1
|
Di Mattía RA, Gallo D, Ciarrocchi S, Gonano LA, Blanco PG, Valverde CA, Portiansky EL, Sommese LM, Toischer K, Bleckwedel F, Zelarayán LC, Aiello EA, Orlowski A. Cardiac hypertrophy induced by overexpression of IP3-released inositol 1, 4, 5-trisphosphate receptor-binding protein (IRBIT). J Mol Cell Cardiol 2025; 201:1-15. [PMID: 39929439 DOI: 10.1016/j.yjmcc.2025.02.001] [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: 03/06/2024] [Revised: 01/22/2025] [Accepted: 02/06/2025] [Indexed: 03/22/2025]
Abstract
INTRODUCTION IRBIT, also known as Ahcyl1, is an IP3 receptor (IP3R)-binding protein released with IP3 and was first described as a competitive inhibitor of the mentioned receptor. Studies have shown that overexpression of IP3Rs is associated with cardiac hypertrophy in both human and animal models. Given that IP3Rs play a role in cardiac hypertrophy, IRBIT may also be involved in this condition. AIM Although IRBIT heart expression has been reported, its function in cardiac tissues remains unclear. Thus, we aimed to study the cardiac outcomes of up-and downregulation of IRBIT to establish its pathophysiological role. METHODS AND RESULTS We found that IRBIT is expressed in mouse ventricles and atria, fibroblasts and cardiomyocytes isolated from neonatal mice, and in the myoblast cell line H9c2. Mice with transverse aortic constriction showed a significant increase in both the mRNA and protein expression of IRBIT. Furthermore, we described the differential expression of IRBIT in human myocardial samples of dilated and ischemic cardiomyopathy. IRBIT cardiac overexpression in mice using an adenoassociated virus (AAV9) at two different time points (neonatal mice, day 4 and adult mice, 3 months) resulted in the development of cardiac hypertrophy with impaired systolic function by four months of age. A decrease in the mRNA levels of the IP3 receptor was also observed in both models. Isolated myocytes from the IRBIT-overexpressing neonatal model showed a significantly decreased Ca2+ transient amplitude and slower rise of the global Ca2+ transient, without changes in sarcoplasmic reticulum (SR) Ca2+ content or spontaneous Ca2+ wave frequency. However, the velocity of Ca2+ wave propagation was reduced. Moreover, we found that the dyssynchrony index (DI) is significantly increased under IRBIT overexpression. Nuclear Ca2+ dynamics were assessed, showing no significant changes, but IRBIT overexpression reduced the number of nuclear envelope invaginations. In addition, reducing IRBIT expression using AAV9-shRNA did not result in any changes in the heart morphometric parameters. CONCLUSION Our study describes for the first time that IRBIT plays a critical role in the pathophysiology of the heart. Our findings demonstrate that IRBIT overexpression disrupts Ca2+ signaling, contributing to hypertrophic remodeling and impaired cardiac function. The altered wave propagation, the increase in DI and the decrease of the rate of the Ca2+ transient suggests that IRBIT influences Ca2+ - induced Ca2+ release. This study provides the first evidence linking IRBIT to pathological cardiac remodeling and Ca2+ handling dysregulation. Although significant progress has been made, further research is required to better understand the cardiovascular function of IRBIT and its mechanisms.
Collapse
Affiliation(s)
- R A Di Mattía
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - D Gallo
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - S Ciarrocchi
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - L A Gonano
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - P G Blanco
- Centro de Fisiología Reproductiva & Métodos Complementarios de Diagnóstico (CEFIRE & MECODIAG), Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - C A Valverde
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - E L Portiansky
- Laboratorio de Análisis de Imágenes, Facultad de Ciencias Veterinarias, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - L M Sommese
- Departamento de Ciencia y Tecnología, CONICET, Universidad Nacional de Quilmes, Bernal, Argentina
| | - K Toischer
- German Center for Cardiovascular Research (DZHK) Partner Site, 37075 Goettingen, Germany; Clinic for Cardiology and Pneumology, University Medical Center, Göttingen, Germany
| | - F Bleckwedel
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen (UMG), 37075 Goettingen, Germany; German Center for Cardiovascular Research (DZHK) Partner Site, 37075 Goettingen, Germany
| | - L C Zelarayán
- Institute of Pharmacology and Toxicology, University Medical Center Goettingen (UMG), 37075 Goettingen, Germany; German Center for Cardiovascular Research (DZHK) Partner Site, 37075 Goettingen, Germany; Justus Liebig University, Medical Clinic I, Department of Cardiology and Angiology, Klinikstraße 33, 35392 Giessen, Germany
| | - E A Aiello
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina
| | - A Orlowski
- Centro de Investigaciones Cardiovasculares "Dr. Horacio E. Cingolani", Facultad de Ciencias Médicas, Universidad Nacional de La Plata, CONICET, La Plata, Argentina.
| |
Collapse
|
2
|
Extracellular vesicles DJ-1 derived from hypoxia-conditioned hMSCs alleviate cardiac hypertrophy by suppressing mitochondria dysfunction and preventing ATRAP degradation. Pharmacol Res 2023; 187:106607. [PMID: 36509316 DOI: 10.1016/j.phrs.2022.106607] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND As a pathological myocardial remodeling process in a variety of cardiovascular diseases, cardiac hypertrophy still has no effective treatment. Human mesenchymal stem cells (hMSCs) derived extracellular vesicles (EVs) has been recognized as a promising treatment strategy for cardiac disease. METHODS In this study, the inhibitory effects on cardiac hypertrophy are compared between normoxia-conditioned hMSC-derived EVs (Nor-EVs) and hypoxia-conditioned hMSC-derived EVs (Hypo-EVs) in neonatal rat cardiomyocytes (NRCMs) after angiotensin II (Ang II) stimulation and in a mouse model of transverse aortic constriction (TAC). RESULTS We demonstrate that Hypo-EVs exert an increased inhibitory effect on cardiac hypertrophy compared with Nor-EVs. Parkinson disease protein 7 (PARK7/DJ-1) is identify as a differential protein between Nor-EVs and Hypo-EVs by quantitative proteomics analysis. Results show that DJ-1, which is rich in Hypo-EVs, alleviates mitochondrial dysfunction and excessive mitochondrial reactive oxygen species (mtROS) production as an antioxidant. Mechanistic studies demonstrate for the first time that DJ-1 may suppress cardiac hypertrophy by inhibiting the activity of proteasome subunit beta type 10 (PSMB10) through a direct physical interaction. This interaction can inhibit angiotensin II type 1 receptor (AT1R)-mediated signaling pathways resulting in cardiac hypertrophy through alleviating ubiquitination degradation of AT1R-associated protein (ATRAP). CONCLUSIONS When taken together, our study suggests that Hypo-EVs have significant potential as a novel therapeutic agent for the treatment of cardiac hypertrophy.
Collapse
|
3
|
NMR-Based Metabolomic Analysis of Cardiac Tissues Clarifies Molecular Mechanisms of CVB3-Induced Viral Myocarditis and Dilated Cardiomyopathy. Molecules 2022; 27:molecules27186115. [PMID: 36144851 PMCID: PMC9500976 DOI: 10.3390/molecules27186115] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/10/2022] [Accepted: 09/15/2022] [Indexed: 11/17/2022] Open
Abstract
Viral myocarditis (VMC), which is defined as inflammation of the myocardium with consequent myocardial injury, may develop chronic disease eventually leading to dilated cardiomyopathy (DCM). Molecular mechanisms underlying the progression from acute VMC (aVMC), to chronic VMC (cVMC) and finally to DCM, are still unclear. Here, we established mouse models of VMC and DCM with Coxsackievirus B3 infection and conducted NMR-based metabolomic analysis of aqueous metabolites extracted from cardiac tissues of three histologically classified groups including aVMC, cVMC and DCM. We showed that these three pathological groups were metabolically distinct from their normal counterparts and identified three impaired metabolic pathways shared by these pathological groups relative to normal controls, including nicotinate and nicotinamide metabolism; alanine, aspartate and glutamate metabolism; and D-glutamine and D-glutamate metabolism. We also identified two extra impaired metabolic pathways in the aVMC group, including glycine, serine and threonine metabolism; and taurine and hypotaurine metabolism Furthermore, we identified potential cardiac biomarkers for metabolically distinguishing these three pathological stages from normal controls. Our results indicate that the metabolomic analysis of cardiac tissues can provide valuable insights into the molecular mechanisms underlying the progression from acute VMC to DCM.
Collapse
|
4
|
Zhang F, Yan Y, Peng W, Wang L, Wang T, Xie Z, Luo H, Zhang J, Dong W. PARK7 promotes repair in early steroid-induced osteonecrosis of the femoral head by enhancing resistance to stress-induced apoptosis in bone marrow mesenchymal stem cells via regulation of the Nrf2 signaling pathway. Cell Death Dis 2021; 12:940. [PMID: 34645791 PMCID: PMC8514492 DOI: 10.1038/s41419-021-04226-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 09/13/2021] [Accepted: 09/24/2021] [Indexed: 12/13/2022]
Abstract
Novel therapies for the treatment of early steroid-induced osteonecrosis of the femoral head (SONFH) are urgently needed in orthopedics. Transplantation of bone marrow mesenchymal stem cells (BMSCs) provides new strategies for treating this condition at the early stage. However, stress-induced apoptosis of BMSCs transplanted into the femoral head necrotic area limits the efficacy of BMSC transplantation. Inhibiting BMSC apoptosis is key to improving the efficacy of this procedure. In our previous studies, we confirmed that Parkinson disease protein 7 (PARK7) is active in antioxidant defense and can clear reactive oxygen species (ROS), protect the mitochondria, and impart resistance to stress-induced apoptosis in BMSCs. In this study, we investigated the mechanism driving this PARK7-mediated resistance to apoptosis in BMSCs. Our results indicate that PARK7 promoted the disintegration of nuclear factor (erythroid-derived 2)-like 2 (Nrf2)/Kelch-like echinacoside-associated protein 1 (Keap1) complex. The free Nrf2 then entered the nucleus and activated the genetic expression of manganese superoxide dismutase (MnSOD), catalase (CAT), glutathione peroxidase (GPx), and other antioxidant enzymes that clear excessive ROS, thereby protecting BMSCs from stress-induced apoptosis. To further explore whether PARK7-mediated resistance to stress-induced apoptosis could improve the efficacy of BMSC transplantation in early-stage SONFH, we transplanted BMSCs-overexpressing PARK7 into rats with early-stage SONFH. We then evaluated the survival of transplanted BMSCs and bone regeneration in the femoral head necrotic area of these rats. The results indicated that PARK7 promoted the survival of BMSCs in the osteonecrotic area and improved the transplantation efficacy of BMSCs on early-stage SONFH. This study provides new ideas and methods for resisting the stress-induced apoptosis of BMSCs and improving the transplantation effect of BMSCs on early-stage SONFH.
Collapse
Affiliation(s)
- Fei Zhang
- Department of Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Yanglin Yan
- Department of Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Wuxun Peng
- Department of Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China.
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China.
| | - Lei Wang
- Department of Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Tao Wang
- Department of Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Zhihong Xie
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Hong Luo
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Jian Zhang
- Department of Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Wentao Dong
- Department of Orthopedics, The Affliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| |
Collapse
|
5
|
Müller J, Bertsch T, Volke J, Schmid A, Klingbeil R, Metodiev Y, Karaca B, Kim SH, Lindner S, Schupp T, Kittel M, Poschet G, Akin I, Behnes M. Narrative review of metabolomics in cardiovascular disease. J Thorac Dis 2021; 13:2532-2550. [PMID: 34012599 PMCID: PMC8107570 DOI: 10.21037/jtd-21-22] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiovascular diseases are accompanied by disorders in the cardiac metabolism. Furthermore, comorbidities often associated with cardiovascular disease can alter systemic and myocardial metabolism contributing to worsening of cardiac performance and health status. Biomarkers such as natriuretic peptides or troponins already support diagnosis, prognosis and treatment of patients with cardiovascular diseases and are represented in international guidelines. However, as cardiovascular diseases affect various pathophysiological pathways, a single biomarker approach cannot be regarded as ideal to reveal optimal clinical application. Emerging metabolomics technology allows the measurement of hundreds of metabolites in biological fluids or biopsies and thus to characterize each patient by its own metabolic fingerprint, improving our understanding of complex diseases, significantly altering the management of cardiovascular diseases and possibly personalizing medicine. This review outlines current knowledge, perspectives as well as limitations of metabolomics for diagnosis, prognosis and treatment of cardiovascular diseases such as heart failure, atherosclerosis, ischemic and non-ischemic cardiomyopathy. Furthermore, an ongoing research project tackling current inconsistencies as well as clinical applications of metabolomics will be discussed. Taken together, the application of metabolomics will enable us to gain more insights into pathophysiological interactions of metabolites and disease states as well as improving therapies of patients with cardiovascular diseases in the future.
Collapse
Affiliation(s)
- Julian Müller
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Thomas Bertsch
- Institute of Clinical Chemistry, Laboratory Medicine and Transfusion Medicine, Nuremburg General Hospital, Paracelsus Medical University, Nuremberg, Germany
| | - Justus Volke
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Alexander Schmid
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Rebecca Klingbeil
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Yulian Metodiev
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Bican Karaca
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Seung-Hyun Kim
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Simon Lindner
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Tobias Schupp
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Maximilian Kittel
- Institute for Clinical Chemistry, Faculty of Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Gernot Poschet
- Centre for Organismal Studies (COS), University of Heidelberg, Heidelberg, Germany
| | - Ibrahim Akin
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael Behnes
- First Department of Medicine, Faculty of Medicine Mannheim, University of Heidelberg, Mannheim, Germany
| |
Collapse
|
6
|
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
|
7
|
Zhang F, Peng W, Zhang J, Wang L, Dong W, Zheng Y, Wang Z, Xie Z, Wang T, Wang C, Yan Y. PARK7 enhances antioxidative-stress processes of BMSCs via the ERK1/2 pathway. J Cell Biochem 2020; 122:222-234. [PMID: 32918333 PMCID: PMC7820948 DOI: 10.1002/jcb.29845] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 08/15/2020] [Accepted: 08/31/2020] [Indexed: 12/15/2022]
Abstract
Oxidative stresss in the microenvironment surrounding lesions induces apoptosis of transplanted bone‐marrow‐derived mesenchymal stem cells (BMSCs). Hence, there is an urgent need for improving antioxidative‐stress processes of transplanted BMSCs to further promote their survival. The present study reports the role and mechanism of Parkinson's disease protein 7 (PARK7) in enhancing antioxidative activity in BMSCs. We used a PARK7 lentivirus to transfect BMSCs to up‐ or downregulate PARK7, and then used H2O2 to simulate oxidative stress in BMSCs in vitro. Overexpression of PARK7 effectively reduced reactive oxygen species and malondialdehyde, protected mitochondrial membrane potential, and resisted oxidative‐stress‐induced apoptosis of BMSCs, but the expression of PARK7 was downregulated, these results were reversed. At the same time, we also found that overexpression of PARK7 increased extracellular‐regulated protein kinase 1/2 (ERK1/2) phosphorylation and nuclear translocation, as well as upregulated Elk1 phosphorylation and superoxide dismutase (SOD) expression. In contrast, when U0126 was used to block the ERK1/2 pathway, ERK1/2 and Elk1 phosphorylation levels were downregulated, ERK1/2 nuclear translocation and SOD content were significantly reduced, and PARK7‐overexperssion‐induced antioxidative activity was completely blocked. Collectively, our results suggest that PARK7 overexpression increased antioxidative‐stress processes and survival of BMSCs subjected to H2O2 via activating the ERK1/2 signaling pathway. Our findings may guide the development of a PARK7‐specific strategy for improving the transplantation efficacy of BMSCs.
Collapse
Affiliation(s)
- Fei Zhang
- Department of Traumatologic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Wuxun Peng
- Department of Traumatologic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Jian Zhang
- Department of Traumatologic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Lei Wang
- Department of Statistics, Guizhou Maternal and Child Health Hospital, Guiyang, Guizhou, China
| | - Wentao Dong
- Department of Traumatologic Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, China.,School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yinggang Zheng
- Department of Orthopedics, The Affiliated Wudang Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhenwen Wang
- Department of Orthopedics, The Affiliated Cancer Hospital of Guizhou Medical University, Guiyang, Guizhou, China
| | - Zhihong Xie
- School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Tao Wang
- School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Chuan Wang
- School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| | - Yanglin Yan
- School of clinical medicine, Guizhou Medical University, Guiyang, Guizhou, China
| |
Collapse
|
8
|
Zhang J, Xu M, Zhou W, Li D, Zhang H, Chen Y, Ning L, Zhang Y, Li S, Yu M, Chen Y, Zeng H, Cen L, Zhou T, Zhou X, Lu C, Yu C, Li Y, Sun J, Kong X, Shen Z. Deficiency in the anti-apoptotic protein DJ-1 promotes intestinal epithelial cell apoptosis and aggravates inflammatory bowel disease via p53. J Biol Chem 2020; 295:4237-4251. [PMID: 32075910 PMCID: PMC7105307 DOI: 10.1074/jbc.ra119.010143] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 02/11/2020] [Indexed: 12/31/2022] Open
Abstract
Parkinson disease autosomal recessive, early onset 7 (PARK7 or DJ-1) is involved in multiple physiological processes and exerts anti-apoptotic effects on multiple cell types. Increased intestinal epithelial cell (IEC) apoptosis and excessive activation of the p53 signaling pathway is a hallmark of inflammatory bowel disease (IBD), which includes ulcerative colitis (UC) and Crohn's disease (CD). However, whether DJ-1 plays a role in colitis is unclear. To determine whether DJ-1 deficiency is involved in the p53 activation that results in IEC apoptosis in colitis, here we performed immunostaining, real-time PCR, and immunoblotting analyses to assess DJ-1 expression in human UC and CD samples. In the inflamed intestines of individuals with IBD, DJ-1 expression was decreased and negatively correlated with p53 expression. DJ-1 deficiency significantly aggravated colitis, evidenced by increased intestinal inflammation and exacerbated IEC apoptosis. Moreover, DJ-1 directly interacted with p53, and reduced DJ-1 levels increased p53 levels both in vivo and in vitro and were associated with decreased p53 degradation via the lysosomal pathway. We also induced experimental colitis with dextran sulfate sodium in mice and found that compared with DJ-1-/- mice, DJ-1-/-p53-/- mice have reduced apoptosis and inflammation and increased epithelial barrier integrity. Furthermore, pharmacological inhibition of p53 relieved inflammation in the DJ-1-/- mice. In conclusion, reduced DJ-1 expression promotes inflammation and IEC apoptosis via p53 in colitis, suggesting that the modulation of DJ-1 expression may be a potential therapeutic strategy for managing colitis.
Collapse
Affiliation(s)
- Jie Zhang
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Min Xu
- Department of Liver Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Weihua Zhou
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Dejian Li
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Hong Zhang
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yi Chen
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Longgui Ning
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yuwei Zhang
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Sha Li
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Mengli Yu
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Yishu Chen
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Hang Zeng
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Li Cen
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Tianyu Zhou
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Xinxin Zhou
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Chao Lu
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Chaohui Yu
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China
| | - Youming Li
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| | - Jing Sun
- Department of Gastroenterology, Rui Jin Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200031, China.
| | - Xiaoni Kong
- Institute of Clinical Immunology, Department of Liver Diseases, ShuGuang Hospital Affiliated to Shanghai University of Chinese Traditional Medicine, Shanghai, China.
| | - Zhe Shen
- Department of Gastroenterology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, China.
| |
Collapse
|
9
|
Ko YU, Kim SJ, Lee J, Song MY, Park KS, Park JB, Cho HS, Oh YJ. Protein kinase A-induced phosphorylation at the Thr154 affects stability of DJ-1. Parkinsonism Relat Disord 2019; 66:143-150. [DOI: 10.1016/j.parkreldis.2019.07.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 06/28/2019] [Accepted: 07/22/2019] [Indexed: 10/26/2022]
|
10
|
Zhang CJ, Huang Y, Lu JD, Lin J, Ge ZR, Huang H. Retracted: Upregulated microRNA-132 rescues cardiac fibrosis and restores cardiocyte proliferation in dilated cardiomyopathy through the phosphatase and tensin homolog-mediated PI3K/Akt signal transduction pathway. J Cell Biochem 2019; 120:1232-1244. [PMID: 30216493 DOI: 10.1002/jcb.27081] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/26/2018] [Indexed: 02/02/2023]
Abstract
Cardiac fibrosis is known to be present in dilated cardiomyopathy (DCM) and it predicts the occurrence of sudden death and congestive heart failure. The aim of our study is to investigate the expression of microRNA-132 (miR-132) and its effect on cardiocyte proliferation, apoptosis, and cardiac fibrosis by binding to phosphatase and tensin homolog (PTEN) through the phosphateidylinositol 3-kinase (PI3K)/protein kinase (Akt) signal transduction pathway in DCM rats. DCM rat models induced by doxorubicin were established and confirmed by an ultrasonic cardiogram. Epithelial cells were treated with inhibitors, activators, and small interfering RNAs to identify the mechanisms by which miR-132 controls cardiocyte activity and cardiac fibrosis. Angiotensin II (Ang II) and aldosterone (ALD) expressions were detected by an enzyme-linked immunosorbent assay. The relationship between PTEN and miR-132 was verified by a dual-luciferase reporter assay. Cell proliferation and apoptosis were tested by the MTT assay and flow cytometry. PTEN was determined to be the target gene of miR-132. Rat models of DCM exhibited a lower level of miR-132, PI3K, Akt, B-cell lymphoma 2, collagen I, and collagen III, but a higher level of PTEN, Bcl-2-associated X protein, and proliferating cell nuclear antigen as well as inflammatory response (Ang II and ALD), accompanied by declined cardiocyte proliferation and elevated apoptosis and cardiac fibrosis. Upregulated miR-132 or silenced PTEN activated the PI3K/Akt pathway, thus facilitating cardiocyte proliferation and repressing cardiocyte apoptosis and cardiac fibrosis, as well as inflammatory responses. Downregulated miR-132 reversed this tendency. These findings indicate that miR-132 activates the PI3K/Akt pathway by inhibiting PTEN expression, thus facilitating cardiocyte proliferation and inhibiting apoptosis and cardiac fibrosis in DCM rats.
Collapse
Affiliation(s)
- Chen-Jun Zhang
- Department of Cardiology, Gongli Hospital, Shanghai, China
| | - Yu Huang
- Department of Cardiology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Ji-De Lu
- Department of Cardiology, Gongli Hospital, Shanghai, China
| | - Jie Lin
- Department of Cardiology, Gongli Hospital, Shanghai, China
| | - Zhi-Ru Ge
- Department of Cardiology, Gongli Hospital, Shanghai, China
| | - Hui Huang
- Department of Cardiology, Gongli Hospital, Shanghai, China
| |
Collapse
|
11
|
Jiang F, Zhang DL, Jia M, Hao WH, Li YJ. Mangiferin inhibits high-fat diet induced vascular injury via regulation of PTEN/AKT/eNOS pathway. J Pharmacol Sci 2018; 137:265-273. [DOI: 10.1016/j.jphs.2018.07.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 05/22/2018] [Accepted: 06/27/2018] [Indexed: 02/09/2023] Open
|
12
|
Kim HS, Nam ST, Mun SH, Lee SK, Kim HW, Park YH, Kim B, Won KJ, Kim HR, Park YM, Kim HS, Beaven MA, Kim YM, Choi WS. DJ-1 controls bone homeostasis through the regulation of osteoclast differentiation. Nat Commun 2017; 8:1519. [PMID: 29142196 PMCID: PMC5688089 DOI: 10.1038/s41467-017-01527-y] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 09/25/2017] [Indexed: 11/09/2022] Open
Abstract
Receptor activator of NF-kB ligand (RANKL) generates intracellular reactive oxygen species (ROS), which increase RANKL-mediated signaling in osteoclast (OC) precursor bone marrow macrophages (BMMs). Here we show that a ROS scavenging protein DJ-1 negatively regulates RANKL-driven OC differentiation, also called osteoclastogenesis. DJ-1 ablation in mice leads to a decreased bone volume and an increase in OC numbers. In vitro, the activation of RANK-dependent signals is enhanced in DJ-1-deficient BMMs as compared to wild-type BMMs. DJ-1 suppresses the activation of both RANK-TRAF6 and RANK-FcRγ/Syk signaling pathways because of activation of Src homology region 2 domain-containing phosphatase-1, which is inhibited by ROS. Ablation of DJ-1 in mouse models of arthritis and RANKL-induced bone disease leads to an increase in the number of OCs, and exacerbation of bone damage. Overall, our results suggest that DJ-1 plays a role in bone homeostasis in normal physiology and in bone-associated pathology by negatively regulating osteoclastogenesis.
Collapse
Affiliation(s)
- Hyuk Soon Kim
- Department of Immunology and Physiology, School of Medicine, Konkuk University, Chungju, 380-701, Republic of Korea
| | - Seung Taek Nam
- Department of Immunology and Physiology, School of Medicine, Konkuk University, Chungju, 380-701, Republic of Korea
| | - Se Hwan Mun
- Department of Immunology and Physiology, School of Medicine, Konkuk University, Chungju, 380-701, Republic of Korea
- Department of Medicine, University of Connecticut Health Center, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Sun-Kyeong Lee
- Department of Medicine, University of Connecticut Health Center, 263 Farmington Ave, Farmington, CT, 06030, USA
| | - Hyun Woo Kim
- Department of Immunology and Physiology, School of Medicine, Konkuk University, Chungju, 380-701, Republic of Korea
| | - Young Hwan Park
- Department of Immunology and Physiology, School of Medicine, Konkuk University, Chungju, 380-701, Republic of Korea
| | - Bokyung Kim
- Department of Immunology and Physiology, School of Medicine, Konkuk University, Chungju, 380-701, Republic of Korea
| | - Kyung-Jong Won
- Department of Immunology and Physiology, School of Medicine, Konkuk University, Chungju, 380-701, Republic of Korea
| | - Hae-Rim Kim
- Department of Rheumatology, School of Medicine, Konkuk University, Chungju, 380-701, Republic of Korea
| | - Yeong-Min Park
- Department of Immunology and Physiology, School of Medicine, Konkuk University, Chungju, 380-701, Republic of Korea
| | - Hyung Sik Kim
- Department of Toxicology, School of Pharmacy, Sungkyunkwan University, Suwon, 440-746, Republic of Korea
| | - Michael A Beaven
- Laboratory of Molecular Immunology, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Young Mi Kim
- Department of Preventive Pharmacy, College of Pharmacy, Duksung Women's University, Seoul, 132-714, Republic of Korea
| | - Wahn Soo Choi
- Department of Immunology and Physiology, School of Medicine, Konkuk University, Chungju, 380-701, Republic of Korea.
| |
Collapse
|
13
|
Piqueras-Flores J, López-García A, Moreno-Reig Á, González-Martínez A, Hernández-González A, Vaamonde-Gamo J, Jurado-Román A. Structural and functional alterations of the heart in Parkinson’s disease. Neurol Res 2017; 40:53-61. [DOI: 10.1080/01616412.2017.1390933] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jesús Piqueras-Flores
- Cardiology Department, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Ana López-García
- Neurology Department. Complejo Hospitalario Universitario de Albacete, Albacete, Spain
| | - Álvaro Moreno-Reig
- Cardiology Department, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | | | | | - Julia Vaamonde-Gamo
- Neurology Department. Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Alfonso Jurado-Román
- Cardiology Department, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| |
Collapse
|
14
|
Biosa A, Sandrelli F, Beltramini M, Greggio E, Bubacco L, Bisaglia M. Recent findings on the physiological function of DJ-1: Beyond Parkinson's disease. Neurobiol Dis 2017; 108:65-72. [PMID: 28823929 DOI: 10.1016/j.nbd.2017.08.005] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Revised: 07/26/2017] [Accepted: 08/16/2017] [Indexed: 01/16/2023] Open
Abstract
Several mutations in the gene coding for DJ-1 have been associated with early onset forms of parkinsonism. In spite of the massive effort spent by the scientific community in understanding the physiological role of DJ-1, a consensus on what DJ-1 actually does within the cells has not been reached, with several diverse functions proposed. At present, the most accepted function for DJ-1 is a neuronal protective role against oxidative stress. However, how exactly this function is exerted by DJ-1 is not clear. In recent years, novel molecular mechanisms have been suggested that may account for the antioxidant properties of DJ-1. In this review, we critically analyse the experimental evidence, including some very recent findings, supporting the purported neuroprotective role of DJ-1 through different mechanisms linked to oxidative stress handling, as well as the relevance of these processes in the context of Parkinson's disease.
Collapse
Affiliation(s)
- Alice Biosa
- Molecular Physiology and Biophysics Unit, Department of Biology, University of Padova, 35131 Padova, Italy
| | - Federica Sandrelli
- Neurogenetics and Chronobiology Unit, Department of Biology, University of Padova, 35131 Padova, Italy
| | - Mariano Beltramini
- Molecular Physiology and Biophysics Unit, Department of Biology, University of Padova, 35131 Padova, Italy
| | - Elisa Greggio
- Molecular Physiology and Biophysics Unit, Department of Biology, University of Padova, 35131 Padova, Italy
| | - Luigi Bubacco
- Molecular Physiology and Biophysics Unit, Department of Biology, University of Padova, 35131 Padova, Italy
| | - Marco Bisaglia
- Molecular Physiology and Biophysics Unit, Department of Biology, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
15
|
Zhou Y, Shi X, Chen H, Zhang S, Salker MS, Mack AF, Föller M, Mak TW, Singh Y, Lang F. DJ-1/Park7 Sensitive Na + /H + Exchanger 1 (NHE1) in CD4 + T Cells. J Cell Physiol 2017; 232:3050-3059. [PMID: 27509531 DOI: 10.1002/jcp.25516] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/09/2016] [Indexed: 01/03/2023]
Abstract
DJ-1/Park7 is a redox-sensitive chaperone protein counteracting oxidation and presumably contributing to the control of oxidative stress responses and thus inflammation. DJ-1 gene deletion exacerbates the progression of Parkinson's disease presumably by augmenting oxidative stress. Formation of reactive oxygen species (ROS) is paralleled by activation of the Na+ /H+ exchanger 1 (NHE1). ROS formation in CD4+ T cells plays a decisive role in regulating inflammatory responses. In the present study, we explored whether DJ-1 is expressed in CD4+ T cells, and affects ROS production as well as NHE1 in those cells. To this end, DJ-1 and NHE1 transcript, and protein levels were quantified by qRT-PCR and Western blotting, respectively, intracellular pH (pHi ) utilizing bis-(2-carboxyethyl)-5-(and-6)-carboxyfluorescein (BCECF) fluorescence, NHE activity from realkalinization after an ammonium pulse, and ROS production utilizing 2',7' -dichlorofluorescin diacetate (DCFDA) fluorescence. As a result DJ-1 was expressed in CD4+ T cells. ROS formation, NHE1 transcript levels, NHE1 protein, and NHE activity were higher in CD4+ T cells from DJ-1 deficient mice than in CD4+ T cells from wild type mice. Antioxidant N-acetyl-cysteine (NAC) and protein tyrosine kinase (PTK) inhibitor staurosporine decreased the NHE activity in DJ-1 deficient CD4+ T cells, and blunted the difference between DJ-1-/- and DJ-1+/+ CD4+ T cells, an observation pointing to a role of ROS in the up-regulation of NHE1 in DJ-1-/- CD4+ T cells. In conclusion, DJ-1 is a powerful regulator of ROS production as well as NHE1 expression and activity in CD4+ T cells. J. Cell. Physiol. 232: 3050-3059, 2017. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Yuetao Zhou
- Department of Cardiology, Vascular Medicine and Physiology, Eberhard-Karls-University, Tübingen, Germany
| | - Xiaolong Shi
- Department of Cardiology, Vascular Medicine and Physiology, Eberhard-Karls-University, Tübingen, Germany
| | - Hong Chen
- Department of Cardiology, Vascular Medicine and Physiology, Eberhard-Karls-University, Tübingen, Germany
| | - Shaqiu Zhang
- Department of Cardiology, Vascular Medicine and Physiology, Eberhard-Karls-University, Tübingen, Germany.,Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City, Sichuan, China
| | - Madhuri S Salker
- Department of Cardiology, Vascular Medicine and Physiology, Eberhard-Karls-University, Tübingen, Germany
| | - Andreas F Mack
- Institute of Clinical Anatomy and Cell Analysis, Eberhard-Karls-University, Tübingen, Germany
| | - Michael Föller
- Department of Cardiology, Vascular Medicine and Physiology, Eberhard-Karls-University, Tübingen, Germany.,Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, UHN, Toronto, Canada.,Institute of Agricultural and Nutritional Sciences, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| | - Tak W Mak
- Campbell Family Institute for Breast Cancer Research, Ontario Cancer Institute, UHN, Toronto, Canada
| | - Yogesh Singh
- Department of Cardiology, Vascular Medicine and Physiology, Eberhard-Karls-University, Tübingen, Germany
| | - Florian Lang
- Department of Cardiology, Vascular Medicine and Physiology, Eberhard-Karls-University, Tübingen, Germany
| |
Collapse
|
16
|
Amatullah H, Shan Y, Beauchamp BL, Gali PL, Gupta S, Maron-Gutierrez T, Speck ER, Fox-Robichaud AE, Tsang JLY, Mei SHJ, Mak TW, Rocco PRM, Semple JW, Zhang H, Hu P, Marshall JC, Stewart DJ, Harper ME, Liaw PC, Liles WC, dos Santos CC. DJ-1/PARK7 Impairs Bacterial Clearance in Sepsis. Am J Respir Crit Care Med 2017; 195:889-905. [DOI: 10.1164/rccm.201604-0730oc] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Hajera Amatullah
- The Keenan Research Centre for Biomedical Science of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Physiology, Faculty of Medicine, and
| | - Yuexin Shan
- The Keenan Research Centre for Biomedical Science of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | | | - Patricia L. Gali
- The Keenan Research Centre for Biomedical Science of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Sahil Gupta
- The Keenan Research Centre for Biomedical Science of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, and
| | - Tatiana Maron-Gutierrez
- Ottawa Hospital Research Institute, University of Ottawa, Ottawa, Ontario, Canada
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - Edwin R. Speck
- The Keenan Research Centre for Biomedical Science of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Alison E. Fox-Robichaud
- Laboratory of Pulmonary Investigation, Carlos Chagas Filho Institute of Biophysics, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Jennifer L. Y. Tsang
- The Keenan Research Centre for Biomedical Science of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
- Thrombosis and Atherosclerosis Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Shirley H. J. Mei
- Department of Medicine, McMaster University, Hamilton (Niagara Campus), Ontario, Canada
| | - Tak W. Mak
- Department of Medical Biophysics and Immunology, The Campbell Family Institute for Breast Cancer Research at Princess Margaret Hospital, Ontario Cancer Institute, University Health Network, Toronto, Ontario, Canada
| | - Patricia R. M. Rocco
- Laboratory of Immunopharmacology, Oswaldo Cruz Institute, Fiocruz, Rio de Janeiro, Brazil
| | - John W. Semple
- The Keenan Research Centre for Biomedical Science of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Haibo Zhang
- The Keenan Research Centre for Biomedical Science of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba, Canada; and
| | - John C. Marshall
- The Keenan Research Centre for Biomedical Science of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
| | - Duncan J. Stewart
- Department of Medicine, McMaster University, Hamilton (Niagara Campus), Ontario, Canada
| | - Mary-Ellen Harper
- Institute of Medical Sciences, University of Toronto, Toronto, Ontario, Canada
| | - Patricia C. Liaw
- Thrombosis and Atherosclerosis Research Institute, Department of Medicine, McMaster University, Hamilton, Ontario, Canada
| | - W. Conrad Liles
- Department of Medicine, University of Washington, Seattle, Washington
| | - Claudia C. dos Santos
- The Keenan Research Centre for Biomedical Science of the Li Ka Shing Knowledge Institute of St. Michael's Hospital, Toronto, Ontario, Canada
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, and
| |
Collapse
|
17
|
Mokou M, Lygirou V, Vlahou A, Mischak H. Proteomics in cardiovascular disease: recent progress and clinical implication and implementation. Expert Rev Proteomics 2017; 14:117-136. [DOI: 10.1080/14789450.2017.1274653] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Marika Mokou
- Biotechnology Division, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Vasiliki Lygirou
- Biotechnology Division, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Antonia Vlahou
- Biotechnology Division, Biomedical Research Foundation Academy of Athens, Athens, Greece
| | - Harald Mischak
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Mosaiques Diagnostics, Hannover, Germany
| |
Collapse
|
18
|
Oh SE, Mouradian MM. Regulation of Signal Transduction by DJ-1. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1037:97-131. [PMID: 29147906 DOI: 10.1007/978-981-10-6583-5_8] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The ability of DJ-1 to modulate signal transduction has significant effects on how the cell regulates normal processes such as growth, senescence, apoptosis, and autophagy to adapt to changing environmental stimuli and stresses. Perturbations of DJ-1 levels or function can disrupt the equilibrium of homeostatic signaling networks and set off cascades that play a role in the pathogenesis of conditions such as cancer and Parkinson's disease.DJ-1 plays a major role in various pathways. It mediates cell survival and proliferation by activating the extracellular signal-regulated kinase (ERK1/2) pathway and the phosphatidylinositol-3-kinase (PI3K)/Akt pathway. It attenuates cell death signaling by inhibiting apoptosis signal-regulating kinase 1 (ASK1) activation as well as by inhibiting mitogen-activated protein kinase kinase kinase 1 (MEKK1/MAP3K1) activation of downstream apoptotic cascades. It also modulates autophagy through the ERK, Akt, or the JNK/Beclin1 pathways. In addition, DJ-1 regulates the transcription of genes essential for male reproductive function, such as spermatogenesis, by relaying nuclear receptor androgen receptor (AR) signaling. In this chapter, we summarize the ways that DJ-1 regulates these pathways, focusing on how its role in signal transduction contributes to cellular homeostasis and the pathologic states that result from dysregulation.
Collapse
Affiliation(s)
- Stephanie E Oh
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA
| | - M Maral Mouradian
- Center for Neurodegenerative and Neuroimmunologic Diseases, Department of Neurology, Rutgers - Robert Wood Johnson Medical School, Piscataway, NJ, 08854, USA.
| |
Collapse
|
19
|
Gene expression profiling reveals genes and transcription factors associated with dilated and ischemic cardiomyopathies. Pathol Res Pract 2016; 213:548-557. [PMID: 28318762 DOI: 10.1016/j.prp.2016.12.017] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 10/09/2016] [Accepted: 12/22/2016] [Indexed: 12/18/2022]
Abstract
AIMS Dilated cardiomyopathy (DCM) and ischemic cardiomyopathy (ICM) can cause heart failure, and this study aims to identify genes and transcription factors (TFs) associated with DCM and ICM. METHODS Gene expression dataset GSE42955 was generated from GEO database, and it contained 12 DCM, 12 ICM, and 5 control samples. Differentially expressed genes (DEGs) were identified between DCM (or ICM) and controls. Gene functions were investigated, and their associations were analyzed using Enrichmentmap plugin in Cytoscape. Protein-protein interactions (PPIs) between DEGs were determined, and DEGs with high degree were defined as key DEGs. Potential TFs of key DEGs were predicted using iRegulon plugin. Common DEGs were found, and their functional interactions were investigated using GeneMANIA. RESULTS A total of 362 and 300 DEGs were respectively identified for DCM and ICM in comparison with controls, and these DEGs mainly participated in similar functions about extracellular region, membrane, immune process, and defense response. PPI networks were respectively constructed for DCM and ICM, and 26 key DEGs (e.g. CXCL10, IL6, TLR3, and VCAM1) were found, which might be targeted by 35 TFs (e.g. IRF1). Besides, 47 common up-regulated DEGs were found, which participated in 14 pathways like Apoptosis, Collagen formation, as well as 127 common down-regulated DEGs that involved in 20 pathways like Adaptive immune system, Interferon γ signaling (e.g. IRF1, VCAM1), and Toll-like receptor signaling pathway (e.g. CXCL10, IL6, TLR3). CONCLUSION DCM and ICM may share similar mechanism, and TFs (e.g. IRF1) play crucial roles in their development via regulating gene expression.
Collapse
|
20
|
Raghow R. An 'Omics' Perspective on Cardiomyopathies and Heart Failure. Trends Mol Med 2016; 22:813-827. [PMID: 27499035 DOI: 10.1016/j.molmed.2016.07.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 07/15/2016] [Accepted: 07/15/2016] [Indexed: 12/27/2022]
Abstract
Pathological enlargement of the heart, represented by hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM), occurs in response to many genetic and non-genetic factors. The clinical course of cardiac hypertrophy is remarkably variable, ranging from lifelong absence of symptoms to rapidly declining heart function and sudden cardiac death (SCD). Unbiased omics studies have begun to provide a glimpse into the molecular framework underpinning altered mechanotransduction, mitochondrial energetics, oxidative stress, and extracellular matrix in the heart undergoing physiological and pathological hypertrophy. Omics analyses indicate that post-transcriptional regulation of gene expression plays an overriding role in the normal and diseased heart. Studies to date highlight a need for more effective bioinformatics to better integrate patient omics data with their comprehensive clinical histories.
Collapse
Affiliation(s)
- Rajendra Raghow
- Department of Pharmacology, College of Medicine, The University of Tennessee Health Science Center and the VA Medical Center, Memphis, TN 38104, USA.
| |
Collapse
|
21
|
Saito Y, Akazawa-Ogawa Y, Matsumura A, Saigoh K, Itoh S, Sutou K, Kobayashi M, Mita Y, Shichiri M, Hisahara S, Hara Y, Fujimura H, Takamatsu H, Hagihara Y, Yoshida Y, Hamakubo T, Kusunoki S, Shimohama S, Noguchi N. Oxidation and interaction of DJ-1 with 20S proteasome in the erythrocytes of early stage Parkinson's disease patients. Sci Rep 2016; 6:30793. [PMID: 27470541 PMCID: PMC4965792 DOI: 10.1038/srep30793] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 07/11/2016] [Indexed: 02/05/2023] Open
Abstract
Parkinson's disease (PD) is a progressive, age-related, neurodegenerative disorder, and oxidative stress is an important mediator in its pathogenesis. DJ-1, the product of the causative gene of a familial form of PD, plays a significant role in anti-oxidative defence to protect cells from oxidative stress. DJ-1 undergoes preferential oxidation at the cysteine residue at position 106 (Cys-106) under oxidative stress. Here, using specific antibodies against Cys-106-oxidized DJ-1 (oxDJ-1), it was found that the levels of oxDJ-1 in the erythrocytes of unmedicated PD patients (n = 88) were higher than in those of medicated PD patients (n = 62) and healthy control subjects (n = 33). Elevated oxDJ-1 levels were also observed in a non-human primate PD model. Biochemical analysis of oxDJ-1 in erythrocyte lysates showed that oxDJ-1 formed dimer and polymer forms, and that the latter interacts with 20S proteasome. These results clearly indicate a biochemical alteration in the blood of PD patients, which could be utilized as an early diagnosis marker for PD.
Collapse
Affiliation(s)
- Yoshiro Saito
- Systems Life Sciences laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
- National Institute of Advanced Industrial Science and Technology (AIST), Ikeda, Osaka 563-8577, Japan
| | - Yoko Akazawa-Ogawa
- National Institute of Advanced Industrial Science and Technology (AIST), Ikeda, Osaka 563-8577, Japan
| | - Akihiro Matsumura
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Kazumasa Saigoh
- Department of Neurology, Kinki University Faculty of Medicine, Osaka 589-8511, Japan
| | - Sayoko Itoh
- Hamamatsu Pharma Research Inc., Hamamatsu 431-2103, Japan
| | - Kenta Sutou
- Systems Life Sciences laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Mayuka Kobayashi
- Systems Life Sciences laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Yuichiro Mita
- Systems Life Sciences laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Mototada Shichiri
- National Institute of Advanced Industrial Science and Technology (AIST), Ikeda, Osaka 563-8577, Japan
| | - Shin Hisahara
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Yasuo Hara
- Hara Clinic, Ikeda, Osaka 563-0025, Japan
| | - Harutoshi Fujimura
- Department of Neurology, National Hospital Organization Toneyama National Hospital, Toyonaka, Osaka 560-8552, Japan
| | | | - Yoshihisa Hagihara
- National Institute of Advanced Industrial Science and Technology (AIST), Ikeda, Osaka 563-8577, Japan
| | - Yasukazu Yoshida
- National Institute of Advanced Industrial Science and Technology (AIST), Ikeda, Osaka 563-8577, Japan
| | - Takao Hamakubo
- Laboratory of Systems Biology and Medicine, Research Center for Advanced Science and Technology, University of Tokyo, Tokyo 153-0041, Japan
| | - Susumu Kusunoki
- Department of Neurology, Kinki University Faculty of Medicine, Osaka 589-8511, Japan
| | - Shun Shimohama
- Department of Neurology, School of Medicine, Sapporo Medical University, Sapporo 060-8556, Japan
| | - Noriko Noguchi
- Systems Life Sciences laboratory, Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| |
Collapse
|
22
|
Fernandez-Caggiano M, Schröder E, Cho HJ, Burgoyne J, Barallobre-Barreiro J, Mayr M, Eaton P. Oxidant-induced Interprotein Disulfide Formation in Cardiac Protein DJ-1 Occurs via an Interaction with Peroxiredoxin 2. J Biol Chem 2016; 291:10399-410. [PMID: 26945066 DOI: 10.1074/jbc.m115.699850] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Indexed: 12/24/2022] Open
Abstract
The role and responses of the dimeric DJ-1 protein to cardiac oxidative stress is incompletely understood. H2O2 induces a 50-kDa DJ-1 interprotein homodimer disulfide, known to form between Cys-53 on each subunit. A trimeric 75-kDa DJ-1 complex that mass spectrometry shows contained 2-Cys peroxiredoxin also formed and precedes the appearance of the disulfide dimer. These observations may represent peroxiredoxin sensing and transducing the oxidant signal to DJ-1. The dimeric disulfide DJ-1 complex was stabilized by auranofin, suggesting that thioredoxin recycles it in cells. Higher concentrations of H2O2 concomitantly induce DJ-1 Cys-106 hyperoxidation (sulfination or sulfonation) in myocytes, perfused heart, or HEK cells. An oxidation-resistant C53A DJ-1 shows potentiated H2O2-induced Cys-106 hyperoxidation. DJ-1 also forms multiple disulfides with unknown target proteins during H2O2 treatment, the formation of which is also potentiated in cells expressing the C53A mutant. This suggests that the intersubunit disulfide induces a conformational change that limits Cys-106 forming heterodisulfide protein complexes or from hyperoxidizing. High concentrations of H2O2 also induce cell death, with DJ-1 Cys-106 sulfonation appearing causal in these events, as expressionof C53A DJ-1 enhanced both Cys-106 sulfonation and cell death. Nonetheless, expression of the DJ-1 C106A mutant, which fully prevents hyperoxidation, also showed exacerbated cell death responses to H2O2 A rational explanation for these findings is that DJ-1 Cys-106 forms disulfides with target proteins to limit oxidant-induced cell death. However, when Cys-106 is hyperoxidized, formation of these potentially protective heterodimeric disulfide complexes is limited, and so cell death is exacerbated.
Collapse
Affiliation(s)
- Mariana Fernandez-Caggiano
- From the King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom and
| | - Ewald Schröder
- From the King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom and
| | - Hyun-Ju Cho
- From the King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom and
| | - Joseph Burgoyne
- From the King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom and
| | - Javier Barallobre-Barreiro
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The James Black Centre, King's College Hospital, London SE5 9NU, United Kingdom
| | - Manuel Mayr
- King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The James Black Centre, King's College Hospital, London SE5 9NU, United Kingdom
| | - Philip Eaton
- From the King's College London, Cardiovascular Division, The British Heart Foundation Centre of Excellence, The Rayne Institute, St. Thomas' Hospital, London SE1 7EH, United Kingdom and
| |
Collapse
|
23
|
Abstract
Onset of cancer and neurodegenerative disease occurs by abnormal cell growth and neuronal cell death, respectively, and the number of patients with both diseases has been increasing in parallel with an increase in mean lifetime, especially in developed countries. Although both diseases are sporadic, about 10% of the diseases are genetically inherited, and analyses of such familial forms of gene products have contributed to an understanding of the molecular mechanisms underlying the onset and pathogenesis of these diseases. I have been working on c-myc, a protooncogene, for a long time and identified various c-Myc-binding proteins that play roles in c-Myc-derived tumorigenesis. Among these proteins, some proteins have been found to be also responsible for the onset of neurodegenerative diseases, including Parkinson's disease, retinitis pigmentosa and cerebellar atrophy. In this review, I summarize our findings indicating the common mechanisms of onset between cancer and neurodegenerative diseases, with a focus on genes such as DJ-1 and Myc-Modulator 1 (MM-1) and signaling pathways that contribute to the onset and pathogenesis of cancer and neurodegenerative diseases.
Collapse
|
24
|
West JA, Beqqali A, Ament Z, Elliott P, Pinto YM, Arbustini E, Griffin JL. A targeted metabolomics assay for cardiac metabolism and demonstration using a mouse model of dilated cardiomyopathy. Metabolomics 2016; 12:59. [PMID: 27069442 PMCID: PMC4781888 DOI: 10.1007/s11306-016-0956-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2015] [Accepted: 11/14/2015] [Indexed: 12/12/2022]
Abstract
Metabolomics can be performed either as an 'open profiling' tool where the aim is to measure, usually in a semi-quantitative manner, as many metabolites as possible or perform 'closed' or 'targeted' analyses where instead a pre-defined set of metabolites are measured. Targeted methods can be designed to be more sensitive and quantitative and so are particularly appropriate to systems biology for quantitative models of systems or when metabolomics is performed in a hypothesis driven manner to test whether a particular pathway is perturbed. We describe a targeted metabolomics assay that quantifies a broad range of over 130 metabolites relevant to cardiac metabolism including the pathways of the citric acid cycle, fatty acid oxidation, glycolysis, the pentose phosphate pathway, amino acid metabolism, the urea cycle, nucleotides and reactive oxygen species using tandem mass spectrometry to produce quantitative, sensitive and robust data. This assay is illustrated by profiling cardiac metabolism in a lamin A/C (Lmna) mouse model of dilated cardiomyopathy (DCM). The model of DCM was characterised by increases in concentrations of proline and methyl-histidine suggestive of increased myofibrillar and collagen degradation, as well as decreases in a number of citric acid cycle intermediates and carnitine derivatives indicating reduced energy metabolism in the dilated heart. These assays could be used for any other cardiac or cardiovascular disease in that they cover central core metabolism and key pathways involved in cardiac metabolism, and may provide a general start for many mammalian systems.
Collapse
Affiliation(s)
- James A. West
- The Department of Biochemistry & The Cambridge Systems Biology Centre, University of Cambridge, Tennis Court Road, Cambridge, CB2 1GA UK
- The Elsie Widdowson Laboratory, Medical Research Council Human Nutrition Research, 120 Fulbourn Road, Cambridge, CB1 9NL UK
| | - Abdelaziz Beqqali
- Department of Experimental Cardiology, Academic Medical Centre, Amsterdam, The Netherlands
| | - Zsuzsanna Ament
- The Department of Biochemistry & The Cambridge Systems Biology Centre, University of Cambridge, Tennis Court Road, Cambridge, CB2 1GA UK
- The Elsie Widdowson Laboratory, Medical Research Council Human Nutrition Research, 120 Fulbourn Road, Cambridge, CB1 9NL UK
| | - Perry Elliott
- Heart Hospital, University College London, London, W1G 8PH UK
| | - Yigal M. Pinto
- Department of Experimental Cardiology, Academic Medical Centre, Amsterdam, The Netherlands
| | | | - Julian L. Griffin
- The Department of Biochemistry & The Cambridge Systems Biology Centre, University of Cambridge, Tennis Court Road, Cambridge, CB2 1GA UK
- The Elsie Widdowson Laboratory, Medical Research Council Human Nutrition Research, 120 Fulbourn Road, Cambridge, CB1 9NL UK
| |
Collapse
|
25
|
Direct Evidence that Myocardial Insulin Resistance following Myocardial Ischemia Contributes to Post-Ischemic Heart Failure. Sci Rep 2015; 5:17927. [PMID: 26659007 PMCID: PMC4677294 DOI: 10.1038/srep17927] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/23/2015] [Indexed: 12/31/2022] Open
Abstract
A close link between heart failure (HF) and systemic insulin resistance has been well documented, whereas myocardial insulin resistance and its association with HF are inadequately investigated. This study aims to determine the role of myocardial insulin resistance in ischemic HF and its underlying mechanisms. Male Sprague-Dawley rats subjected to myocardial infarction (MI) developed progressive left ventricular dilation with dysfunction and HF at 4 wk post-MI. Of note, myocardial insulin sensitivity was decreased as early as 1 wk after MI, which was accompanied by increased production of myocardial TNF-α. Overexpression of TNF-α in heart mimicked impaired insulin signaling and cardiac dysfunction leading to HF observed after MI. Treatment of rats with a specific TNF-α inhibitor improved myocardial insulin signaling post-MI. Insulin treatment given immediately following MI suppressed myocardial TNF-α production and improved cardiac insulin sensitivity and opposed cardiac dysfunction/remodeling. Moreover, tamoxifen-induced cardiomyocyte-specific insulin receptor knockout mice exhibited aggravated post-ischemic ventricular remodeling and dysfunction compared with controls. In conclusion, MI induces myocardial insulin resistance (without systemic insulin resistance) mediated partly by ischemia-induced myocardial TNF-α overproduction and promotes the development of HF. Our findings underscore the direct and essential role of myocardial insulin signaling in protection against post-ischemic HF.
Collapse
|
26
|
Differential effect of DJ-1/PARK7 on development of natural and induced regulatory T cells. Sci Rep 2015; 5:17723. [PMID: 26634899 PMCID: PMC4669505 DOI: 10.1038/srep17723] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 11/04/2015] [Indexed: 12/19/2022] Open
Abstract
Regulatory T cells (Tregs) are essential for maintaining an effective immune tolerance and a homeostatic balance of various other immune cells. To manipulate the immune response during infections and autoimmune disorders, it is essential to know which genes or key molecules are involved in the development of Tregs. Transcription factor Foxp3 is required for the development of Tregs and governs most of the suppressive functions of these cells. Inhibited PI3K/AKT/mTOR signalling is critical for Foxp3 stability. Previous studies have suggested that DJ-1 or PARK7 protein is a positive regulator of the PI3K/AKT/mTOR pathway by negatively regulating the activity of PTEN. Thus, we hypothesised that a lack of DJ-1 could promote the development of Tregs. As a result, loss of DJ-1 decreased the total CD4(+) T cell numbers but increased the fraction of thymic and peripheral nTregs. In contrast, Foxp3 generation was not augmented following differentiation of DJ-1-deficient naïve CD4(+) T cells. DJ-1-deficient-iTregs were imperfect in replication, proliferation and more prone to cell death. Furthermore, DJ-1 deficient iTregs were less sensitive to pSmad2 and pStat5 signalling but had activated AKT/mTOR signalling. These observations reveal an unexpected differential role of DJ-1 in the development of nTregs and iTregs.
Collapse
|
27
|
Abstract
Ischemic heart disease (IHD) is the leading cause of death and disability worldwide. Therefore, novel therapeutic targets for protecting the heart against acute ischemia/reperfusion injury (IRI) are required to attenuate cardiomyocyte death, preserve myocardial function, and prevent the onset of heart failure. In this regard, a specific group of mitochondrial proteins, which have been linked to familial forms of Parkinson's disease (PD), may provide novel therapeutic targets for cardioprotection. In dopaminergic neurons of the substantia nigra, these PD proteins, which include Parkin, PINK1, DJ-1, LRRK2, and α-synuclein, play essential roles in preventing cell death-through maintaining normal mitochondrial function, protecting against oxidative stress, mediating mitophagy, and preventing apoptosis. These rare familial forms of PD may therefore provide important insights into the pathophysiology underlying mitochondrial dysfunction and the development of PD. Interestingly, these PD proteins are also present in the heart, but their role in myocardial health and disease is not clear. In this article, we review the role of these PD proteins in the heart and explore their potential as novel mitochondrial targets for cardioprotection.
Collapse
Affiliation(s)
- Uma A Mukherjee
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore
| | - Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford University School of Medicine, CA, USA
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK; Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore, Singapore; National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore; The National Institute of Health Research University College London Hospitals Biomedical Research Centre, London, UK.
| |
Collapse
|
28
|
|
29
|
Zhang W, Xing B, Yang L, Shi J, Zhou X. Icaritin Attenuates Myocardial Ischemia and Reperfusion Injury Via Anti-Inflammatory and Anti-Oxidative Stress Effects in Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2015; 43:1083-97. [PMID: 26364662 DOI: 10.1142/s0192415x15500627] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Icaritin (ICT) is a traditional Chinese medicinal herb proved to be neuroprotective and exerts promoting effects on cardiac differentiation. However, its role in cardioprotection against myocardial ischemia/reperfusion (MI/R) injury remains largely unknown. This study aimed to investigate the effects of ICT treatment on MI/R injury and the underlying mechanisms. Rats were subjected to 30 min of myocardial ischemic insult followed by 3 h of reperfusion. ICT (3, 10, and 30 mg/kg) was administered intraperitoneally 10 min before reperfusion. ICT treatment at the dose of 10 and 30 mg/kg improved cardiac function and limited infarct size following MI/R. Meanwhile, ICT reduced plasma creatine kinase (CK), lactate dehydrogenase (LDH) activities and cardiomyocyte apoptosis in I/R heart tissue. Moreover, ICT treatment not only inhibited the pro-inflammatory cytokine TNF-α production and increased the anti-inflammatory cytokine IL-10 level in myocardium but also reduced the increase in the generation of superoxide content and malondialdehyde (MDA) formation and simultaneously increased the anti-oxidant capability in I/R hearts. Furthermore, ICT treatment increased Akt phosphorylation and inhibited PTEN expression in I/R hearts. PI3K inhibitor wortmannin inhibited ICT-enhanced Akt phosphorylation, and blunted ICT-mediated anti-oxidative and anti-inflammatory effects and cardioprotection. Our study indicated for the first time that ICT reduces inflammation and oxidative stress and protects against MI/R injury in rats, which might be via a PI3K–Akt-dependent mechanism.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
- Department of Cardiothoracic Surgery, Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Baichun Xing
- Department of Anesthesiology, Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Linlin Yang
- Department of Anesthesiology, Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Jialun Shi
- Department of Cardiothoracic Surgery, Peace Hospital of Changzhi Medical College, Changzhi, Shanxi 046000, P.R. China
| | - Xinmin Zhou
- Department of Cardiothoracic Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
30
|
Wen XR, Fu YY, Liu HZ, Wu J, Shao XP, Zhang XB, Tang M, Shi Y, Ma K, Zhang F, Wang YW, Tang H, Han D, Zhang P, Wang SL, Xu Z, Song YJ. Neuroprotection of Sevoflurane Against Ischemia/Reperfusion-Induced Brain Injury Through Inhibiting JNK3/Caspase-3 by Enhancing Akt Signaling Pathway. Mol Neurobiol 2015; 53:1661-1671. [PMID: 25687432 DOI: 10.1007/s12035-015-9111-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2014] [Accepted: 01/21/2015] [Indexed: 01/27/2023]
Abstract
In this study, we investigated the neuroprotective effect of sevoflurane against ischemic brain injury and its underlying molecular mechanisms. Transient global brain ischemia was induced by 4-vessel occlusion in adult male Sprague-Dawley rats. The rats were pretreated with sevoflurane alone or sevoflurane combined with LY294002/wortmannin (selective inhibitor of PI3K) before ischemia. Cresyl violet staining was used to examine the survival of hippocampal CA1 pyramidal neurons. Immunoblotting and immunoprecipitation were performed to measure the phosphorylation of Akt1, PRAS40, ASK1, and JNK3 and the expression of cleaved-caspase-3. The results demonstrated that a moderate dose of sevoflurane inhalation of 2% for 2 h had significant neuroprotective effects against ischemia/reperfusion induced hippocampal neuron death. Sevoflurane significantly increased Akt and PRAS40 phosphorylation and decreased the phosphorylation of ASK1 at 6 h after reperfusion and the phosphorylation of JNK3 at 3 days after reperfusion following 15 min of transient global brain ischemia. Conversely, LY294002 and wortmannin significantly inhibited the effects of sevoflurane. Taken together, the results suggest that sevoflurane could suppress ischemic brain injury by downregulating the activation of the ASK1/JNK3 cascade via increasing the phosphorylation of Akt1 during ischemia/reperfusion.
Collapse
Affiliation(s)
- Xiang-Ru Wen
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China.,School of Basic Education Sciences, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Yan-Yan Fu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China.,Department of Genetics, Research Center for Neurobiology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Hong-Zhi Liu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Jian Wu
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China.,Department of Genetics, Research Center for Neurobiology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Xiao-Ping Shao
- School of Public Health, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Xun-Bao Zhang
- School of Public Health, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Man Tang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Yue Shi
- School of Public Health, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Kai Ma
- Department of Medical Information, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Fang Zhang
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Yi-Wen Wang
- School of Public Health, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Hui Tang
- School of Public Health, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Dong Han
- Jiangsu Province Key Laboratory of Anesthesiology, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China
| | - Pu Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, 221002, People's Republic of China
| | - Shu-Ling Wang
- Department of Respiratory Medicine, The Affiliated Municipal Hospital of Xuzhou Medical College, Xuzhou, Jiangsu, 221002, People's Republic of China
| | - Zhou Xu
- School of Basic Education Sciences, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China.
| | - Yuan-Jian Song
- Jiangsu Key Laboratory of Brain Disease Bioinformation, Xuzhou Medical College, Xuzhou, Jiangsu, 221004, People's Republic of China. .,Department of Genetics, Research Center for Neurobiology, Xuzhou Medical College, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, People's Republic of China.
| |
Collapse
|
31
|
Next generation of cardiovascular studies: transcriptional responses of the human myocardium during cardiac surgery. Anesthesiology 2015; 122:486-8. [PMID: 25569809 DOI: 10.1097/aln.0000000000000581] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
DJ-1 interacts with RACK1 and protects neurons from oxidative-stress-induced apoptosis. Biochem J 2014; 462:489-97. [PMID: 24947010 DOI: 10.1042/bj20140235] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PD (Parkinson's disease) is a complex disorder that is associated with neuronal loss or dysfunction caused by genetic risks, environmental factors and advanced aging. It has been reported that DJ-1 mutations rendered neurons sensitive to oxidative damage, which led to the onset of familiar PD. However, the molecular mechanism is still unclear. In the present study we show that DJ-1 interacts with RACK1 (receptor of activated C kinase 1) and increases its dimerization and protein stability. The DJ-1 transgene protects cortical neurons from H2O2-induced apoptosis, and this protective effect is abrogated by knocking down RACK1. Similarly, deletion of DJ-1 in cortical neurons increases the sensitivity to H2O2, and the damage can be significantly rescued by DJ-1 or DJ-1/RACK1 co-transfection, but not by RACK1 alone. We observed further that the interaction of DJ-1 and RACK1 is disrupted by H2O2 or MPP+ (1-methyl-4-phenylpyridinium) treatment, and the protein levels of DJ-1 and RACK1 decreased in neurodegenerative disease models. Taken together, the DJ-1-RACK1 complex protects neurons from oxidative stress-induced apoptosis, with the implication that DJ-1 and RACK1 might be novel targets in the treatment of neurodegenerative diseases.
Collapse
|
33
|
Schechter MA, Hsieh MKH, Njoroge LW, Thompson JW, Soderblom EJ, Feger BJ, Troupes CD, Hershberger KA, Ilkayeva OR, Nagel WL, Landinez GP, Shah KM, Burns VA, Santacruz L, Hirschey MD, Foster MW, Milano CA, Moseley MA, Piacentino V, Bowles DE. Phosphoproteomic profiling of human myocardial tissues distinguishes ischemic from non-ischemic end stage heart failure. PLoS One 2014; 9:e104157. [PMID: 25117565 PMCID: PMC4130503 DOI: 10.1371/journal.pone.0104157] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2014] [Accepted: 07/06/2014] [Indexed: 12/31/2022] Open
Abstract
The molecular differences between ischemic (IF) and non-ischemic (NIF) heart failure are poorly defined. A better understanding of the molecular differences between these two heart failure etiologies may lead to the development of more effective heart failure therapeutics. In this study extensive proteomic and phosphoproteomic profiles of myocardial tissue from patients diagnosed with IF or NIF were assembled and compared. Proteins extracted from left ventricular sections were proteolyzed and phosphopeptides were enriched using titanium dioxide resin. Gel- and label-free nanoscale capillary liquid chromatography coupled to high resolution accuracy mass tandem mass spectrometry allowed for the quantification of 4,436 peptides (corresponding to 450 proteins) and 823 phosphopeptides (corresponding to 400 proteins) from the unenriched and phospho-enriched fractions, respectively. Protein abundance did not distinguish NIF from IF. In contrast, 37 peptides (corresponding to 26 proteins) exhibited a ≥ 2-fold alteration in phosphorylation state (p<0.05) when comparing IF and NIF. The degree of protein phosphorylation at these 37 sites was specifically dependent upon the heart failure etiology examined. Proteins exhibiting phosphorylation alterations were grouped into functional categories: transcriptional activation/RNA processing; cytoskeleton structure/function; molecular chaperones; cell adhesion/signaling; apoptosis; and energetic/metabolism. Phosphoproteomic analysis demonstrated profound post-translational differences in proteins that are involved in multiple cellular processes between different heart failure phenotypes. Understanding the roles these phosphorylation alterations play in the development of NIF and IF has the potential to generate etiology-specific heart failure therapeutics, which could be more effective than current therapeutics in addressing the growing concern of heart failure.
Collapse
Affiliation(s)
- Matthew A. Schechter
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Michael K. H. Hsieh
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Linda W. Njoroge
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - J. Will Thompson
- Duke Proteomics Core, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Erik J. Soderblom
- Duke Proteomics Core, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Bryan J. Feger
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Constantine D. Troupes
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Kathleen A. Hershberger
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Olga R. Ilkayeva
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Whitney L. Nagel
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Gina P. Landinez
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Kishan M. Shah
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Virginia A. Burns
- Duke Translational Research Institute, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Lucia Santacruz
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Matthew D. Hirschey
- Sarah W. Stedman Nutrition and Metabolism Center, Duke University Medical Center, Durham, North Carolina, United States of America
- Department of Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Matthew W. Foster
- Division of Pulmonary, Allergy and Critical Care, Medicine, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Carmelo A. Milano
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - M. Arthur Moseley
- Duke Proteomics Core, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Valentino Piacentino
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Dawn E. Bowles
- Department of Surgery, Duke University Medical Center, Durham, North Carolina, United States of America
| |
Collapse
|
34
|
Han Z, Chen F, Ge X, Tan J, Lei P, Zhang J. miR-21 alleviated apoptosis of cortical neurons through promoting PTEN-Akt signaling pathway in vitro after experimental traumatic brain injury. Brain Res 2014; 1582:12-20. [PMID: 25108037 DOI: 10.1016/j.brainres.2014.07.045] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Revised: 07/25/2014] [Accepted: 07/29/2014] [Indexed: 12/24/2022]
Abstract
Traumatic brain injury (TBI) is a major cause of chronic disability and death in young adults worldwide. Multiple cellular, molecular and biochemical changes impact the development and outcome of TBI. Neuronal cell apoptosis, which is an important pathological change in secondary brain damage, is crucial to determine the functional recovery after TBI. miR-21, a widely-reported oncogene, which can reduce cell apoptosis in cancer, has been confirmed to be a pronounced up-regulated miRNA after TBI in animal model. Our study is designed to investigate whether miR-21 has the function of antiapoptosis in experimental TBI model in vitro and to explore the possible regulatory mechanism of miR-21 on neuronal apoptosis. The scratch cell injury was performed to mimic TBI-induced apoptosis in neurons, and miR-21 agomir/antagomir was transfected to up-/down-regulate the miR-21 level. Our data suggests that miR-21 can reduce the number of TUNEL-positive neurons. Meanwhile, miR-21 decreased the expression level of PTEN, and increased the phosphorylation of Akt significantly. In neurons transfected with miR-21 agomir, the expression of Bcl-2 was promoted while the caspase-3, caspase-9 and Bax level were down-regulated, which are crucially the downstream apoptosis-related proteins of PTEN-Akt signaling pathway. In conclusion, miR-21 can exert the function of reducing neuronal apoptosis through activating the PTEN-Akt signaling pathway. Our research provides new insights into the molecular mechanisms of neuronal apoptosis following TBI, which reminds that miR-21may be a potential therapeutic target for TBI treatment.
Collapse
Affiliation(s)
- Zhaoli Han
- Department of Neurosurgery, Tianjin Neurological Institute General Hospital, Tianjin Medical University, Tianjin 300352, China
| | - Fanglian Chen
- Department of Neurosurgery, Tianjin Neurological Institute General Hospital, Tianjin Medical University, Tianjin 300352, China
| | - Xintong Ge
- Department of Neurosurgery, Tianjin Neurological Institute General Hospital, Tianjin Medical University, Tianjin 300352, China
| | - Jin Tan
- Tianjin Institute of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300352, China
| | - Ping Lei
- Department of Neurosurgery, Tianjin Neurological Institute General Hospital, Tianjin Medical University, Tianjin 300352, China; Tianjin Institute of Geriatrics, Tianjin Medical University General Hospital, Tianjin 300352, China.
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Neurological Institute General Hospital, Tianjin Medical University, Tianjin 300352, China
| |
Collapse
|
35
|
Stechschulte LA, Hinds TD, Ghanem SS, Shou W, Najjar SM, Sanchez ER. FKBP51 reciprocally regulates GRα and PPARγ activation via the Akt-p38 pathway. Mol Endocrinol 2014; 28:1254-64. [PMID: 24933248 DOI: 10.1210/me.2014-1023] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
FK506-binding protein 51 (FKBP51) is a negative regulator of glucocorticoid receptor-α (GRα), although the mechanism is unknown. We show here that FKBP51 is also a chaperone to peroxisome proliferator-activated receptor-γ (PPARγ), which is essential for activity, and uncover the mechanism underlying this differential regulation. In COS-7 cells, FKBP51 overexpression reduced GRα activity at a glucocorticoid response element-luciferase reporter, while increasing PPARγ activity at a peroxisome proliferator response element reporter. Conversely, FKBP51-deficient (knockout) (51KO) mouse embryonic fibroblasts (MEFs) showed elevated GRα but reduced PPARγ activities compared with those in wild-type MEFs. Phosphorylation is known to exert a similar pattern of reciprocal modulation of GRα and PPARγ. Knockdown of FKBP51 in 3T3-L1 preadipocytes increased phosphorylation of PPARγ at serine 112, a phospho-residue that inhibits activity. In 51KO cells, elevated phosphorylation of GRα at serines 220 and 234, phospho-residues that promote activity, was observed. Because FKBP51 is an essential chaperone to the Akt-specific phosphatase PH domain leucine-rich repeat protein phosphatase, Akt signaling was investigated. Elevated Akt activation and increased activation of p38 kinase, a downstream target of Akt that phosphorylates GRα and PPARγ, were seen in 51KO MEFs, causing activation and inhibition, respectively. Inactivation of p38 with PD169316 reversed the effects of FKBP51 deficiency on GRα and PPARγ activities and reduced PPARγ phosphorylation. Last, loss of FKBP51 caused a shift of PPARγ from cytoplasm to nucleus, as previously shown for GRα. A model is proposed in which FKBP51 loss reciprocally regulates GRα and PPARγ via 2 complementary mechanisms: activation of Akt-p38-mediated phosphorylation and redistribution of the receptors to the nucleus for direct targeting by p38.
Collapse
Affiliation(s)
- Lance A Stechschulte
- Center for Diabetes and Endocrine Research (L.A.S., T.D.H., S.S.G., S.M.N., E.R.S.), Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio 43614; and Herman B. Wells Center for Pediatric Research (W.S.), Section of Pediatric Cardiology, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana 46202
| | | | | | | | | | | |
Collapse
|
36
|
Song X, Shen YF, Cao M, Yuan Q, Tang Y, Lei C, Ji Yang Y. Dynamic expression of early responsible genes to acute left-ventricular ischemia in a time-dependent pattern. Anim Cells Syst (Seoul) 2014. [DOI: 10.1080/19768354.2014.911202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
37
|
Saito Y. Oxidized DJ-1 as a possible biomarker of Parkinson's disease. J Clin Biochem Nutr 2014; 54:138-144. [PMID: 24894116 PMCID: PMC4042152 DOI: 10.3164/jcbn.13-108] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 01/30/2014] [Indexed: 02/05/2023] Open
Abstract
Parkinson's disease is a progressive, age-related, neurodegenerative disorder, and oxidative stress is an important mediator in its pathogenesis. DJ-1 is a causative gene of a familial form of Parkinson's disease, namely PARK7, and plays a significant role in antioxidative defense to protect the cells from oxidative stress. DJ-1 undergoes preferential oxidation at the cysteine residue at position 106, Cys-106, under oxidative stress. The critical role of Cys-106 in the biological function of DJ-1 has been demonstrated, and recent studies indicate that DJ-1 acts as a sensor of oxidative stress by regulating the gene expression of antioxidative defense. Specific antibodies against Cys-106-oxidized DJ-1 have been developed, and the generation of oxidized DJ-1 in cellular and animal models of Parkinson's disease has been investigated. This review focuses on the role of DJ-1 in antioxidative defense and the importance of oxidizable Cys-106 in its function. The significance of the identification of early-phase Parkinson's disease biomarkers and the nature of oxidized DJ-1 as a biomarker for Parkinson's disease are discussed here.
Collapse
Affiliation(s)
- Yoshiro Saito
- Systems Life Sciences Laboratory, Department of Medical Life Systems, Faculty of Medical and Life Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| |
Collapse
|
38
|
Zhao Y, Liu XZ, Tian WW, Guan YF, Wang P, Miao CY. Extracellular visfatin has nicotinamide phosphoribosyltransferase enzymatic activity and is neuroprotective against ischemic injury. CNS Neurosci Ther 2014; 20:539-47. [PMID: 24750959 DOI: 10.1111/cns.12273] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2014] [Revised: 03/12/2014] [Accepted: 03/26/2014] [Indexed: 12/25/2022] Open
Abstract
AIM Visfatin, a novel adipokine, is predominantly produced by visceral adipose tissue and exists in intracellular and extracellular compartments. The intracellular form of visfatin is proved to be nicotinamide phosphoribosyltransferase (NAMPT) and exhibits neuroprotection through maintaining intracellular NAD(+) pool. However, whether extracellular form of visfatin has NAMPT activity and the effect of extracellular visfatin in cerebral ischemia are unknown. METHODS AND RESULTS Plasma concentrations of visfatin, NAD(+) , and ATP were increased in mice upon cerebral ischemia. Cultured glia, but not neuron, was able to secrete visfatin. Oxygen-glucose deprivation (OGD) stress increased the secretion of visfatin from glia. Extracellular recombinant mouse wild-type visfatin, but not mouse H247A-mutant enzymatic-dead visfatin, had NAMPT enzymatic function in vitro. Treatment of wild-type visfatin, but not H247A-mutant enzymatic-dead visfatin, significantly attenuated detrimental effect of OGD on the cell viability and apoptosis in both cultured mouse neuron and glia. Treatment of neutralizing antibody, abolished the protective effect of extracellular visfatin on cell viability, but failed to block the antiapoptotic effect of extracellular visfatin. At last, we observed that plasma visfatin concentrations decreased in 6-month-old but not 3-month-old SHR-SP compared with that in age-matched Wistar-Kyoto rats. Inhibition of NAMPT enzymatic function of visfatin (by FK866) accelerated the occurrence of stroke in SHR-SP. CONCLUSIONS Extracellular visfatin has NAMPT enzymatic activity and maybe be neuroprotective just as intracellular visfatin in cerebral ischemic injury.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Pharmacology, Second Military Medical University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
39
|
Wang Y, Liu W, He X, Zhou F. Parkinson's Disease-Associated Dj-1 Mutations Increase Abnormal Phosphorylation of Tau Protein through Akt/Gsk-3β Pathways. J Mol Neurosci 2013; 51:911-8. [DOI: 10.1007/s12031-013-0099-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 08/12/2013] [Indexed: 10/26/2022]
|