ-1 transfection. Moreover, HPC promoted the association of DJ-1 with mitochondrial complex I subunits ND1 and NDUFA4, improved complex I activity, and inhibited mitochondria-derived ROS production and subsequent oxidative stress damage after H/R, which was also mimicked by pFlag-DJ-1 transfection. Intriguingly, these effects of HPC and pFlag-DJ-1 transfection were also prevented by Grp75 knockdown. In conclusion, these results indicated that HPC promotes the translocation of DJ-1 from cytosol to mitochondria in a Grp75-dependent manner and Grp75 is required for DJ-1-mediated protection of HPC on H/R-induced mitochondrial complex I defect and subsequent oxidative stress damage.
Collapse
|
32
|
Niacin and Selenium Attenuate Brain Injury After Cardiac Arrest in Rats by Up-Regulating DJ-1-Akt Signaling. Crit Care Med 2019; 46:e788-e796. [PMID: 29742581 DOI: 10.1097/ccm.0000000000003198] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVES To determine neuroprotective effects and mechanism of the combination therapy of niacin and selenium in cardiac arrest rats. DESIGN Prospective laboratory study. SETTING University laboratory. SUBJECTS Rat cortex neurons and male Sprague-Dawley rats (n = 68). INTERVENTIONS In rat cortex neurons underwent 90 minutes of oxygen-glucose deprivation and 22.5 hours of reoxygenation, effects of the combination therapy of niacin (0.9 mM) and selenium (1.5 μM) were investigated. The role of DJ-1 was determined using DJ-1 knockdown cells. In cardiac arrest rats, posttreatment effects of the combination therapy of niacin (360 mg/kg) and selenium (60 μg/kg) were evaluated. MEASUREMENTS AND MAIN RESULTS In oxygen-glucose deprivation and 22.5 hours of reoxygenation cells, combination therapy synergistically activated the glutathione redox cycle by a niacin-induced increase in glutathione reductase and a selenium-induced increase in glutathione peroxidase activities and reduced hydrogen peroxide level. It increased phosphorylated Akt and intranuclear Nuclear factor erythroid 2-related factor 2 expression and attenuated neuronal injury. However, these benefits were negated by DJ-1 knockdown. In cardiac arrest rats, combination therapy increased DJ-1, phosphorylated Akt, and intranuclear nuclear factor erythroid 2-related factor 2 expression, suppressed caspase 3 cleavage, and attenuated histologic injury in the brain tissues. It also improved the 7-day Neurologic Deficit Scales from 71.5 (66.0-74.0) to 77.0 (74.-80.0) (p = 0.02). CONCLUSIONS The combination therapy of clinically relevant doses of niacin and selenium attenuated brain injury and improved neurologic outcome in cardiac arrest rats. Its benefits were associated with reactive oxygen species reduction and subsequent DJ-1-Akt signaling up-regulation.
Collapse
|
33
|
Ong SB, Kwek XY, Katwadi K, Hernandez-Resendiz S, Crespo-Avilan GE, Ismail NI, Lin YH, Yap EP, Lim SY, Ja KPMM, Ramachandra CJA, Tee N, Toh JJ, Shim W, Wong P, Cabrera-Fuentes HA, Hausenloy DJ. Targeting Mitochondrial Fission Using Mdivi-1 in A Clinically Relevant Large Animal Model of Acute Myocardial Infarction: A Pilot Study. Int J Mol Sci 2019; 20:E3972. [PMID: 31443187 PMCID: PMC6720595 DOI: 10.3390/ijms20163972] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Revised: 08/09/2019] [Accepted: 08/14/2019] [Indexed: 12/19/2022] Open
Abstract
Background: New treatments are needed to reduce myocardial infarct size (MI) and prevent heart failure (HF) following acute myocardial infarction (AMI), which are the leading causes of death and disability worldwide. Studies in rodent AMI models showed that genetic and pharmacological inhibition of mitochondrial fission, induced by acute ischemia and reperfusion, reduced MI size. Whether targeting mitochondrial fission at the onset of reperfusion is also cardioprotective in a clinically-relevant large animal AMI model remains to be determined. Methods: Adult pigs (30-40 kg) were subjected to closed-chest 90-min left anterior descending artery ischemia followed by 72 h of reperfusion and were randomized to receive an intracoronary bolus of either mdivi-1 (1.2 mg/kg, a small molecule inhibitor of the mitochondrial fission protein, Drp1) or vehicle control, 10-min prior to reperfusion. The left ventricular (LV) size and function were both assessed by transthoracic echocardiography prior to AMI and after 72 h of reperfusion. MI size and the area-at-risk (AAR) were determined using dual staining with Tetrazolium and Evans blue. Heart samples were collected for histological determination of fibrosis and for electron microscopic analysis of mitochondrial morphology. Results: A total of 14 pigs underwent the treatment protocols (eight control and six mdivi-1). Administration of mdivi-1 immediately prior to the onset of reperfusion did not reduce MI size (MI size as % of AAR: Control 49.2 ± 8.6 vs. mdivi-1 50.5 ± 11.4; p = 0.815) or preserve LV systolic function (LV ejection fraction %: Control 67.5 ± 0.4 vs. mdivi-1 59.6 ± 0.6; p = 0.420), when compared to vehicle control. Similarly, there were no differences in mitochondrial morphology or myocardial fibrosis between mdivi-1 and vehicle control groups. Conclusion: Our pilot study has shown that treatment with mdivi-1 (1.2 mg/kg) at the onset of reperfusion did not reduce MI size or preserve LV function in the clinically-relevant closed-chest pig AMI model. A larger study, testing different doses of mdivi-1 or using a more specific Drp1 inhibitor are required to confirm these findings.
Collapse
Affiliation(s)
- Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore.
- Department of Cardiovascular, Renal and Metabolic Medicine, School of Medicine, Sapporo Medical University, Hokkaido 060-8543, Japan.
| | - Xiu-Yi Kwek
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Khairunnisa Katwadi
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Sauri Hernandez-Resendiz
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Gustavo E Crespo-Avilan
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
- Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany
| | - Nur Izzah Ismail
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
| | - Ying-Hsi Lin
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - En Ping Yap
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Song-Yi Lim
- Innoheart Pte Ltd., Singapore 119844, Singapore
| | - K P Myu Mai Ja
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Chrishan J A Ramachandra
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | - Nicole Tee
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
| | | | - Winston Shim
- Innoheart Pte Ltd., Singapore 119844, Singapore
- Health and Social Sciences Cluster, Singapore Institute of Technology, Singapore 138683, Singapore
| | - Philip Wong
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
- Innoheart Pte Ltd., Singapore 119844, Singapore
| | - Hector A Cabrera-Fuentes
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore.
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore.
- Institute of Biochemistry, Medical School, Justus-Liebig University, 35392 Giessen, Germany.
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, NL 64849, Mexico.
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, 420008 Kazan, Russian.
| | - Derek J Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, Singapore 169857, Singapore
- National Heart Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore
- Tecnologico de Monterrey, Centro de Biotecnologia-FEMSA, Monterrey, NL 64849, Mexico
- Yong Loo Lin School of Medicine, National University Singapore, Singapore 119228, Singapore
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London WC1E 6HX, UK
- The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London W1T 7DN, UK
| |
Collapse
|
34
|
Mantri S, Morley JF, Siderowf AD. The importance of preclinical diagnostics in Parkinson disease. Parkinsonism Relat Disord 2019; 64:20-28. [DOI: 10.1016/j.parkreldis.2018.09.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Revised: 08/02/2018] [Accepted: 09/08/2018] [Indexed: 01/21/2023]
|
35
|
Essack M, Salhi A, Stanimirovic J, Tifratene F, Bin Raies A, Hungler A, Uludag M, Van Neste C, Trpkovic A, Bajic VP, Bajic VB, Isenovic ER. Literature-Based Enrichment Insights into Redox Control of Vascular Biology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:1769437. [PMID: 31223421 PMCID: PMC6542245 DOI: 10.1155/2019/1769437] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Revised: 04/11/2019] [Accepted: 05/02/2019] [Indexed: 02/07/2023]
Abstract
In cellular physiology and signaling, reactive oxygen species (ROS) play one of the most critical roles. ROS overproduction leads to cellular oxidative stress. This may lead to an irrecoverable imbalance of redox (oxidation-reduction reaction) function that deregulates redox homeostasis, which itself could lead to several diseases including neurodegenerative disease, cardiovascular disease, and cancers. In this study, we focus on the redox effects related to vascular systems in mammals. To support research in this domain, we developed an online knowledge base, DES-RedoxVasc, which enables exploration of information contained in the biomedical scientific literature. The DES-RedoxVasc system analyzed 233399 documents consisting of PubMed abstracts and PubMed Central full-text articles related to different aspects of redox biology in vascular systems. It allows researchers to explore enriched concepts from 28 curated thematic dictionaries, as well as literature-derived potential associations of pairs of such enriched concepts, where associations themselves are statistically enriched. For example, the system allows exploration of associations of pathways, diseases, mutations, genes/proteins, miRNAs, long ncRNAs, toxins, drugs, biological processes, molecular functions, etc. that allow for insights about different aspects of redox effects and control of processes related to the vascular system. Moreover, we deliver case studies about some existing or possibly novel knowledge regarding redox of vascular biology demonstrating the usefulness of DES-RedoxVasc. DES-RedoxVasc is the first compiled knowledge base using text mining for the exploration of this topic.
Collapse
Affiliation(s)
- Magbubah Essack
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Adil Salhi
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Julijana Stanimirovic
- Vinca Institute, University of Belgrade, Laboratory for Molecular Endocrinology and Radiobiology, Belgrade, Serbia
| | - Faroug Tifratene
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Arwa Bin Raies
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Arnaud Hungler
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Mahmut Uludag
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Christophe Van Neste
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Andreja Trpkovic
- Vinca Institute, University of Belgrade, Laboratory for Molecular Endocrinology and Radiobiology, Belgrade, Serbia
| | - Vladan P. Bajic
- Vinca Institute, University of Belgrade, Laboratory for Molecular Endocrinology and Radiobiology, Belgrade, Serbia
| | - Vladimir B. Bajic
- King Abdullah University of Science and Technology, Computational Bioscience Research Center, Thuwal, Saudi Arabia
| | - Esma R. Isenovic
- Vinca Institute, University of Belgrade, Laboratory for Molecular Endocrinology and Radiobiology, Belgrade, Serbia
| |
Collapse
|
36
|
Yin J, Xu R, Wei J, Zhang S. The protective effect of glutaredoxin 1/DJ-1/HSP70 signaling in renal tubular epithelial cells injury induced by ischemia. Life Sci 2019; 223:88-94. [PMID: 30858124 DOI: 10.1016/j.lfs.2019.03.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 03/07/2019] [Indexed: 12/20/2022]
Abstract
AIMS Gluaredoxin1 (GRX1) is an important protein of the cellular antioxidant defense system, but its role in renal epithelial cell injury caused by ischemia remains unclear. In this study, we aimed to gain insight into the role of GRX1 in HK-2 cells with oxygen glucose deprivation (OGD) injury, which served as an in vitro cell model of renal epithelial cell ischemic injury. We investigated the underlying regulation of GRX1, DJ-1, and HSP70 as well as the role of the GRX1/DJ-1/HSP70 signaling pathway in this model. MATERIALS AND METHODS The protein and mRNA expressions were measured by Western blot and qRT-PCR assays, respectively. GRX1 was overexpressed by transfection of pcDNA.3.1-GRX1 and DJ-1 was inhibited by transfection with DJ-1 siRNA. Cell apoptosis, caspase-3 activity, lactate dehydrogenase (LDH) leakage, or superoxide dismutase (SOD) content was tested by the related detection kit. Reactive oxygen species (ROS) level was detected via carboxy-H2DCF-DA. KEY FINDINGS We found that GRX1 was distinctly down-regulated in HK-2 cells after incubation under the OGD condition. GRX1 overexpression markedly constrained cell apoptosis, caspase-3 activity, LDH leakage, and the ROS level, while SOD content was elevated. GRX1 up-regulation increased DJ-1 and HSP70 protein expression, while DJ-1 inhibition significantly offset the effect of GRX1 overexpression on HSP70, indicating that GRX1 could regulate HSP70 via control of DJ-1. Moreover, we observed that HSP70 inhibition removed the constraints imposed by GRX1 overexpression on ROS level, LDH leakage, and caspase-3 activity. SIGNIFICANCE Overall, this study showed that GRX1 minimizes cell injury and apoptosis in HK-2 cells under OGD conditions via regulation of DJ-1 and HSP70 expression.
Collapse
Affiliation(s)
- Jian Yin
- Department of Vascular Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Ruisi Xu
- Endoscopy Center, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Jun Wei
- Department of Pharmacology Base, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Siqi Zhang
- Department of Nephrology, China-Japan Union Hospital of Jilin University, Changchun 130033, China.
| |
Collapse
|
37
|
Katunina EA, Ilina EP, Sadekhova GA, Gaisenuk EI. Approaches to early diagnosis of Parkinson's disease. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:119-127. [DOI: 10.17116/jnevro2019119061119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
38
|
Cheng Y, Marion TN, Cao X, Wang W, Cao Y. Park 7: A Novel Therapeutic Target for Macrophages in Sepsis-Induced Immunosuppression. Front Immunol 2018; 9:2632. [PMID: 30542343 PMCID: PMC6277877 DOI: 10.3389/fimmu.2018.02632] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 10/25/2018] [Indexed: 02/05/2023] Open
Abstract
Sepsis remains a serious and life-threatening condition with high morbidity and mortality due to uncontrolled inflammation together with immunosuppression with few therapeutic options. Macrophages are recognized to play essential roles throughout all phases of sepsis and affect both immune homeostasis and inflammatory processes, and macrophage dysfunction is considered to be one of the major causes for sepsis-induced immunosuppression. Currently, Parkinson disease protein 7 (Park 7) is known to play an important role in regulating the production of reactive oxygen species (ROS) through interaction with p47phox, a subunit of NADPH oxidase. ROS are key mediators in initiating toll-like receptor (TLR) signaling pathways to activate macrophages. Emerging evidence has strongly implicated Park 7 as an antagonist for sepsis-induced immunosuppression, which suggests that Park 7 may be a novel therapeutic target for reversing immunosuppression compromised by sepsis. Here, we review the main characteristics of sepsis-induced immunosuppression caused by macrophages and provide a detailed mechanism for how Park 7 antagonizes sepsis-induced immunosuppression initiated by the macrophage inflammatory response. Finally, we further discuss the most promising approach to develop innovative drugs that target Park 7 in patients whose initial presentation is at the late stage of sepsis.
Collapse
Affiliation(s)
- Yanwei Cheng
- West China Hospital Emergency Department, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, China.,Disaster Medicine Center, Sichuan University, Chengdu, China
| | - Tony N Marion
- Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China.,Department of Microbiology, Immunology, and Biochemistry, The University of Tennessee Health Science Center, Memphis, TN, United States
| | - Xue Cao
- Disaster Medicine Center, Sichuan University, Chengdu, China.,Department of Rheumatology and Immunology, West China Hospital, Sichuan University, Chengdu, China
| | - Wanting Wang
- West China Hospital Emergency Department, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, China
| | - Yu Cao
- West China Hospital Emergency Department, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center of Biotherapy, Chengdu, China.,Disaster Medicine Center, Sichuan University, Chengdu, China
| |
Collapse
|
39
|
Abstract
AbstractBackground: The relationship between vascular disease and Parkinson’s disease (PD) is controversial. We performed a cross-sectional study to investigate the association of two common vascular diseases (stroke and coronary artery disease [CAD]) with Parkinson’s disease. Methods: We identified 63 and 62 PD cases in two population-based cohorts (Malu rural community and Wuliqiao urban community) and collected information of PD and non-PD by means of questionnaires. Logistic regression analysis was used to investigate the association between stroke, coronary artery disease and PD, after adjusting for age, sex, hypertension, diabetes mellitus, hypercholesterolemia, smoking status, alcohol consumption, tea consumption and body mass index. Results: After adjustment for potential confounders, we found that CAD and stroke were associated with PD in the Malu rural community (CAD: odds ratio [OR]=7.11, 95% confidence intervals [CI]: 3.09-16.40, p<0.001; stroke: OR=6.77, 95% CI: 3.09-14.81, p<0.001) and stroke was associated with PD in the Wuliqiao urban community (OR=2.58, 95% CI: 1.36-4.89, p=0.004), especially in women. In a subgroup analysis of PD with age- and sex-matched controls, the results were similar in the Malu rural community (CAD: OR=12.72, 95% CI: 2.92-55.32, p=0.001; stroke: OR=6.26, 95% CI: 1.83-21.42, p=0.003), whereas in the Wuliqiao urban community the results were different in that CAD (but not stroke) was found to be associated with PD (CAD: OR=2.44, 95% CI: 1.09-5.47, p=0.03; stroke: OR=1.79, 95% CI: 0.77-4.17, p=0.18). Conclusions: Our study suggested that stroke and CAD are associated with PD in two Chinese population-based cohorts, indicating a probable vascular component in the pathogenesis of PD.
Collapse
|
40
|
Neuronal Preconditioning Requires the Mitophagic Activity of C-terminus of HSC70-Interacting Protein. J Neurosci 2018; 38:6825-6840. [PMID: 29934347 DOI: 10.1523/jneurosci.0699-18.2018] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 06/08/2018] [Accepted: 06/13/2018] [Indexed: 11/21/2022] Open
Abstract
The C terminus of HSC70-interacting protein (CHIP, STUB1) is a ubiquitously expressed cytosolic E3-ubiquitin ligase. CHIP-deficient mice exhibit cardiovascular stress and motor dysfunction before premature death. This phenotype is more consistent with animal models in which master regulators of autophagy are affected rather than with the mild phenotype of classic E3-ubiquitin ligase mutants. The cellular and biochemical events that contribute to neurodegeneration and premature aging in CHIP KO models remain poorly understood. Electron and fluorescent microscopy demonstrates that CHIP deficiency is associated with greater numbers of mitochondria, but these organelles are swollen and misshapen. Acute bioenergetic stress triggers CHIP induction and relocalization to mitochondria, where it plays a role in the removal of damaged organelles. This mitochondrial clearance is required for protection following low-level bioenergetic stress in neurons. CHIP expression overlaps with stabilization of the redox stress sensor PTEN-inducible kinase 1 (PINK1) and is associated with increased LC3-mediated mitophagy. Introducing human promoter-driven vectors with mutations in either the E3 ligase or tetracopeptide repeat domains of CHIP in primary neurons derived from CHIP-null animals enhances CHIP accumulation at mitochondria. Exposure to autophagy inhibitors suggests that the increase in mitochondrial CHIP is likely due to diminished clearance of these CHIP-tagged organelles. Proteomic analysis of WT and CHIP KO mouse brains (four male, four female per genotype) reveals proteins essential for maintaining energetic, redox, and mitochondrial homeostasis undergo significant genotype-dependent expression changes. Together, these data support the use of CHIP-deficient animals as a predictive model of age-related degeneration with selective neuronal proteotoxicity and mitochondrial failure.SIGNIFICANCE STATEMENT Mitochondria are recognized as central determinants of neuronal function and survival. We demonstrate that C terminus of HSC70-Interacting Protein (CHIP) is critical for neuronal responses to stress. CHIP upregulation and localization to mitochondria is required for mitochondrial autophagy (mitophagy). Unlike other disease-associated E3 ligases such as Parkin and Mahogunin, CHIP controls homeostatic and stress-induced removal of mitochondria. Although CHIP deletion results in greater numbers of mitochondria, these organelles have distorted inner membranes without clear cristae. Neuronal cultures derived from animals lacking CHIP are more vulnerable to acute injuries and transient loss of CHIP renders neurons incapable of mounting a protective response after low-level stress. Together, these data suggest that CHIP is an essential regulator of mitochondrial number, cell signaling, and survival.
Collapse
|
41
|
Ghaderi S, Alidadiani N, SoleimaniRad J, Heidari HR, Dilaver N, Heim C, Ramsperger-Gleixner M, Baradaran B, Weyand M. DJ1 and microRNA-214 act synergistically to rescue myoblast cells after ischemia/reperfusion injury. J Cell Biochem 2018; 119:7192-7203. [PMID: 29806880 DOI: 10.1002/jcb.26842] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 03/09/2018] [Indexed: 12/18/2022]
Abstract
Ischemia/reperfusion injury is a tissue injury occurring post-reperfusion of tissues with pre-existing ischemia. A good blood supply to tissues aids in the survival of ischemic tissue, however, due to prolonged ischemia the levels of ATP decrease and pH declines leading to acidosis. Reduced ATP leads to an increase in the AMP/ATP ratio, causing cessation of intracellular calcium transport, hence calcium overload and cell death. In this study, we demonstrate the synergistic and antagonistic effect of DJ1 and microR-214 (miR-214) in rescuing myoblast C2C12 cells after ischemia/reperfusion in an in vitro model. Both DJ1 and miR-214 were cloned into a hypoxic inducible expression cassette and transfected into the C2C12 cells. We showed that DJ1 and miR-214 have synergistic effects in reducing intracellular lactate dehydrogenase and intracellular transient calcium levels after reoxygenation compared to control cells, in addition to reducing cell death via necrosis. Western blotting revealed a decrease in autophagosome formation in LC3II/I ratio and an increase in AKT expression in cells transfected with DJ1 and miR-214. Using quantitative real-time PCR, we demonstrated that DJ1 and miR-214 significantly reduced the expression of pro-apoptotic factors and autophagy compared to control. The results indicated DJ1 is an endogenous oxidative stress molecule and miR-214 is a potent inhibitor of the sodium calcium exchanger channel. DJ1 had the greatest effect to inhibiting mitochondrial cell death pathways by possibly acting as a modulator of autophagy. Additionally, we have concluded that miR-214 has an inhibitory effect on extrinsic cell death pathways such as necrosis and autophagy.
Collapse
Affiliation(s)
- Shahrooz Ghaderi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Advanced Medical Sciences, Department of Molecular Medicine, Tabriz, Iran
| | - Neda Alidadiani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hamid R Heidari
- Faculty of Pharmacy, Department of Pharmaceutical Biotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nafi Dilaver
- Swansea University Medical School, Swansea University, Swansea, UK
| | - Christian Heim
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| | | | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael Weyand
- Department of Cardiac Surgery, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
42
|
Leak RK. Conditioning Against the Pathology of Parkinson's disease. CONDITIONING MEDICINE 2018; 1:143-162. [PMID: 30370426 PMCID: PMC6200356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Parkinson's disease is delayed in clinical onset, asymmetric in initial appearance, and slow in progression. One explanation for these characteristics may be a boost in natural defenses after early exposure to mild cellular stress. As the patient ages and resilience recedes, however, stress levels may become sufficiently high that toxic cellular responses can no longer be curbed, culminating in inverted U-shaped stress-response curves as a function of disease duration. If dopaminergic systems are indeed capable of responding to mild stress with effective natural defenses, experimental models of Parkinson's disease should adhere to the principles of preconditioning, whereby stress exposure fortifies cells and tempers the toxic sequelae of subsequent stressors. Here, I review evidence favoring the efficacy of preconditioning in dopaminergic systems. Recent animal work also raises the possibility that cross-hemispheric preconditioning may arrest the spread of asymmetric Parkinson's pathology to the other side of the brain. Indeed, compensatory homeostatic systems have long been hypothesized to maintain neurological function until a threshold of cell loss is exceeded and are often displayed as inverted U-shaped curves. However, some stress responses assume an exponential or sigmoidal profile as a function of disease severity, suggesting end-stage deceleration of disease processes. Thus, surviving dopaminergic neurons may become progressively harder to kill, with the dorsal nigral tier dying slower due to superior baseline defenses, inducible conditioning capacity, or delayed dorsomedial nigral spread of disease. In addition, compensatory processes may be useful as biomarkers to distinguish "responder patients" from "nonresponders" before clinical trials. However, another possibility is that defenses are already maximally conditioned in most patients and no further boost is possible. A third alternative is that genuinely diseased human cells cannot be conditioned, in contrast to preclinical models, none of which faithfully recapitulate age-related human conditions. Disease-related "conditioning deficiencies" would then explain how Parkinson's pathology takes root, progressively shrinks defenses, and eventually kills the patient.
Collapse
Affiliation(s)
- Rehana K. Leak
- For correspondence please address: Rehana K. Leak,
Ph.D., Graduate School of Pharmaceutical Sciences, Duquesne University, 600
Forbes Ave, Pittsburgh, PA 15282, ,
412.396.4734
| |
Collapse
|
43
|
Ammal Kaidery N, Thomas B. Current perspective of mitochondrial biology in Parkinson's disease. Neurochem Int 2018; 117:91-113. [PMID: 29550604 DOI: 10.1016/j.neuint.2018.03.001] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/05/2018] [Accepted: 03/06/2018] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative movement disorder characterized by preferential loss of dopaminergic neurons of the substantia nigra pars compacta and the presence of Lewy bodies containing α-synuclein. Although the cause of PD remains elusive, remarkable advances have been made in understanding the possible causative mechanisms of PD pathogenesis. An explosion of discoveries during the past two decades has led to the identification of several autosomal dominant and recessive genes that cause familial forms of PD. The investigations of these familial PD gene products have shed considerable insights into the molecular pathogenesis of the more common sporadic PD. A growing body of evidence suggests that the etiology of PD is multifactorial and involves a complex interplay between genetic and environmental factors. Substantial evidence from human tissues, genetic and toxin-induced animal and cellular models indicates that mitochondrial dysfunction plays a central role in the pathophysiology of PD. Deficits in mitochondrial functions due to bioenergetics defects, alterations in the mitochondrial DNA, generation of reactive oxygen species, aberrant calcium homeostasis, and anomalies in mitochondrial dynamics and quality control are implicated in the underlying mechanisms of neuronal cell death in PD. In this review, we discuss how familial PD-linked genes and environmental factors interface the pathways regulating mitochondrial functions and thereby potentially converge both familial and sporadic PD at the level of mitochondrial integrity. We also provide an overview of the status of therapeutic strategies targeting mitochondrial dysfunction in PD. Unraveling potential pathways that influence mitochondrial homeostasis in PD may hold the key to therapeutic intervention for this debilitating neurodegenerative movement disorder.
Collapse
Affiliation(s)
| | - Bobby Thomas
- Departments of Pharmacology and Toxicology, Augusta, GA 30912, United States; Neurology Medical College of Georgia, Augusta University, Augusta, GA 30912, United States.
| |
Collapse
|
44
|
Li Y, Liu X. Novel insights into the role of mitochondrial fusion and fission in cardiomyocyte apoptosis induced by ischemia/reperfusion. J Cell Physiol 2018. [PMID: 29528108 DOI: 10.1002/jcp.26522] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
As the main source of energy in the body, mitochondria are highly dynamic organelles, which are constantly going through fusion and fission. The fine balance of mitochondrial fusion and fission plays an important role in maintaining the stability of cardiomyocyte homeostasis. The processes of mitochondrial fusion and fission are very complex, which is mediated by fusion and fission proteins. Disruptions in these processes through controlling fusion and fission proteins affect mitochondrial functions and cardiomyocyte survival. Ischemia/reperfusion (I/R) can regulate the expression and post-translational modifications of fusion and fission proteins thereby inducing the abnormality of mitochondrial fusion and fission and cardiomyocyte apoptosis. Furthermore, intervention with the expression and function of fusion and fission proteins influences on cardiomyocyte apoptosis under I/R conditions. In this review, we focus on the current developments in the effects of mitochondrial fusion and fission on cardiomyocyte functions, the implications for cardiomyocyte apoptosis in response to I/R, and possible mechanisms. And we review their roles as a potential therapeutic target for treating I/R-induced cardiomyocyte injury.
Collapse
Affiliation(s)
- YuZhen Li
- Department of Pathophysiology, Institute of Basic Medical Science, PLA General Hospital, Beijing, China
| | - XiuHua Liu
- Department of Pathophysiology, Institute of Basic Medical Science, PLA General Hospital, Beijing, China
| |
Collapse
|
45
|
Cistanche deserticola polysaccharides protects PC12 cells against OGD/RP-induced injury. Biomed Pharmacother 2018; 99:671-680. [PMID: 29710464 DOI: 10.1016/j.biopha.2018.01.114] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Revised: 01/19/2018] [Accepted: 01/24/2018] [Indexed: 12/25/2022] Open
Abstract
Ischemia stroke is a disease with high morbidity and mortality. Cistanche deserticola polysaccharides (CDP) possess a wide range of beneficial effects, including hepatoprotection and immune homeostasis. As far as we know, the protective effect of CDP on neurons injured by oxygen-glucose deprivation/reperfusion (OGD/RP) has not been investigated. In this study, OGD/RP injured a PC12 cell model. Briefly, CDP (0.05, 0.5 and 5??g/ml) was administered before reperfusion. The protective effect of CDP was then evaluated on the basis of cell viability, lactate dehydrogenase (LDH) leakage, [Ca2+]i, mitochondrial membrane potential (MMP)and cell apoptosis, and redox status after reperfusion was evaluated by assaying reactive oxygen species (ROS), catalase (CAT), glutathione peroxidase (GSH-Px) and total antioxidant capacity. Basing on the fact that Parkinson's disease-associated protein DJ-1 participates in endogenous antioxidation and performs neuroprotective effects after ischemia stroke, we investigated the interaction between CDP and DJ-1. DJ-1 expression was detected through ELISA and Western blot analysis, and the translocation of DJ-1 was evaluated through immunofluorescence. Result showed that CDP (0.05, 0.5 and 5??g/ml) attenuated PC12 cell death, preserved MMP and calcium homeostasis; inhibited oxidative stress and decreased cell apoptosis. Moreover, CDP (5??g/ml) markedly stimulated DJ-1 secretion and expression. Overall, the results suggested that CDP exerts neuroprotective effect against OGD/RP-induced injury by inhibiting oxidative stress and regulating the DJ-1 pathway.
Collapse
|
46
|
Ding H, Xu XW, Wang H, Xiao L, Zhao L, Duan GL, Li XR, Ma ZX, Chen HP. DJ-1 plays an obligatory role in the cardioprotection of delayed hypoxic preconditioning against hypoxia/reoxygenation-induced oxidative stress through maintaining mitochondrial complex I activity. Cell Biochem Funct 2018; 36:147-154. [DOI: 10.1002/cbf.3326] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 01/13/2018] [Accepted: 01/22/2018] [Indexed: 11/10/2022]
Affiliation(s)
- Hao Ding
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Xing-Wang Xu
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Huan Wang
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Lin Xiao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Le Zhao
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Guang-Ling Duan
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Xiao-Ran Li
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - Zhao-Xia Ma
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| | - He-Ping Chen
- The Key Laboratory of Basic Pharmacology, School of Pharmaceutical Science; Nanchang University; Nanchang People's Republic of China
| |
Collapse
|
47
|
Han T, Liu M, Yang S. DJ-1 Alleviates Angiotensin II-Induced Endothelial Progenitor Cell Damage by Activating the PPARγ/HO-1 Pathway. J Cell Biochem 2018; 119:392-400. [PMID: 28600848 DOI: 10.1002/jcb.26191] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2017] [Accepted: 06/05/2017] [Indexed: 01/06/2023]
Abstract
There is evidence that angiotensin II (Ang II) may impair the functions of endothelial progenitor cells (EPCs). It was revealed that DJ-1 could resist oxidative stress. In this study, we investigated whether DJ-1 could protect EPCs against Ang II-induced cell damage. The proliferation and migration of EPCs were strongly reduced in the Ang II group and were increased by overexpression of DJ-1. Western blotting indicated that the increased expression of the senescence marker β-galactosidase and decreased expression of adhesion molecules (ICAM-1, VCAM-1) induced by Ang II were reversed after Ad-DJ-1 transfection. The reduced angiogenic capacity of EPCs caused by Ang II was also improved after Ad-DJ-1 transfection. Moreover, Ang II significantly increased the levels of reactive oxygen species (ROS), malondialdehyde (MDA), and inflammatory cytokines (TNF-α and IL-1β), reduced the levels of superoxide dismutase (SOD), glutathione (GSH), and these were reversed by Ad-DJ-1 transfection. Expression of peroxisome proliferator-activated receptor-γ (PPARγ) and heme oxygenase (HO-1) was increased by DJ-1. Therefore, HO-1 siRNA were constructed and transfected into EPCs, and the results showed that HO-1 siRNA transfection inhibited the effects of DJ-1 on EPC function. Thus, our study implies that DJ-1 may protect EPCs against Ang II-induced dysfunction by activating the PPARγ/HO-1. J. Cell. Biochem. 119: 392-400, 2018. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Tao Han
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033,, Jilin, China
| | - Meihan Liu
- Department of Ultrasonography, China-Japan Union Hospital, Jilin University, Changchun, 130033,, Jilin, China
| | - Songbai Yang
- Department of Vascular Surgery, China-Japan Union Hospital, Jilin University, Changchun, 130033,, Jilin, China
| |
Collapse
|
48
|
Tian H, Li S, Yu K. DJ‑1 alleviates high glucose‑induced endothelial cells injury via PI3K/Akt‑eNOS signaling pathway. Mol Med Rep 2017; 17:1205-1211. [PMID: 29115508 DOI: 10.3892/mmr.2017.7975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2017] [Accepted: 10/13/2017] [Indexed: 11/06/2022] Open
Abstract
Hyperglycemia mediated endothelial cells (ECs) injury is closely associated with diabetic vascular complications. It was revealed that DJ‑1 possesses cellular protective effects by suppressing oxidative stress. The present study aimed to investigate the beneficial effects of DJ‑1 on high glucose (HG)‑induced human umbilical vein endothelial cell (HUVEC) injury and to elucidate its underlying mechanisms. HUVECs were incubated under 5.5 mM (control group) or 25 mM D‑glucose (HG group) and then transfected with recombinant adenoviral vectors to overexpression of DJ‑1. Cell proliferation and apoptosis were measured using the EdU incorporation assay and flow cytometry with Annexin V-FITC/propidium iodide double staining, respectively. Apoptotic‑related proteins were determined using western blot analysis. Reactive oxygen species (ROS) production, lactate dehydrogenase (LDH) and nitric oxide (NO) levels, the content of malondialdehyde (MDA), and the activities of superoxide dismutase (SOD) were measured. Results demonstrated that overexpression of DJ‑1 promoted cell proliferation and inhibited HUVECs apoptosis stimulated by HG. DJ‑1 also suppressed the HG‑induced reduction in the Bcl‑2/Bax ratio and HG activated ROS generation in HUVECs. Furthermore, HG significantly increased the levels of LDH and MDA, and reduced the level of SOD; however, these effects were reversed by Ad‑DJ‑1 transfection. Furthermore, the cellular protective effect of overexpression of DJ‑1 enhanced p‑Akt/Akt ratio, eNOS activation and NO production, and these trends were partially reversed by a phosphatidylinositol‑4,5‑bisphosphate 3‑kinase (PI3K) inhibitor (LY294002). Taken together, the present study highlighted the involvement of DJ‑1 in HG‑related EC injury and identified that DJ‑1 exerts a cellular protective effect in HUVECs exposed to HG induced oxidative stress via activation of the PI3K/Akt‑eNOS signaling pathway.
Collapse
Affiliation(s)
- Hongan Tian
- Radiology Department, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Shunzhen Li
- Radiology Department, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| | - Kaihu Yu
- Radiology Department, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, Hubei 437100, P.R. China
| |
Collapse
|
49
|
Xue R, Jiang J, Dong B, Tan W, Sun Y, Zhao J, Chen Y, Dong Y, Liu C. DJ-1 activates autophagy in the repression of cardiac hypertrophy. Arch Biochem Biophys 2017; 633:124-132. [PMID: 28941803 DOI: 10.1016/j.abb.2017.09.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 09/10/2017] [Accepted: 09/18/2017] [Indexed: 01/01/2023]
Abstract
Cardiac hypertrophy is the risk factor of heart failure when the heart is confronted with pressure overload or neurohumoral stimuli. Autophagy, a conserved degradative pathway, is one of the important mechanisms involved in the regulation of cardiac hypertrophy. DJ-1 is a traditional anti-oxidative protein and emerging evidence suggested that DJ-1 might modulate autophagy. However, the regulation of autophagy by DJ-1 in the process of cardiac hypertrophy remains unknown. In our study, we firstly discovered that the expression of DJ-1declined in the process of pressure overload cardiac hypertrophy, and its alteration was parallel with the impairment of autophagy. Furthermore, we proved that DJ-1 knockout mice exhibited a more hypertrophied phenotype than wildtype mice in cardiac hypertrophy which indicated that DJ-1 is responsible for the repression of cardiac hypertrophy. Furthermore, DJ-1 knockout significantly exacerbated pulmonary edema due to cardiac hypertrophy. In the process of cardiac hypertrophy, DJ-1 knockout significantly impaired autophagy activation and enhanced mTORC1 and mTORC2 phosphorylation were found. Similarly, our in vitro study proved that DJ-1 overexpression ameliorated phenylephrine (PE)-induced cardiac hypertrophy and promoted autophagy activation. Taken together, DJ-1 might repress both pressure overload and PE-induced cardiac hypertrophy via the activation of autophagy.
Collapse
Affiliation(s)
- Ruicong Xue
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Jingzhou Jiang
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Bin Dong
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Weiping Tan
- Department of Respiratory, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yu Sun
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Jingjing Zhao
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Yili Chen
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China
| | - Yugang Dong
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China.
| | - Chen Liu
- Department of Cardiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China; Key Laboratory on Assisted Circulation, Ministry of Health, Guangzhou, China.
| |
Collapse
|
50
|
Yang RX, Lei J, Wang BD, Feng DY, Huang L, Li YQ, Li T, Zhu G, Li C, Lu FF, Nie TJ, Gao GD, Gao L. Pretreatment with Sodium Phenylbutyrate Alleviates Cerebral Ischemia/Reperfusion Injury by Upregulating DJ-1 Protein. Front Neurol 2017. [PMID: 28649223 PMCID: PMC5465296 DOI: 10.3389/fneur.2017.00256] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Oxidative stress and mitochondrial dysfunction play critical roles in ischemia/reperfusion (I/R) injury. DJ-1 is an endogenous antioxidant that attenuates oxidative stress and maintains mitochondrial function, likely acting as a protector of I/R injury. In the present study, we explored the protective effect of a possible DJ-1 agonist, sodium phenylbutyrate (SPB), against I/R injury by protecting mitochondrial dysfunction via the upregulation of DJ-1 protein. Pretreatment with SPB upregulated the DJ-1 protein level and rescued the I/R injury-induced DJ-1 decrease about 50% both in vivo and in vitro. SPB also improved cellular viability and mitochondrial function and alleviated neuronal apoptosis both in cell and animal models; these effects of SPB were abolished by DJ-1 knockdown with siRNA. Furthermore, SPB improved the survival rate about 20% and neurological functions, as well as reduced about 50% of the infarct volume and brain edema, of middle cerebral artery occlusion mice 23 h after reperfusion. Therefore, our findings demonstrate that preconditioning of SPB possesses a neuroprotective effect against cerebral I/R injury by protecting mitochondrial function dependent on the DJ-1 upregulation, suggesting that DJ-1 is a potential therapeutic target for clinical ischemic stroke.
Collapse
Affiliation(s)
- Rui-Xin Yang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jie Lei
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Bo-Dong Wang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Da-Yun Feng
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lu Huang
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yu-Qian Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Tao Li
- Research Center of Traditional Chinese Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Gang Zhu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Chen Li
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Fang-Fang Lu
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Tie-Jian Nie
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Guo-Dong Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Li Gao
- Department of Neurosurgery, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| |
Collapse
|