1
|
Li Q, Wang Z, Li F, Liu S, Ding Y, Yan J, Feng X, Li M. AIM2 exacerbates hypoxic-ischemic brain damage in neonatal rats via promoting neuronal pyroptosis. Brain Res Bull 2025; 224:111305. [PMID: 40101806 DOI: 10.1016/j.brainresbull.2025.111305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/03/2025] [Accepted: 03/12/2025] [Indexed: 03/20/2025]
Abstract
BACKGROUND Pyroptosis has been reported to play a pathogenic role in neonatal hypoxic-ischemic brain damage (HIBD). Absence in melanoma 2 (AIM2) is an inflammasome involved in pyroptosis. OBJECTIVE This study aimed to investigate the role of AIM2 in hypoxic-ischemia (HI)-induced pyroptosis and brain damage in a neonatal rat HIBD model. METHODS In vivo, we injected a lentivirus that overexpressed or knocked down AIM2 into the lateral ventricle of rats within 24 h after birth and prepared a 7-day Sprague Dawley (SD) rat HIBD model. In vitro, we transfected lentiviruses overexpressing or knocking down AIM2 into cultured primary neurons and established an oxygen/glucose deprivation/reoxygenation (OGD/R) model. 2,3,5-triphenyltetrazolium chloride (TTC) staining was used to determine infarct size. Hematoxylin and eosin and Nissl staining were used to evaluate morphological changes in the damaged brain. Cell Counting Kit-8 (CCK-8) and lactate dehydrogenase (LDH) assays were used to determine cell viability and toxicity. Pyroptosis was observed using transmission electron microscopy. RESULTS AIM2 expression significantly increased in the HI-induced cortex of neonatal rats. Lentivirus-mediated overexpression of AIM2 significantly aggravates HI-induced brain injury and OGD/R-induced neuronal injury in vivo and in vitro. The lentivirus-mediated AIM2 knockdown significantly reversed these adverse effects. In addition, AIM2 overexpression increased HI-induced pyroptosis in neonatal rats in vivo and in vitro, whereas AIM2 knockdown suppressed HI-induced pyroptosis via the AIM2/Caspase-1/GSDMD pathway. CONCLUSION These findings show that the upregulation of AIM2 activates pyroptosis and plays a pathogenic role in neonatal HIBD.
Collapse
Affiliation(s)
- Qianqian Li
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, Jiangsu Province, China; Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zengqin Wang
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, Jiangsu Province, China
| | - Fengli Li
- Department of Intensive Care Unit, Zibo Central Hospital, Zibo, Shandong Province, China
| | - Songlin Liu
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, Jiangsu Province, China
| | - Yuhong Ding
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, Jiangsu Province, China
| | - Junmei Yan
- Department of Neonatology, Xuzhou Maternity and Child Health Care Hospital, Xuzhou, Jiangsu Province, China.
| | - Xing Feng
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China.
| | - Mei Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China.
| |
Collapse
|
2
|
Song C, Zhao L, Deng J, Wang L, Mao M, Peng S, Tang W. E2F8-induced GRPEL2 promoted colorectal cancer progression via targeting TIGAR. J Transl Med 2025; 23:466. [PMID: 40269881 PMCID: PMC12020167 DOI: 10.1186/s12967-025-06451-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 04/02/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is the leading cause for cancer mortality across the world. GRPEL2 is a critical regulator of mitochondria's function with an oncogenic role in different kinds of cancer. The exact function of GRPEL2 -driven mitochondrial regulation and CRC progression have not been elucidated. METHODS RNA-seq data from TCGA database was analyzed to identify biomarkers and therapeutic targets of CRC. The gene expression profile was validated by quantitative real-time PCR on 68 paired tumor and non-tumor samples from CRC patients. Tumorigenesis regulated by GRPEL2 was tested through EdU staining, Transwell assay, in vivo tumor growth and in vivo metastasis. The function of Mitochondria mediated by GRPEL2 was evaluated by transmission electron microscopy, DCFH-DA staining, mitochondrial membrane potential detection, and Calcein staining. LC-MS/MS screening and Co-IP were performed to discover protein partners of GRPEL2. E2F8-mediated transcriptional regulation of GRPEL2 was verified via Luciferase reporter and ChIP assays. RESULTS GRPEL2 was upregulated in CRC tumor tissues and cell lines. High expression of GRPEL2 was associated with poor prognosis of CRC and inhibition of GRPEL2 suppressed CRC proliferation and migration by inducing mitochondria injury. Meanwhile, TIGAR was shown to interact with GRPEL2 and overexpression of TIGAR rescued CRC progression in the presence of GRPEL2 inhibition. Moreover, E2F8 was the upstream regulator of GRPEL2, which positively induced GRPEL2 transcription and expression in CRC. CONCLUSION Our work illustrated the oncogenic role of GRPEL2 in CRC development and determined the molecular mechanism of E2F8/GRPEL2/TIGAR pathway. These findings will provide novel insights and promising therapeutic targets for CRC treatment in the future.
Collapse
Affiliation(s)
- Cheng Song
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Lei Zhao
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Jing Deng
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Li Wang
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Min Mao
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Siyuan Peng
- Center of Integrated Traditional Chinese and Western Medicine, Hunan Cancer Hospital, Changsha, Hunan, China
| | - Wei Tang
- Department of Oncology, Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, No. 58, Lushan Road, Yuelu District, Changsha, Hunan, China.
| |
Collapse
|
3
|
Chen L, Tang J, Liu XQ, Li QQ, Li JY, Li YY, Zheng WH, Qin ZH, Sheng R. TIGAR Suppresses ER Stress-Induced Neuronal Injury through Targeting ATF4 Signaling in Cerebral Ischemia/Reperfusion. J Neurosci 2025; 45:e1406242025. [PMID: 39919831 PMCID: PMC11949484 DOI: 10.1523/jneurosci.1406-24.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 01/03/2025] [Accepted: 01/31/2025] [Indexed: 02/09/2025] Open
Abstract
Endoplasmic reticulum (ER) stress is crucial in cerebral ischemia/reperfusion injury by triggering cellular apoptosis and exacerbating neuronal damage. This study elucidates the dynamics of TP53-induced glycolysis and apoptosis regulator (TIGAR) translocation and its role in regulating neural fate during cerebral ischemia-induced ER stress, specifically in male mice. We found enhanced nuclear localization of TIGAR in neurons after transient middle cerebral artery occlusion/reperfusion (tMCAO/R) in male mice, as well as oxygen glucose deprivation/reperfusion (OGD/R) and treatment with ER stress inducer (tunicamycin and thapsigargin) in neuronal cells. Conditional neuronal knockdown of Tigar aggravated the injury following ischemia-reperfusion, whereas overexpression of Tigar attenuated cerebral ischemic injury and ameliorated intraneuronal ER stress. Additionally, TIGAR overexpression reduced the elevation of ATF4 target genes and attenuated ER stress-induced cell death. Notably, TIGAR colocalized and interacted with ATF4 in the nucleus, inhibiting its downstream proapoptotic gene transcription, consequently protecting against ischemic injury. In vitro and in vivo experiments revealed that ATF4 overexpression reversed the protective effects of TIGAR against cerebral ischemic injury. Intriguingly, our study identified the Q141/K145 residues of TIGAR, crucial for its nuclear translocation and interaction with ATF4, highlighting a novel aspect of TIGAR's function distinct from its known phosphatase activity or mitochondrial localization domains. These findings reveal a novel neuroprotective mechanism of TIGAR in regulating ER stress through ATF4-mediated signaling pathways. These insights may guide targeted therapeutic strategies to protect neuronal function and alleviate the deleterious effects of cerebral ischemic injury.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou 215123, China
| | - Jie Tang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou 215123, China
| | - Xue-Qing Liu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou 215123, China
| | - Qi-Qi Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou 215123, China
| | - Jia-Ying Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou 215123, China
| | - Yan-Yan Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou 215123, China
| | - Wen-Hua Zheng
- Center of Reproduction, Development and Aging, Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Macau SAR 519000, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou 215123, China
- Institute of Health Technology, Suzhou Gaobo Vocational College, Suzhou High-Technology District, Science & Technology Town, Suzhou 215163, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou 215123, China
| |
Collapse
|
4
|
Sun M, Wang Y, Xu H, Shen Y, Liu B, Ma Y, Jiang C, Wang S, Li Q, Lu Y, Han F, Li T, Qin Y. Novel Hypochlorous Acid-Activated Near-Infrared Probe Monitors the Dynamic Changes of Myeloperoxidase Activity in Ischemic Brain. J Med Chem 2025; 68:5382-5399. [PMID: 40014579 DOI: 10.1021/acs.jmedchem.4c02431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Myeloperoxidase (MPO) contributes to the progression of ischemic damage. To fully understand MPO biology, highly sensitive and specific probes that can trace the activity of endogenous MPO fluxes are indispensable. Here, we developed two hypochlorous acid (HClO)-activated near-infrared probes to image MPO activity in a noninvasive manner. The probe MPO-NIR-II could track MPO-induced HClO in real time and in situ upon various stimuli with high sensitivity and specificity. Furthermore, MPO-NIR-II could monitor the MPO activity by in vivo fluorescence imaging and confocal laser scanning microscopy in mice with ischemic stroke. Moreover, a high-content screening system for MPO inhibitors was established by combining MPO-NIR-II with MPO-overexpressed cells and mouse brain slices with ischemic stroke, and the candidate compound AZD5904 was found to effectively attenuate ischemic brain injury. Overall, this work provides a versatile fluorescence tool that holds great promise for visualizing endogenous MPO fluxes of brain ischemia.
Collapse
Affiliation(s)
- Meiling Sun
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Yuting Wang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Huijun Xu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuting Shen
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Bin Liu
- Department of Geriatrics, The Affiliated Brain Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Yuchen Ma
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Chenchen Jiang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Supeng Wang
- The First Clinical Medical College of Nanjing Medical University, Nanjing 211166, China
| | - Qi Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yingmei Lu
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Feng Han
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Tingyou Li
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yajuan Qin
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
5
|
Xiao J, Wu M, Li H, Zhang S, Deng J, Wu B. TIGAR alleviates cognitive impairment in rats with chronic cerebral hypoperfusion by suppressing oxidative stress and pyroptosis. Am J Transl Res 2025; 17:1223-1236. [PMID: 40092124 PMCID: PMC11909525 DOI: 10.62347/nwqs1671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 01/18/2025] [Indexed: 03/19/2025]
Abstract
BACKGROUND It is postulated that oxidative stress and pyroptosis, which are induced by chronic cerebral hypoperfusion (CCH), contribute to the pathogenesis of vascular cognitive impairment (VCI). The protective role of TIGAR in neurologic illnesses has been the subject of extensive examination, yet its role in models of CCH-induced cognitive impairment remains unexplored. The objective of this study was to ascertain whether TIGAR is a neuroprotective agent in rats with CCH that reduces oxidative stress and pyroptosis. METHODS A CCH model was established in rats through the use of bilateral common carotid artery occlusion (BCCAO). The effects of TIGAR on cognitive function and anxiety-depressive behaviors in rats with CCH were examined. To this end, the Y-maze and open field tests were employed. Nissl and hematoxylin-eosin (H&E) staining were used to assess histologic changes in the CA1 area of the hippocampus. Hippocampal glutathione (GSH) activity, malondialdehyde (MDA) content, and NADPH/NADP+ ratio were measured using the WST-8 colorimetric method to assess oxidative stress. The expression of TIGAR and pyroptosis-related proteins was assessed by western blotting. RESULTS A model of CCH-induced cognitive impairment was successfully established. Four weeks after BCCAO, cerebral blood flow returned to normal in the rats, and cognitive impairment and anxiety-depression-like behavior developed. In rats with CCH, MDA levels increased, GSH activity and NADPH/NADP+ ratio decreased, and pyroptosis-related protein expression increased. The pathologic findings indicated that there was an exacerbation of neuronal injury in the CA1 area of the hippocampus and that the cells were loosely arranged. In rats with CCH, overexpression of TIGAR reduced pyroptosis-associated protein expression while increasing MDA content, GSH activity, and NADPH/NADP+ ratio. It promoted neuronal cell survival, and improved cognitive function and anxiety-depression-like behavior. CONCLUSION Overexpression of TIGAR reduced CCH-induced oxidative stress and pyroptosis and ameliorated cognitive dysfunction and anxiety-depression-like behavior in rats. These findings suggest that TIGAR counteracts oxidative stress and prevents pyroptosis, making it a promising target for the treatment of VCI.
Collapse
Affiliation(s)
- Jie Xiao
- Department of Internal Medicine, Nanchong Mental Health Center Nanchong 637000, Sichuan, China
- Innovation Centre for Science and Technology, North Sichuan Medical College Nanchong 637000, Sichuan, China
| | - Min Wu
- Department of Internal Medicine, Nanchong Mental Health Center Nanchong 637000, Sichuan, China
- Innovation Centre for Science and Technology, North Sichuan Medical College Nanchong 637000, Sichuan, China
| | - Hailong Li
- Innovation Centre for Science and Technology, North Sichuan Medical College Nanchong 637000, Sichuan, China
- Department of Gastroenterology, Wusheng People's Hospital Guang'an 638500, Sichuan, China
| | - Shufan Zhang
- Department of Internal Medicine, Nanchong Mental Health Center Nanchong 637000, Sichuan, China
- Innovation Centre for Science and Technology, North Sichuan Medical College Nanchong 637000, Sichuan, China
| | - Jing Deng
- Department of Internal Medicine, Nanchong Mental Health Center Nanchong 637000, Sichuan, China
- Innovation Centre for Science and Technology, North Sichuan Medical College Nanchong 637000, Sichuan, China
| | - Bihua Wu
- Department of Internal Medicine, Nanchong Mental Health Center Nanchong 637000, Sichuan, China
| |
Collapse
|
6
|
Li Y, Gao Y, Yu G, Ye Y, Zhu H, Wang J, Li Y, Chen L, Gu L. G6PD protects against cerebral ischemia-reperfusion injury by inhibiting excessive mitophagy. Life Sci 2025; 362:123367. [PMID: 39756510 DOI: 10.1016/j.lfs.2024.123367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 12/20/2024] [Accepted: 12/31/2024] [Indexed: 01/07/2025]
Abstract
AIMS Cerebral ischemia-reperfusion injury (CIRI) exacerbates post-stroke brain damage. We aimed to understand the role of glucose-6-phosphate dehydrogenase (G6PD) in CIRI and mitophagy. MATERIALS AND METHODS Lentivirus and small interfering RNA were utilized to suppress G6PD in tissues and cells, leading to the establishment of in vivo and in vitro models of ischemia-reperfusion following middle cerebral artery occlusion (MCAO) and oxygen-glucose deprivation/ reoxygenation (OGD/R). The expression and function of G6PD were investigated through differential gene analysis and weighted correlation network analysis (WGCNA), immunofluorescence, and western blotting (WB). KEY FINDINGS G6PD mRNA levels increased 3 d after MCAO, and G6PD protein expression was elevated in the ischemic penumbra of mice and HT22 cells following OGD/R. G6PD knockdown increased neural deficits, enlarged infarct volume in mice after CIRI, and reduced HT22 cell survival during OGD/R. WGCNA indicated a correlation between G6PD and mitophagy in CIRI. Following G6PD knockdown, the p-DRP1/DRP ratio increased, the PINK1/Parkin pathway was further activated, and TOMM20 expression was downregulated. The mitophagy inhibitor Mdivi-1 reversed these changes, as well as the nerve damage caused by G6PD knockdown, and alleviated mitochondrial damage in the ischemic penumbra. SIGNIFICANCE The role of G6PD in CIRI was revealed and its interaction with mitophagy was explored, providing important insights for understanding the molecular mechanism of CIRI and developing new therapeutic strategies.
Collapse
Affiliation(s)
- Yina Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yikun Gao
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Guixiang Yu
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China; Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Hua Zhu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China; Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Jin Wang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China; Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Yilin Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China
| | - Lei Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China; Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Province, China.
| |
Collapse
|
7
|
Sun M, Wang L, Cao Q, Wang X, Zhang Y, Guo M, Chen J, Ma Y, Niu L, Zhang Y, Hu M, Gu M, Zhu Z, Yao X, Yao J, Zhao C, Wu J, Liu X, Lu Y, Wang Z, Xiang Q, Han F, Zhu D. Discovery of HZS60 as a Novel Brain Penetrant NMDAR/TRPM4 Interaction Interface Inhibitor with Improved Activity and Pharmacokinetic Properties for the Treatment of Cerebral Ischemia. J Med Chem 2025; 68:2008-2043. [PMID: 39745498 DOI: 10.1021/acs.jmedchem.4c02772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
The death signaling complex comprising extrasynaptic NMDAR and TRPM4 plays a pivotal role in the pathogenesis of ischemic stroke. Targeting the protein-protein interactions between NMDAR and TRPM4 represents a promising therapeutic strategy for ischemic stroke. Herein, we describe the discovery of a novel series of NMDAR/TRPM4 interaction interface inhibitors aimed at enhancing neuroprotective efficacy and optimizing pharmacokinetic profiles. The representative compound HZS60 displayed significant neuroprotective effects against both NMDA and oxygen-glucose deprivation/reoxygenation-induced ischemic injury in primary neurons. Notably, HZS60 exhibited a favorable pharmacokinetic profile and excellent brain permeability. Furthermore, HZS60 provided effective neuroprotection following brain ischemia and reperfusion injury in vivo. Collectively, these findings underscore the potential of HZS60 as a promising candidate for the development of novel therapeutic strategies for ischemic stroke.
Collapse
Affiliation(s)
- Meiling Sun
- Department of Neurology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Lin Wang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Qiaofeng Cao
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xuechun Wang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ying Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Manyu Guo
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jie Chen
- Department of Neurology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Yuchen Ma
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Le Niu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yanping Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Mengdie Hu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Mengli Gu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Zhihui Zhu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xinyi Yao
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Junchen Yao
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Chen Zhao
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jin Wu
- Department of Neurology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Xiuxiu Liu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yingmei Lu
- Department of Neurology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Zhen Wang
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, #345 Lingling Rd., Shanghai 200032, China
| | - Qiuping Xiang
- Guoke Ningbo Life Science and Health Industry Research Institute, Ningbo No.2 Hospital, Ningbo 315000, China
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- The affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Northern Jiangsu Institute of Clinical Medicine, Huaian 223300, China
| | - Dongsheng Zhu
- Department of Neurology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
8
|
Sajrawi C, Odeh M, Tiwari AK, Agranovich B, Abramovich I, Zubedat S, Saar G, Shaulov L, Avital A, Reznik D, Benhar M, Radzishevsky I, Engelender S, Wolosker H. Endogenous histidine peptides are physiological antioxidants that prevent oligodendrocyte cell death and myelin loss in vivo. Glia 2025; 73:122-139. [PMID: 39360557 DOI: 10.1002/glia.24624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Histidine dipeptides (HDs) are synthesized in brain oligodendrocytes by carnosine synthase (carns1), but their role is unknown. Using metabolomics and in vivo experiments with both constitutive and oligodendrocyte-selective carns1-KO mouse models, we found that HDs are critical for oligodendrocyte survival and protect against oxidative stress. Carns1-KO mouse models had lower numbers of mature oligodendrocytes, increased lipid peroxidation, and behavioral changes. Cuprizone administration, which increases reactive oxygen species in vivo, resulted in higher oligodendrocyte death, demyelination, axonal alterations, and oxidative damage in the corpus callosum of carns1-KO mice. Gliosis and oxidative damage by cuprizone were prevented by pretreatment with the antioxidant N-acetylcysteine. NADPH levels were increased threefold in the brains of carns1-KO mice as an antioxidant response to oxidative stress through acceleration of the pentose phosphate pathway (PPP). This was due to overexpression of glucose-6-phosphate dehydrogenase, the rate-limiting enzyme of the PPP. Likewise, expression of NAD kinase, the biosynthetic enzyme for NADP+, and NAMPT, which replenishes the NAD+ pool, was higher in carns1-KO mice brains than in controls. Our observations suggest that HDs cell-autonomously protect oligodendrocytes from oxidative stress, with implications for demyelinating diseases.
Collapse
Affiliation(s)
- Clara Sajrawi
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Maali Odeh
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Akshay K Tiwari
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Bella Agranovich
- Laura and Isaac Perlmutter Metabolomics Center, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ifat Abramovich
- Laura and Isaac Perlmutter Metabolomics Center, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Salman Zubedat
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| | - Galit Saar
- In vivo Imaging Unit, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Lihi Shaulov
- Electron Microscopy Unit, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Avi Avital
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| | - Dan Reznik
- Data Science Consulting, Rio de Janeiro, RJ, Brazil
| | - Moran Benhar
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Inna Radzishevsky
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Simone Engelender
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Herman Wolosker
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Laura and Isaac Perlmutter Metabolomics Center, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
9
|
Li Y, Song H, Xu J, Wang Y, Bai L, Wang H, Zhang J. TIGAR relieves PCOS by inhibiting granulosa cell apoptosis and oxidative stress through activating Nrf2. Mol Cell Endocrinol 2024; 594:112381. [PMID: 39341450 DOI: 10.1016/j.mce.2024.112381] [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: 06/27/2024] [Revised: 09/13/2024] [Accepted: 09/25/2024] [Indexed: 10/01/2024]
Abstract
This study aimed to elucidate the role of TP53-induced glycolysis and apoptosis regulator (TIGAR) in polycystic ovary syndrome (PCOS). A rat model PCOS was constructed by subcutaneous injection with dehydroepiandrosterone (DHEA). Follicular atresia and reduced granular cells (GCs) in ovaries suggested successful modeling. The low expression of TIGAR was observed in ovarian tissue of PCOS rat. To explore the role of TIGAR in PCOS, lentivirus carrying the TIGAR were used to up-regulate TIGAR expression. TIGAR overexpression reduced the DHEA-induced increase of ovarian weight, the levels of estradiol (E2), and the ratio of luteinizing hormone/follicle-stimulating hormone (LH/FSH) in the serum, as well as improved the morphology of the follicle, especially increased the thickness of the GC layer, which attributed to the inhibition of apoptosis by TIGAR. In addition, high expression of TIGAR inhibited oxidative stress in ovaries of PCOS rat, as evidenced by decreased level of malondialdehyde (MDA), and reactive oxygen species (ROS), and enhanced activity of glutathione peroxidase (GPX) and superoxide dismutase (SOD). Mechanically, Nrf2/OH-1 signal pathway was activated by TIGAR. The effect of TIGAR on PCOS were verified in the primary rat GCs treated with dihydrotestosterone, but also the rescue experiment was performed. Downregulation of Nrf2 reversed the effects of TIGAR, indicating that TIGAR suppressed oxidative stress and GC apoptosis by activating Nrf2/OH-1 pathway in PCOS. Finally, non-targeted metabolomics revealed that TIGAR might affect the energy metabolic pathway, thereby altering the metabolic profile of primary rat GCs. This study provided new insights into the prevention and treatment of PCOS.
Collapse
Affiliation(s)
- Yan Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China
| | - Hui Song
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China
| | - Jia Xu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China
| | - Yunping Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China
| | - Lu Bai
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China
| | - Haixu Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China.
| | - Jianfang Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of the Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
10
|
Li G, Ma Y, Zhang S, Lin W, Yao X, Zhou Y, Zhao Y, Rao Q, Qu Y, Gao Y, Chen L, Zhang Y, Han F, Sun M, Zhao C. A mechanistic systems biology model of brain microvascular endothelial cell signaling reveals dynamic pathway-based therapeutic targets for brain ischemia. Redox Biol 2024; 78:103415. [PMID: 39520909 PMCID: PMC11584692 DOI: 10.1016/j.redox.2024.103415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 10/31/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
Ischemic stroke is a significant threat to human health. Currently, there is a lack of effective treatments for stroke, and progress in new neuron-centered drug target development is relatively slow. On the other hand, studies have demonstrated that brain microvascular endothelial cells (BMECs) are crucial components of the neurovascular unit and play pivotal roles in ischemic stroke progression. To better understand the complex multifaceted roles of BMECs in the regulation of ischemic stroke pathophysiology and facilitate BMEC-based drug target discovery, we utilized a transcriptomics-informed systems biology modeling approach and constructed a mechanism-based computational multipathway model to systematically investigate BMEC function and its modulatory potential. Extensive multilevel data regarding complex BMEC pathway signal transduction and biomarker expression under various pathophysiological conditions were used for quantitative model calibration and validation, and we generated dynamic BMEC phenotype maps in response to various stroke-related stimuli to identify potential determinants of BMEC fate under stress conditions. Through high-throughput model sensitivity analyses and virtual target perturbations in model-based single cells, our model predicted that targeting succinate could effectively reverse the detrimental cell phenotype of BMECs under oxygen and glucose deprivation/reoxygenation, a condition that mimics stroke pathogenesis, and we experimentally validated the utility of this new target in terms of regulating inflammatory factor production, free radical generation and tight junction protection in vitro and in vivo. Our work is the first that complementarily couples transcriptomic analysis with mechanistic systems-level pathway modeling in the study of BMEC function and endothelium-based therapeutic targets in ischemic stroke.
Collapse
Affiliation(s)
- Geli Li
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China; Gusu School, Nanjing Medical University, 215000, Suzhou, China
| | - Yuchen Ma
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Sujie Zhang
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Wen Lin
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Xinyi Yao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yating Zhou
- The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China
| | - Yanyong Zhao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Qi Rao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yuchen Qu
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China
| | - Yuan Gao
- QSPMed Technologies, 210000, Nanjing, China
| | - Lianmin Chen
- The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China
| | - Yu Zhang
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, 21205, Baltimore, USA
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, Drug Target and Drug Discovery Center, School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China.
| | - Meiling Sun
- School of Basic Medical Sciences, Nanjing Medical University, 210000, Nanjing, China.
| | - Chen Zhao
- School of Pharmacy, Nanjing Medical University, 210000, Nanjing, China; The First Affiliated Hospital of Nanjing Medical University, 210000, Nanjing, China.
| |
Collapse
|
11
|
Nam HY, Park SH, Lee GH, Kim EY, Lee S, Chang HW, Chang EJ, Choi KC, Kim SW. TIGAR coordinates senescence-associated secretory phenotype via lysosome repositioning and α-tubulin deacetylation. Exp Mol Med 2024; 56:2726-2738. [PMID: 39633033 DOI: 10.1038/s12276-024-01362-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/06/2024] [Accepted: 09/25/2024] [Indexed: 12/07/2024] Open
Abstract
TP53-induced glycolysis and apoptosis regulator (TIGAR) regulates redox homeostasis and provides the intermediates necessary for cell growth by reducing the glycolytic rate. During cellular senescence, cells undergo metabolic rewiring towards the glycolytic pathway, along with the development of the senescence-associated secretory phenotype (SASP), also known as the secretome. We observed that TIGAR expression increased during replicative senescence following the in vitro expansion of human mesenchymal stromal cells (MSCs) and that TIGAR knockout (KO) decreased SASP factors and triggered premature senescence with decelerated progression. Additionally, TIGAR KO impaired flexible lysosomal movement to the perinuclear region and decreased the autophagic flux of MSCs. Research on the mechanism of lysosomal movement revealed that, while native senescent MSCs presented low levels of Ac-α-tubulin (lysine 40) and increased sirtuin 2 (SIRT2) activity compared with those in growing cells, TIGAR KO-MSCs maintained Ac-α-tubulin levels and exhibited decreased SIRT2 activity despite being in a senescent state. The overexpression of SIRT2 reduced Ac-α-tubulin as a protein target of SIRT2 and induced the positioning of lysosomes at the perinuclear region, restoring the cytokine secretion of TIGAR KO-MSCs. Furthermore, TIGAR expression was positively correlated with SIRT2 activity, indicating that TIGAR affects SIRT2 activity partly by modulating the NAD+ level. Thus, our study demonstrated that TIGAR provides a foundation that translates the regulation of energy metabolism into lysosome positioning, affecting the secretome for senescence development. Considering the functional value of the cell-secretome in aging-related diseases, these findings suggest the feasibility of TIGAR for the regulation of secretory phenotypes.
Collapse
Affiliation(s)
- Hae Yun Nam
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| | - Seung-Ho Park
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Geun-Hee Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Eun-Young Kim
- Department Hematology and Medical Oncology, Whinship Cancer Institute of Emory University, Atlanta, GA, 30322, USA
| | - SangEun Lee
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Hyo Won Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Eun-Ju Chang
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea
| | - Kyung-Chul Choi
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| | - Seong Who Kim
- Department of Biochemistry and Molecular Biology, Brain Korea 21 project, Asan Medical Center, University of Ulsan College of Medicine, Seoul, 05505, South Korea.
| |
Collapse
|
12
|
Wang H, Liu S, Sun Y, Chen C, Hu Z, Li Q, Long J, Yan Q, Liang J, Lin Y, Yang S, Lin M, Liu X, Wang H, Yu J, Yi F, Tan Y, Yang Y, Chen N, Ai Q. Target modulation of glycolytic pathways as a new strategy for the treatment of neuroinflammatory diseases. Ageing Res Rev 2024; 101:102472. [PMID: 39233146 DOI: 10.1016/j.arr.2024.102472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 09/06/2024]
Abstract
Neuroinflammation is an innate and adaptive immune response initiated by the release of inflammatory mediators from various immune cells in response to harmful stimuli. While initially beneficial and protective, prolonged or excessive neuroinflammation has been identified in clinical and experimental studies as a key pathological driver of numerous neurological diseases and an accelerant of the aging process. Glycolysis, the metabolic process that converts glucose to pyruvate or lactate to produce adenosine 5'-triphosphate (ATP), is often dysregulated in many neuroinflammatory disorders and in the affected nerve cells. Enhancing glucose availability and uptake, as well as increasing glycolytic flux through pharmacological or genetic manipulation of glycolytic enzymes, has shown potential protective effects in several animal models of neuroinflammatory diseases. Modulating the glycolytic pathway to improve glucose metabolism and ATP production may help alleviate energy deficiencies associated with these conditions. In this review, we examine six neuroinflammatory diseases-stroke, Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), amyotrophic lateral sclerosis (ALS), and depression-and provide evidence supporting the role of glycolysis in their treatment. We also explore the potential link between inflammation-induced aging and glycolysis. Additionally, we briefly discuss the critical role of glycolysis in three types of neuronal cells-neurons, microglia, and astrocytes-within physiological processes. This review highlights the significance of glycolysis in the pathology of neuroinflammatory diseases and its relevance to the aging process.
Collapse
Affiliation(s)
- Hanlong Wang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Shasha Liu
- Department of Pharmacy, Changsha Hospital for Matemal&Child Health Care Affiliated to Hunan Normal University, Changsha 410007, China
| | - Yang Sun
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Chen Chen
- Department of Pharmacy, The First Hospital of Lanzhou University, Lanzhou 730000, China
| | - Ziyi Hu
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Qinqin Li
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Junpeng Long
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Qian Yan
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jinping Liang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Yuting Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Meiyu Lin
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Xuan Liu
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Huiqin Wang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Jingbo Yu
- Technology Innovation Center/National Key Laboratory Breeding Base of Chinese Medicine Powders and Innovative Drugs, Hunan University of Chinese Medicine, Changsha 410208, China
| | - Fan Yi
- Key Laboratory of Cosmetic, China National Light Industry, Beijing Technology and Business University, Beijing 100048, China
| | - Yong Tan
- Nephrology Department, Xiangtan Central Hospital, Xiangtan 411100, China
| | - Yantao Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| | - Naihong Chen
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Qidi Ai
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China.
| |
Collapse
|
13
|
Jiang XL, Zhang ZB, Feng CX, Lin CJ, Yang H, Tan LL, Ding X, Xu LX, Li G, Pan T, Qin ZH, Sun B, Feng X, Li M. PHLDA1 contributes to hypoxic ischemic brain injury in neonatal rats via inhibiting FUNDC1-mediated mitophagy. Acta Pharmacol Sin 2024; 45:1809-1820. [PMID: 38750074 PMCID: PMC11336168 DOI: 10.1038/s41401-024-01292-x] [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: 01/22/2024] [Accepted: 04/14/2024] [Indexed: 08/22/2024]
Abstract
Hypoxia-ischemia (HI) is one of the main causes of neonatal brain injury. Mitophagy has been implicated in the degradation of damaged mitochondria and cell survival following neonatal brain HI injury. Pleckstrin homology-like domain family A member 1 (PHLDA1) plays vital roles in the progression of various disorders including the regulation of oxidative stress, the immune responses and apoptosis. In the present study we investigated the role of PHLDA1 in HI-induced neuronal injury and further explored the mechanisms underlying PHLDA1-regulated mitophagy in vivo and in vitro. HI model was established in newborn rats by ligation of the left common carotid artery plus exposure to an oxygen-deficient chamber with 8% O2 and 92% N2. In vitro studies were conducted in primary hippocampal neurons subjected to oxygen and glucose deprivation/-reoxygenation (OGD/R). We showed that the expression of PHLDA1 was significantly upregulated in the hippocampus of HI newborn rats and in OGD/R-treated primary neurons. Knockdown of PHLDA1 in neonatal rats via lentiviral vector not only significantly ameliorated HI-induced hippocampal neuronal injury but also markedly improved long-term cognitive function outcomes, whereas overexpression of PHLDA1 in neonatal rats via lentiviral vector aggravated these outcomes. PHLDA1 knockdown in primary neurons significantly reversed the reduction of cell viability and increase in intracellular reactive oxygen species (ROS) levels, and attenuated OGD-induced mitochondrial dysfunction, whereas overexpression of PHLDA1 decreased these parameters. In OGD/R-treated primary hippocampal neurons, we revealed that PHLDA1 knockdown enhanced mitophagy by activating FUNDC1, which was abolished by FUNDC1 knockdown or pretreatment with mitophagy inhibitor Mdivi-1 (25 μM). Notably, pretreatment with Mdivi-1 or the knockdown of FUNDC1 not only increased brain infarct volume, but also abolished the neuroprotective effect of PHLDA1 knockdown in HI newborn rats. Together, these results demonstrate that PHLDA1 contributes to neonatal HI-induced brain injury via inhibition of FUNDC1-mediated neuronal mitophagy.
Collapse
Affiliation(s)
- Xiao-Lu Jiang
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Zu-Bin Zhang
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China.
- Jiangsu Key Laboratory for Translational Research and Therapeutics of NeuroPsycho Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China.
| | - Chen-Xi Feng
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Chen-Jie Lin
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Hui Yang
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Lan-Lan Tan
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xin Ding
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Li-Xiao Xu
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Gen Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Tao Pan
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Zheng-Hong Qin
- Institute of Health Technology, Global Institute of Software Technology, Qingshan Road, Suzhou Science & Technology Tower, Hi-Tech Area, Suzhou, 215163, China
| | - Bin Sun
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Xing Feng
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China.
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, 215025, China.
- Soochow Key Laboratory of Prevention and Treatment of Child Brain Injury, Children's Hospital of Soochow University, Suzhou, 215025, China.
| | - Mei Li
- Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, 215025, China.
| |
Collapse
|
14
|
Wang X, Li M, Wang F, Mao G, Wu J, Han R, Sheng R, Qin Z, Ni H. TIGAR reduces neuronal ferroptosis by inhibiting succinate dehydrogenase activity in cerebral ischemia. Free Radic Biol Med 2024; 216:89-105. [PMID: 38494143 DOI: 10.1016/j.freeradbiomed.2024.03.011] [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: 10/21/2023] [Revised: 01/12/2024] [Accepted: 03/14/2024] [Indexed: 03/19/2024]
Abstract
Ischemia Stroke (IS) is an acute neurological condition with high morbidity, disability, and mortality due to a severe reduction in local cerebral blood flow to the brain and blockage of oxygen and glucose supply. Oxidative stress induced by IS predisposes neurons to ferroptosis. TP53-induced glycolysis and apoptosis regulator (TIGAR) inhibits the intracellular glycolytic pathway to increase pentose phosphate pathway (PPP) flux, promotes NADPH production and thus generates reduced glutathione (GSH) to scavenge reactive oxygen species (ROS), and thus shows strong antioxidant effects to ameliorate cerebral ischemia/reperfusion injury. However, in the current study, prolonged ischemia impaired the PPP, and TIGAR was unable to produce NADPH but was still able to reduce neuronal ferroptosis and attenuate ischemic brain injury. Ferroptosis is a form of cell death caused by free radical-driven lipid peroxidation, and the vast majority of ROS leading to oxidative stress are generated by mitochondrial succinate dehydrogenase (SDH) driving reverse electron transfer (RET) via the mitochondrial electron transport chain. Overexpression of TIGAR significantly inhibited hypoxia-induced enhancement of SDH activity, and TIGAR deficiency further enhanced SDH activity. We also found that the inhibitory effect of TIGAR on SDH activity was related to its mitochondrial translocation under hypoxic conditions. TIGAR may inhibit SDH activity by mediating post-translational modifications (acetylation and succinylation) of SDH A through interaction with SDH A. SDH activity inhibition reduces neuronal ferroptosis by decreasing ROS production, eliminating MitoROS levels and attenuating lipid peroxide accumulation. Notably, TIGAR-mediated inhibition of SDH activity and ferroptosis was not dependent on the PPP-NADPH-GPX4 pathways. In conclusion, mitochondrial translocation of TIGAR in prolonged ischemia is an important pathway to reduce neuronal ferroptosis and provide sustainable antioxidant defense for the brain under prolonged ischemia, further complementing the mechanism of TIGAR resistance to oxidative stress induced by IS.
Collapse
Affiliation(s)
- Xinxin Wang
- Department of Brain Research, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Mei Li
- Department of Brain Research, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China
| | - Fan Wang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Guanghui Mao
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Junchao Wu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Rong Han
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China
| | - Zhenghong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, 215123, China; Institute of Heath Technology, Global Institute of Software Technology, Qingshan Road, Suzhou Science & Technology Tower, Hi-Tech Area, Suzhou, 215163, China.
| | - Hong Ni
- Department of Brain Research, Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China.
| |
Collapse
|
15
|
Gong L, Liang J, Xie L, Zhang Z, Mei Z, Zhang W. Metabolic Reprogramming in Gliocyte Post-cerebral Ischemia/ Reperfusion: From Pathophysiology to Therapeutic Potential. Curr Neuropharmacol 2024; 22:1672-1696. [PMID: 38362904 PMCID: PMC11284719 DOI: 10.2174/1570159x22666240131121032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 12/08/2023] [Accepted: 12/13/2023] [Indexed: 02/17/2024] Open
Abstract
Ischemic stroke is a leading cause of disability and death worldwide. However, the clinical efficacy of recanalization therapy as a preferred option is significantly hindered by reperfusion injury. The transformation between different phenotypes of gliocytes is closely associated with cerebral ischemia/ reperfusion injury (CI/RI). Moreover, gliocyte polarization induces metabolic reprogramming, which refers to the shift in gliocyte phenotype and the overall transformation of the metabolic network to compensate for energy demand and building block requirements during CI/RI caused by hypoxia, energy deficiency, and oxidative stress. Within microglia, the pro-inflammatory phenotype exhibits upregulated glycolysis, pentose phosphate pathway, fatty acid synthesis, and glutamine synthesis, whereas the anti-inflammatory phenotype demonstrates enhanced mitochondrial oxidative phosphorylation and fatty acid oxidation. Reactive astrocytes display increased glycolysis but impaired glycogenolysis and reduced glutamate uptake after CI/RI. There is mounting evidence suggesting that manipulation of energy metabolism homeostasis can induce microglial cells and astrocytes to switch from neurotoxic to neuroprotective phenotypes. A comprehensive understanding of underlying mechanisms and manipulation strategies targeting metabolic pathways could potentially enable gliocytes to be reprogrammed toward beneficial functions while opening new therapeutic avenues for CI/RI treatment. This review provides an overview of current insights into metabolic reprogramming mechanisms in microglia and astrocytes within the pathophysiological context of CI/RI, along with potential pharmacological targets. Herein, we emphasize the potential of metabolic reprogramming of gliocytes as a therapeutic target for CI/RI and aim to offer a novel perspective in the treatment of CI/RI.
Collapse
Affiliation(s)
- Lipeng Gong
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Junjie Liang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Letian Xie
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Zhanwei Zhang
- Department of Neurosurgery, First Affiliated Hospital of Hunan University of Traditional Chinese Medicine, Changsha, Hunan 410007, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, College of Integrated Traditional Chinese Medicine and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
- Third-Grade Pharmacological Laboratory on Chinese Medicine Approved by State Administration of Traditional Chinese Medicine, College of Medicine and Health Sciences, China Three Gorges University, Yichang, Hubei 443002, China
| | - Wenli Zhang
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| |
Collapse
|
16
|
Lin W, Zhao XY, Cheng JW, Li LT, Jiang Q, Zhang YX, Han F. Signaling pathways in brain ischemia: Mechanisms and therapeutic implications. Pharmacol Ther 2023; 251:108541. [PMID: 37783348 DOI: 10.1016/j.pharmthera.2023.108541] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Ischemic stroke occurs when the arteries supplying blood to the brain are narrowed or blocked, inducing damage to brain tissue due to a lack of blood supply. One effective way to reduce brain damage and alleviate symptoms is to reopen blocked blood vessels in a timely manner and reduce neuronal damage. To achieve this, researchers have focused on identifying key cellular signaling pathways that can be targeted with drugs. These pathways include oxidative/nitrosative stress, excitatory amino acids and their receptors, inflammatory signaling molecules, metabolic pathways, ion channels, and other molecular events involved in stroke pathology. However, evidence suggests that solely focusing on protecting neurons may not yield satisfactory clinical results. Instead, researchers should consider the multifactorial and complex mechanisms underlying stroke pathology, including the interactions between different components of the neurovascular unit. Such an approach is more representative of the actual pathological process observed in clinical settings. This review summarizes recent research on the multiple molecular mechanisms and drug targets in ischemic stroke, as well as recent advances in novel therapeutic strategies. Finally, we discuss the challenges and future prospects of new strategies based on the biological characteristics of stroke.
Collapse
Affiliation(s)
- Wen Lin
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiang-Yu Zhao
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jia-Wen Cheng
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Li-Tao Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Quan Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yi-Xuan Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China.
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China; Institute of Brain Science, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
17
|
Neves D, Salazar IL, Almeida RD, Silva RM. Molecular mechanisms of ischemia and glutamate excitotoxicity. Life Sci 2023; 328:121814. [PMID: 37236602 DOI: 10.1016/j.lfs.2023.121814] [Citation(s) in RCA: 55] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 05/05/2023] [Accepted: 05/23/2023] [Indexed: 05/28/2023]
Abstract
Excitotoxicity is classically defined as the neuronal damage caused by the excessive release of glutamate, and subsequent activation of excitatory plasma membrane receptors. In the mammalian brain, this phenomenon is mainly driven by excessive activation of glutamate receptors (GRs). Excitotoxicity is common to several chronic disorders of the Central Nervous System (CNS) and is considered the primary mechanism of neuronal loss of function and cell death in acute CNS diseases (e.g. ischemic stroke). Multiple mechanisms and pathways lead to excitotoxic cell damage including pro-death signaling cascade events downstream of glutamate receptors, calcium (Ca2+) overload, oxidative stress, mitochondrial impairment, excessive glutamate in the synaptic cleft as well as altered energy metabolism. Here, we review the current knowledge on the molecular mechanisms that underlie excitotoxicity, emphasizing the role of Nicotinamide Adenine Dinucleotide (NAD) metabolism. We also discuss novel and promising therapeutic strategies to treat excitotoxicity, highlighting recent clinical trials. Finally, we will shed light on the ongoing search for stroke biomarkers, an exciting and promising field of research, which may improve stroke diagnosis, prognosis and allow better treatment options.
Collapse
Affiliation(s)
- Diogo Neves
- iBiMED - Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal
| | - Ivan L Salazar
- Multidisciplinary Institute of Ageing, MIA - Portugal, University of Coimbra, Coimbra, Portugal
| | - Ramiro D Almeida
- iBiMED - Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal; CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
| | - Raquel M Silva
- iBiMED - Institute of Biomedicine, Department of Medical Sciences, University of Aveiro, Aveiro, Portugal; Universidade Católica Portuguesa, Faculdade de Medicina Dentária, Centro de Investigação Interdisciplinar em Saúde, Viseu, Portugal.
| |
Collapse
|
18
|
Yapindi L, Bowley T, Kurtaneck N, Bergeson RL, James K, Wilbourne J, Harrod CK, Hernandez BY, Emerling BM, Yates C, Harrod R. Activation of p53-regulated pro-survival signals and hypoxia-independent mitochondrial targeting of TIGAR by human papillomavirus E6 oncoproteins. Virology 2023; 585:1-20. [PMID: 37257253 PMCID: PMC10527176 DOI: 10.1016/j.virol.2023.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 05/02/2023] [Accepted: 05/11/2023] [Indexed: 06/02/2023]
Abstract
The high-risk subtype human papillomaviruses (hrHPVs) infect and oncogenically transform basal epidermal stem cells associated with the development of squamous-cell epithelial cancers. The viral E6 oncoprotein destabilizes the p53 tumor suppressor, inhibits p53 K120-acetylation by the Tat-interacting protein of 60 kDa (TIP60, or Kat5), and prevents p53-dependent apoptosis. Intriguingly, the p53 gene is infrequently mutated in HPV + cervical cancer clinical isolates which suggests a possible paradoxical role for this gatekeeper in viral carcinogenesis. Here, we demonstrate that E6 activates the TP53-induced glycolysis and apoptosis regulator (TIGAR) and protects cells against oncogene-induced oxidative genotoxicity. The E6 oncoprotein induces a Warburg-like stress response and activates PI3K/PI5P4K/AKT-signaling that phosphorylates the TIGAR on serine residues and induces its hypoxia-independent mitochondrial targeting in hrHPV-transformed cells. Primary HPV + cervical cancer tissues contain high levels of TIGAR, p53, and c-Myc and our xenograft studies have further shown that lentiviral-siRNA-knockdown of TIGAR expression inhibits hrHPV-induced tumorigenesis in vivo. These findings suggest the modulation of p53 pro-survival signals and the antioxidant functions of TIGAR could have key ancillary roles during HPV carcinogenesis.
Collapse
Affiliation(s)
- Lacin Yapindi
- Laboratory of Molecular Virology, Department of Biological Sciences and the Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX, 75275-0376, United States
| | - Tetiana Bowley
- Laboratory of Molecular Virology, Department of Biological Sciences and the Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX, 75275-0376, United States
| | - Nick Kurtaneck
- Laboratory of Molecular Virology, Department of Biological Sciences and the Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX, 75275-0376, United States
| | - Rachel L Bergeson
- Laboratory of Molecular Virology, Department of Biological Sciences and the Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX, 75275-0376, United States
| | - Kylie James
- Laboratory of Molecular Virology, Department of Biological Sciences and the Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX, 75275-0376, United States
| | - Jillian Wilbourne
- Laboratory of Molecular Virology, Department of Biological Sciences and the Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX, 75275-0376, United States
| | - Carolyn K Harrod
- Laboratory of Molecular Virology, Department of Biological Sciences and the Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX, 75275-0376, United States
| | - Brenda Y Hernandez
- Hawaii Tumor Registry, University of Hawaii Cancer Center, Honolulu, HI, 96813, United States
| | | | - Courtney Yates
- Laboratory Animal Resource Center, Southern Methodist University, Dallas, TX, 75275, United States
| | - Robert Harrod
- Laboratory of Molecular Virology, Department of Biological Sciences and the Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX, 75275-0376, United States.
| |
Collapse
|
19
|
Pan M, Li X, Xu G, Tian X, Li Y, Fang W. Tripartite Motif Protein Family in Central Nervous System Diseases. Cell Mol Neurobiol 2023; 43:2567-2589. [PMID: 36988770 PMCID: PMC11410135 DOI: 10.1007/s10571-023-01337-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023]
Abstract
Tripartite motif (TRIM) protein superfamily is a group of E3 ubiquitin ligases characterized by the conserved RING domain, the B-box domain, and the coiled-coil domain (RBCC). It is widely involved in various physiological and pathological processes, such as intracellular signal transduction, cell cycle regulation, oncogenesis, and innate immune response. Central nervous system (CNS) diseases are composed of encephalopathy and spinal cord diseases, which have a high disability and mortality rate. Patients are often unable to take care of themselves and their life quality can be seriously declined. Initially, the function research of TRIM proteins mainly focused on cancer. However, in recent years, accumulating attention is paid to the roles they play in CNS diseases. In this review, we integrate the reported roles of TRIM proteins in the pathological process of CNS diseases and related signaling pathways, hoping to provide theoretical bases for further research in treating CNS diseases targeting TRIM proteins. TRIM proteins participated in CNS diseases. TRIM protein family is characterized by a highly conserved RBCC domain, referring to the RING domain, the B-box domain, and the coiled-coil domain. Recent research has discovered the relations between TRIM proteins and various CNS diseases, especially Alzheimer's disease, Parkinson's disease, and ischemic stroke.
Collapse
Affiliation(s)
- Mengtian Pan
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Xiang Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Guangchen Xu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Xinjuan Tian
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Yunman Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China.
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China.
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China.
| |
Collapse
|
20
|
Li J, Wu J, Zhou X, Lu Y, Ge Y, Zhang X. Targeting neuronal mitophagy in ischemic stroke: an update. BURNS & TRAUMA 2023; 11:tkad018. [PMID: 37274155 PMCID: PMC10232375 DOI: 10.1093/burnst/tkad018] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 01/29/2023] [Accepted: 03/19/2023] [Indexed: 06/06/2023]
Abstract
Cerebral ischemia is a neurological disorder associated with complex pathological mechanisms, including autophagic degradation of neuronal mitochondria, or termed mitophagy, following ischemic events. Despite being well-documented, the cellular and molecular mechanisms underlying the regulation of neuronal mitophagy remain unknown. So far, the evidence suggests neuronal autophagy and mitophagy are separately regulated in ischemic neurons, the latter being more likely activated by reperfusional injury. Specifically, given the polarized morphology of neurons, mitophagy is regulated by different neuronal compartments, with axonal mitochondria being degraded by autophagy in the cell body following ischemia-reperfusion insult. A variety of molecules have been associated with neuronal adaptation to ischemia, including PTEN-induced kinase 1, Parkin, BCL2 and adenovirus E1B 19-kDa-interacting protein 3 (Bnip3), Bnip3-like (Bnip3l) and FUN14 domain-containing 1. Moreover, it is still controversial whether mitophagy protects against or instead aggravates ischemic brain injury. Here, we review recent studies on this topic and provide an updated overview of the role and regulation of mitophagy during ischemic events.
Collapse
Affiliation(s)
- Jun Li
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Qingchun Road 79, Xiacheng District, Hangzhou, China
| | - Jiaying Wu
- Department of Clinical Pharmacy, the First Affiliated Hospital, Zhejiang University School of Medicine, Qingchun Road 79, Xiacheng District, Hangzhou, China
| | - Xinyu Zhou
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Yuhangtang Road 866, Xihu District, Hangzhou, China
| | - Yangyang Lu
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Yuhangtang Road 866, Xihu District, Hangzhou, China
| | - Yuyang Ge
- Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Yuhangtang Road 866, Xihu District, Hangzhou, China
| | | |
Collapse
|
21
|
Liu G, Qi H, Shen J. JMJD5 inhibits lung cancer progression by regulating glucose metabolism through the p53/TIGAR pathway. Med Oncol 2023; 40:145. [PMID: 37043051 DOI: 10.1007/s12032-023-02016-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 03/30/2023] [Indexed: 04/13/2023]
Abstract
Metabolic reprogramming is considered one of the main driving forces for tumor progression, providing energy and substrates of biosynthesis to support rapid neoplastic proliferation. Particularly, the tumor suppressor protein p53 was shown to revert the Warburg effect and play complex roles in regulating glucose metabolism. Jumonji C domain-containing protein 5 (JMJD5) has previously been reported as a negative regulator of p53. However, the role of JMJD5 in p53-mediated metabolic reprogramming remains elusive. Here, we discovered that knockdown of JMJD5 significantly enhances TIGAR expression in p53 wild-type non-small cell lung cancer (NSCLC) cells, which could further suppress glycolysis and promote the pentose phosphate pathway. Besides, JMJD5 knockdown promotes the NSCLC cell proliferation in vitro and xenograft tumor growth in vivo, while silencing TIGAR can abolish this effect. Low JMJD5 expression levels are associated with elevated TIGAR levels and correlates with poor prognosis in lung cancer patients. Taken together, our findings suggest that JMJD5 is a key regulator of tumor glucose metabolism by targeting the p53/TIGAR metabolic pathway.
Collapse
Affiliation(s)
- Guiling Liu
- Department of Pathology and Pathophysiology, and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Hongyan Qi
- Department of Pathology and Pathophysiology, and Department of Radiation Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
| | - Jing Shen
- Department of Pathology and Pathophysiology, and Department of Medical Oncology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
| |
Collapse
|
22
|
Zhang Q, Yao Z, Chen F, Wang X, Wang M, Lu J, Meng Y, Xu L, Han Y, Liu W, Wang H. TIGAR Protects Cochlear Hair Cells against Teicoplanin-Induced Damage. Mol Neurobiol 2023; 60:3788-3802. [PMID: 36943624 PMCID: PMC10029784 DOI: 10.1007/s12035-023-03309-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 03/05/2023] [Indexed: 03/23/2023]
Abstract
Teicoplanin is a glycopeptide antibiotic used to treat severe staphylococcal infections. It has been claimed that teicoplanin possesses ototoxic potential, although its toxic effects on cochlear hair cells (HCs) remain unknown. The TP53-induced glycolysis and apoptosis regulator (TIGAR) plays a crucial role in promoting cell survival. Prior research has demonstrated that TIGAR protects spiral ganglion neurons against cisplatin damage. However, the significance of TIGAR in damage to mammalian HCs has not yet been investigated. In this study, firstly, we discovered that teicoplanin caused dose-dependent cell death in vitro in both HEI-OC1 cells and cochlear HCs. Next, we discovered that HCs and HEI-OC1 cells treated with teicoplanin exhibited a dramatically decrease in TIGAR expression. To investigate the involvement of TIGAR in inner ear injury caused by teicoplanin, the expression of TIGAR was either upregulated via recombinant adenovirus or downregulated by shRNA in HEI-OC1 cells. Overexpression of TIGAR increased cell viability, decreased apoptosis, and decreased intracellular reactive oxygen species (ROS) level, whereas downregulation of TIGAR decreased cell viability, exacerbated apoptosis, and elevated ROS level following teicoplanin injury. Finally, antioxidant therapy with N-acetyl-L-cysteine decreased ROS level, prevented cell death, and restored p38/phosphorylation-p38 expression levels in HEI-OC1 cells injured by teicoplanin. This study demonstrates that TIGAR may be a promising novel target for the prevention of teicoplanin-induced ototoxicity.
Collapse
Affiliation(s)
- Qiongmin Zhang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China
- Shandong Institute of Otorhinolaryngology, Jinan, Shandong, China
| | - Zhiqun Yao
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China
- Shandong Institute of Otorhinolaryngology, Jinan, Shandong, China
| | - Fang Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China
- Shandong Institute of Otorhinolaryngology, Jinan, Shandong, China
| | - Xue Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China
- Shandong Institute of Otorhinolaryngology, Jinan, Shandong, China
| | - Man Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China
- Shandong Institute of Otorhinolaryngology, Jinan, Shandong, China
| | - Junze Lu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China
- Shandong Institute of Otorhinolaryngology, Jinan, Shandong, China
| | - Yu Meng
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China
- Shandong Institute of Otorhinolaryngology, Jinan, Shandong, China
| | - Lei Xu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China
- Shandong Institute of Otorhinolaryngology, Jinan, Shandong, China
| | - Yuechen Han
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China
- Shandong Institute of Otorhinolaryngology, Jinan, Shandong, China
| | - Wenwen Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China.
- Shandong Institute of Otorhinolaryngology, Jinan, Shandong, China.
| | - Haibo Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, Shandong, China.
- Shandong Institute of Otorhinolaryngology, Jinan, Shandong, China.
| |
Collapse
|
23
|
Huang B, Lang X, Li X. The role of TIGAR in nervous system diseases. Front Aging Neurosci 2022; 14:1023161. [DOI: 10.3389/fnagi.2022.1023161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Accepted: 10/05/2022] [Indexed: 11/10/2022] Open
Abstract
TP53-induced glycolysis and apoptosis regulator (TIGAR) mainly regulates pentose phosphate pathway by inhibiting glycolysis, so as to synthesize ribose required by DNA, promote DNA damage repair and cell proliferation, maintain cell homeostasis and avoid body injury. Its physiological functions include anti-oxidative stress, reducing inflammation, maintaining mitochondrial function, inhibiting apoptosis, reducing autophagy etc. This paper reviews the research of TIGAR in neurological diseases, including stroke, Parkinson’s disease (PD), Alzheimer’s disease (AD), seizures and brain tumors, aiming to provide reference for the development of new therapeutic targets.
Collapse
|
24
|
Chai X, Pan M, Wang J, Feng M, Wang Y, Zhang Q, Sun Y. Cordycepin exhibits anti-fatigue effect via activating TIGAR/SIRT1/PGC-1α signaling pathway. Biochem Biophys Res Commun 2022; 637:127-135. [DOI: 10.1016/j.bbrc.2022.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 10/28/2022] [Accepted: 11/06/2022] [Indexed: 11/11/2022]
|
25
|
Liu M, Zhou X, Li Y, Ma S, Pan L, Zhang X, Zheng W, Wu Z, Wang K, Ahsan A, Wu J, Jiang L, Lu Y, Hu W, Qin Z, Chen Z, Zhang X. TIGAR alleviates oxidative stress in brain with extended ischemia via a pentose phosphate pathway-independent manner. Redox Biol 2022; 53:102323. [PMID: 35576689 PMCID: PMC9118922 DOI: 10.1016/j.redox.2022.102323] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/24/2022] [Accepted: 04/24/2022] [Indexed: 01/18/2023] Open
Abstract
TP53-induced glycolysis and apoptosis regulator (TIGAR) alleviates oxidative stress and protects against ischemic neuronal injury by shifting glucose metabolism into the pentose phosphate pathway (PPP). However, the brain alters glucose metabolism from PPP to glycolysis during prolonged ischemia. It is still unknown whether and how TIGAR exerts the antioxidant activity and neuroprotection in prolonged ischemic brains. Here, we determined the significant upregulation of TIGAR that was proportional to the duration of ischemia. However, TIGAR failed to upregulate the NADPH level but still alleviated oxidative stress in neuronal cells with prolonged oxygen glucose-deprivation (OGD). Furthermore, inhibiting PPP activity, either by the expression of mutant TIGAR (which lacks enzymatic activity) or by silencing Glucose 6-phosphate dehydrogenase, still retained antioxidant effects and neuroprotection of TIGAR with prolonged OGD. Intriguingly, TIGAR-induced autophagy alleviated oxidative stress, contributing to neuron survival. Further experiments indicated that TIGAR-induced autophagy neutralized oxidative stress by activating Nrf2, which was cancelled by ML385 or Nrf2 knockdown. Remarkably, either Atg7 deletion or Nrf2 silencing abolished the neuroprotection of TIGAR in mice with prolonged ischemia. Taken together, we found a PPP-independent pathway in which TIGAR alleviates oxidative stress. TIGAR induces autophagy and, thus, activates Nrf2, offering sustainable antioxidant defense in brains with extended ischemia. This previously unexplored mechanism of TIGAR may serve as a critical compensation for antioxidant activity caused by the lack of glucose in ischemic stroke. We identified a PPP-independent mechanism of TIGAR to neutralize ROS in neurons with extended ischemia. In neuronal cells with prolonged ischemia, TIGAR-induced autophagy alleviated oxidative stress. TIGAR-induced autophagy activated Nrf2, which compensated for the poor NADPH generation with prolonged ischemia.
Collapse
|
26
|
The inhibition of TRIM35-mediated TIGAR ubiquitination enhances mitochondrial fusion and alleviates renal ischemia-reperfusion injury. Int J Biol Macromol 2022; 209:725-736. [PMID: 35421414 DOI: 10.1016/j.ijbiomac.2022.04.054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 11/23/2022]
Abstract
Tripartite motif 35 (TRIM35) is a member of the tripartite motif protein family and has been recognized to play a key role in immune-inflammatory diseases. However, the role of TRIM35 in renal ischemia-reperfusion injury (IRI) remains unclear. Our study proved that knockdown of TRIM35 alleviates kidney IRI by inhibiting oxidative stress and enhancing mitochondrial fusion. In addition, our experimental results found that TRIM35 interacts with TP53-induced glycolysis and apoptosis regulator (TIGAR) and promotes the polyubiquitination of TIGAR and induces its degradation in the proteasome pathway. Furthermore, TIGAR knockdown significantly inhibited mitochondrial fusion. These results indicate that TRIM35 is a potential therapeutic target for renal IRI.
Collapse
|
27
|
Kanan Y, Hackett SF, Taneja K, Khan M, Campochiaro PA. Oxidative stress-induced alterations in retinal glucose metabolism in Retinitis Pigmentosa. Free Radic Biol Med 2022; 181:143-153. [PMID: 35134532 PMCID: PMC8891093 DOI: 10.1016/j.freeradbiomed.2022.01.032] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/21/2022] [Accepted: 01/31/2022] [Indexed: 10/19/2022]
Abstract
Retinitis pigmentosa occurs due to mutations that cause rod photoreceptor degeneration. Once most rods are lost, gradual degeneration of cone photoreceptors occurs. Oxidative damage and abnormal glucose metabolism have been implicated as contributors to cone photoreceptor death. Herein, we show increased phosphorylation of key enzymes of glucose metabolism in the retinas of rd10 mice, a model of RP, and retinas of wild type mice with paraquat-induced oxidative stress, thereby inhibiting these key enzymes. Dietary supplementation with glucose and pyruvate failed to overcome the inhibition, but increased reducing equivalents in the retina and improved cone function and survival. Dichloroacetate reversed the increased phosphorylation of pyruvate dehydrogenase in rd10 retina and increased histone acetylation and levels of TP53-induced glycolysis and apoptosis regulator (TIGAR), which redirected glucose metabolism toward the pentose phosphate pathway. These data indicate that oxidative stress induced damage can be reversed by shifting glycolytic intermediates toward the pentose phosphate pathway which increases reducing equivalents and provides photoreceptor protection.
Collapse
Affiliation(s)
- Yogita Kanan
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sean F Hackett
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Kamil Taneja
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Mahmood Khan
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Peter A Campochiaro
- The Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
28
|
Lespay-Rebolledo C, Tapia-Bustos A, Perez-Lobos R, Vio V, Casanova-Ortiz E, Farfan-Troncoso N, Zamorano-Cataldo M, Redel-Villarroel M, Ezquer F, Quintanilla ME, Israel Y, Morales P, Herrera-Marschitz M. Sustained Energy Deficit Following Perinatal Asphyxia: A Shift towards the Fructose-2,6-bisphosphatase (TIGAR)-Dependent Pentose Phosphate Pathway and Postnatal Development. Antioxidants (Basel) 2021; 11:74. [PMID: 35052577 PMCID: PMC8773255 DOI: 10.3390/antiox11010074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/16/2022] Open
Abstract
Labor and delivery entail a complex and sequential metabolic and physiologic cascade, culminating in most circumstances in successful childbirth, although delivery can be a risky episode if oxygen supply is interrupted, resulting in perinatal asphyxia (PA). PA causes an energy failure, leading to cell dysfunction and death if re-oxygenation is not promptly restored. PA is associated with long-term effects, challenging the ability of the brain to cope with stressors occurring along with life. We review here relevant targets responsible for metabolic cascades linked to neurodevelopmental impairments, that we have identified with a model of global PA in rats. Severe PA induces a sustained effect on redox homeostasis, increasing oxidative stress, decreasing metabolic and tissue antioxidant capacity in vulnerable brain regions, which remains weeks after the insult. Catalase activity is decreased in mesencephalon and hippocampus from PA-exposed (AS), compared to control neonates (CS), in parallel with increased cleaved caspase-3 levels, associated with decreased glutathione reductase and glutathione peroxidase activity, a shift towards the TIGAR-dependent pentose phosphate pathway, and delayed calpain-dependent cell death. The brain damage continues long after the re-oxygenation period, extending for weeks after PA, affecting neurons and glial cells, including myelination in grey and white matter. The resulting vulnerability was investigated with organotypic cultures built from AS and CS rat newborns, showing that substantia nigra TH-dopamine-positive cells from AS were more vulnerable to 1 mM of H2O2 than those from CS animals. Several therapeutic strategies are discussed, including hypothermia; N-acetylcysteine; memantine; nicotinamide, and intranasally administered mesenchymal stem cell secretomes, promising clinical translation.
Collapse
Affiliation(s)
- Carolyne Lespay-Rebolledo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Andrea Tapia-Bustos
- School of Pharmacy, Faculty of Medicine, Universidad Andres Bello, Santiago 8370149, Chile;
| | - Ronald Perez-Lobos
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Valentina Vio
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Emmanuel Casanova-Ortiz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Nancy Farfan-Troncoso
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Marta Zamorano-Cataldo
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Martina Redel-Villarroel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Fernando Ezquer
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Maria Elena Quintanilla
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| | - Yedy Israel
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Center for Regenerative Medicine, Faculty of Medicine-Clínica Alemana, Universidad del Desarrollo, Santiago 7710162, Chile;
| | - Paola Morales
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
- Department of Neuroscience, Faculty of Medicine, University of Chile, Santiago 8380453, Chile
| | - Mario Herrera-Marschitz
- Molecular & Clinical Pharmacology Program, ICBM, Faculty of Medicine, University of Chile, Santiago 8380453, Chile; (C.L.-R.); (R.P.-L.); (V.V.); (E.C.-O.); (N.F.-T.); (M.Z.-C.); (M.R.-V.); (M.E.Q.); (Y.I.)
| |
Collapse
|
29
|
Huang SS, Sheng YC, Jiang YY, Liu N, Lin MM, Wu JC, Liang ZQ, Qin ZH, Wang Y. TIGAR plays neuroprotective roles in KA-induced excitotoxicity through reducing neuroinflammation and improving mitochondrial function. Neurochem Int 2021; 152:105244. [PMID: 34826530 DOI: 10.1016/j.neuint.2021.105244] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 10/30/2021] [Accepted: 11/19/2021] [Indexed: 02/05/2023]
Abstract
Excitotoxicity refers to the ability of excessive extracellular excitatory amino acids to damage neurons via receptor activation. It is a crucial pathogenetic process in neurodegenerative diseases. TP53 is confirmed to be involved in excitotoxicity. It is demonstrated that TP53 induced glycolysis and apoptotic regulator (TIGAR)-regulated metabolic pathway can protect against neuronal injury. However, the role of TIGAR in excitotoxicity and specific mechanisms is still unknown. In this study, an in vivo excitotoxicity model was constructed via stereotypical kainic acid (KA) injection into the striatum of mice. KA reduced TIGAR expression levels, neuroinflammatory responses and mitochondrial dysfunction. TIGAR overexpression could reverse KA-induced neuronal injury by reducing neuroinflammation and improving mitochondrial function, thereby exerting neuroprotective effects. Therefore, this study could provide a potential therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Si-Si Huang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yi-Chao Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yi-Yue Jiang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Na Liu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Miao-Miao Lin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jun-Chao Wu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zhong-Qin Liang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yan Wang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
30
|
Structure, regulation, and biological functions of TIGAR and its role in diseases. Acta Pharmacol Sin 2021; 42:1547-1555. [PMID: 33510458 PMCID: PMC8463536 DOI: 10.1038/s41401-020-00588-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 11/22/2020] [Indexed: 02/02/2023]
Abstract
TIGAR (TP53-induced glycolysis and apoptosis regulator) is the downstream target gene of p53, contains a functional sequence similar to 6-phosphofructose kinase/fructose-2, 6-bisphosphatase (PFKFB) bisphosphatase domain. TIGAR is mainly located in the cytoplasm; in response to stress, TIGAR is translocated to nucleus and organelles, including mitochondria and endoplasmic reticulum to regulate cell function. P53 family members (p53, p63, and p73), some transcription factors (SP1 and CREB), and noncoding miRNAs (miR-144, miR-885-5p, and miR-101) regulate the transcription of TIGAR. TIGAR mainly functions as fructose-2,6-bisphosphatase to hydrolyze fructose-1,6-diphosphate and fructose-2,6-diphosphate to inhibit glycolysis. TIGAR in turn facilitates pentose phosphate pathway flux to produce nicotinamide adenine dinucleotide phosphate (NADPH) and ribose, thereby promoting DNA repair, and reducing intracellular reactive oxygen species. TIGAR thus maintains energy metabolism balance, regulates autophagy and stem cell differentiation, and promotes cell survival. Meanwhile, TIGAR also has a nonenzymatic function and can interact with retinoblastoma protein, protein kinase B, nuclear factor-kappa B, hexokinase 2, and ATP5A1 to mediate cell cycle arrest, inflammatory response, and mitochondrial protection. TIGAR might be a potential target for the prevention and treatment of cardiovascular and neurological diseases, as well as cancers.
Collapse
|
31
|
Zoccarato A, Nabeebaccus AA, Oexner RR, Santos CXC, Shah AM. The nexus between redox state and intermediary metabolism. FEBS J 2021; 289:5440-5462. [PMID: 34496138 DOI: 10.1111/febs.16191] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/20/2021] [Accepted: 09/07/2021] [Indexed: 12/12/2022]
Abstract
Reactive oxygen species (ROS) are not just a by-product of cellular metabolic processes but act as signalling molecules that regulate both physiological and pathophysiological processes. A close connection exists in cells between redox homeostasis and cellular metabolism. In this review, we describe how intracellular redox state and glycolytic intermediary metabolism are closely coupled. On the one hand, ROS signalling can control glycolytic intermediary metabolism by direct regulation of the activity of key metabolic enzymes and indirect regulation via redox-sensitive transcription factors. On the other hand, metabolic adaptation and reprogramming in response to physiological or pathological stimuli regulate intracellular redox balance, through mechanisms such as the generation of reducing equivalents. We also discuss the impact of these intermediary metabolism-redox circuits in physiological and disease settings across different tissues. A better understanding of the mechanisms regulating these intermediary metabolism-redox circuits will be crucial to the development of novel therapeutic strategies.
Collapse
Affiliation(s)
- Anna Zoccarato
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Adam A Nabeebaccus
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Rafael R Oexner
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Celio X C Santos
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, King's College London British Heart Foundation Centre of Excellence, London, UK
| |
Collapse
|
32
|
Zeng S, Zhao Z, Zheng S, Wu M, Song X, Li Y, Zheng Y, Liu B, Chen L, Gao C, Liu H. The E3 ubiquitin ligase TRIM31 is involved in cerebral ischemic injury by promoting degradation of TIGAR. Redox Biol 2021; 45:102058. [PMID: 34218200 PMCID: PMC8260875 DOI: 10.1016/j.redox.2021.102058] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 11/19/2022] Open
Abstract
Tripartite motif (TRIM) 31 has been implicated in diverse biological and pathological conditions. However, whether TRIM31 plays a role in ischemic stroke progression is not clarified. Here we demonstrated that TRIM31 was significantly downregulated in the ischemic brain and the deficiency of TRIM31 alleviated brain injury induced by middle cerebral artery occlusion by reducing reactive oxygen species production and maintaining mitochondrial homeostasis. Mechanistically, we found that TRIM31 is an E3 ubiquitin ligase for TP53-induced glycolysis and apoptosis regulator (TIGAR), which confers protection against brain ischemia by increasing the pentose phosphate pathway flux and preserving mitochondria function. TRIM31 interacted with TIGAR and promoted the polyubiquitination of TIGAR, consequently facilitated its degradation in a proteasome-dependent pathway. Furthermore, TIGAR knockdown effectively abolished the protective effect of TRIM31 deficiency after cerebral ischemia. In conclusion, we identified that TRIM31 was a novel E3 ubiquitin ligase for TIGAR, played a critical role in regulating its protein level, and subsequently involved in the ischemic brain injury, suggesting TRIM31 as a potential therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Shenglan Zeng
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Ze Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Shengnan Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Mengting Wu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaomeng Song
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yiquan Li
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Lin Chen
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Huiqing Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
33
|
Li QQ, Li JY, Zhou M, Qin ZH, Sheng R. Targeting neuroinflammation to treat cerebral ischemia - The role of TIGAR/NADPH axis. Neurochem Int 2021; 148:105081. [PMID: 34082063 DOI: 10.1016/j.neuint.2021.105081] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/24/2021] [Accepted: 05/22/2021] [Indexed: 01/30/2023]
Abstract
Cerebral ischemia is a disease of ischemic necrosis of brain tissue caused by intracranial artery stenosis or occlusion and cerebral artery embolization. Neuroinflammation plays an important role in the pathophysiology of cerebral ischemia. Microglia, astrocytes, leukocytes and other cells that release a variety of inflammatory factors involved in neuroinflammation may play a damaging or protective role during the process of cerebral ischemia. TP53-induced glycolysis and apoptotic regulators (TIGAR) may facilitate the production of nicotinamide adenine dinucleotide phosphoric acid (NADPH) via the pentose phosphate pathway (PPP) to inhibit oxidative stress and neuroinflammation. TIGAR can also directly inhibit NF-κB to inhibit neuroinflammation. TIGAR thus protect against cerebral ischemic injury. Exogenous NADPH can inhibit neuroinflammation by inhibiting oxidative stress and regulating a variety of signals. However, since NADPH oxidase (NOX) may use NADPH as a substrate to generate reactive oxygen species (ROS) to mediate neuroinflammation, the combination of NADPH and NOX inhibitors may produce more powerful anti-neuroinflammatory effects. Here, we review the cells and regulatory signals involved in neuroinflammation during cerebral ischemia, and discuss the possible mechanisms of targeting neuroinflammation in the treatment of cerebral ischemia with TIGAR/NADPH axis, so as to provide new ideas for the prevention and treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Qi-Qi Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Jia-Ying Li
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Ming Zhou
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China.
| |
Collapse
|
34
|
Zhang S, Lachance BB, Mattson MP, Jia X. Glucose metabolic crosstalk and regulation in brain function and diseases. Prog Neurobiol 2021; 204:102089. [PMID: 34118354 DOI: 10.1016/j.pneurobio.2021.102089] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 04/08/2021] [Accepted: 06/01/2021] [Indexed: 01/11/2023]
Abstract
Brain glucose metabolism, including glycolysis, the pentose phosphate pathway, and glycogen turnover, produces ATP for energetic support and provides the precursors for the synthesis of biological macromolecules. Although glucose metabolism in neurons and astrocytes has been extensively studied, the glucose metabolism of microglia and oligodendrocytes, and their interactions with neurons and astrocytes, remain critical to understand brain function. Brain regions with heterogeneous cell composition and cell-type-specific profiles of glucose metabolism suggest that metabolic networks within the brain are complex. Signal transduction proteins including those in the Wnt, GSK-3β, PI3K-AKT, and AMPK pathways are involved in regulating these networks. Additionally, glycolytic enzymes and metabolites, such as hexokinase 2, acetyl-CoA, and enolase 2, are implicated in the modulation of cellular function, microglial activation, glycation, and acetylation of biomolecules. Given these extensive networks, glucose metabolism dysfunction in the whole brain or specific cell types is strongly associated with neurologic pathology including ischemic brain injury and neurodegenerative disorders. This review characterizes the glucose metabolism networks of the brain based on molecular signaling and cellular and regional interactions, and elucidates glucose metabolism-based mechanisms of neurological diseases and therapeutic approaches that may ameliorate metabolic abnormalities in those diseases.
Collapse
Affiliation(s)
- Shuai Zhang
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, United States
| | - Brittany Bolduc Lachance
- Program in Trauma, Department of Neurology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States
| | - Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, United States
| | - Xiaofeng Jia
- Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD, 21201, United States; Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, 21201, United States; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, 21201, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, United States; Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, United States.
| |
Collapse
|
35
|
Liu F, Li L, Chen J, Wu Y, Cao Y, Zhong P. A Network Pharmacology to Explore the Mechanism of Calculus Bovis in the Treatment of Ischemic Stroke. BIOMED RESEARCH INTERNATIONAL 2021; 2021:6611018. [PMID: 33778069 PMCID: PMC7972848 DOI: 10.1155/2021/6611018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/15/2021] [Accepted: 02/20/2021] [Indexed: 02/06/2023]
Abstract
BACKGROUND Calculus Bovis is a valuable Chinese medicine, which is widely used in the clinical treatment of ischemic stroke. The present study is aimed at investigating its target and the mechanism involved in ischemic stroke treatment by network pharmacology. METHODS Effective compounds of Calculus Bovis were collected using methods of network pharmacology and using the Bioinformatics Analysis Tool for Molecular Mechanism of Traditional Chinese Medicine (BATMAN-TCM) and the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform (TCMSP). Potential compound targets were searched in the TCMSP and SwissTargetPrediction databases. Ischemic stroke-related disease targets were searched in the Drugbank, DisGeNet, OMIM, and TTD databases. These two types of targets were uploaded to the STRING database, and a network of their interaction (PPI) was built with its characteristics calculated, aiming to reveal a number of key targets. Hub genes were selected using a plug-in of the Cytoscape software, and Gene Ontology (GO) biological processes and pathway enrichment analyses of Kyoto Encyclopedia of Genes and Genomes (KEGG) were conducted using the clusterProfiler package of R language. RESULTS Among 12 compounds, deoxycorticosterone, methyl cholate, and biliverdin were potentially effective components. A total of 344 Calculus Bovis compound targets and 590 ischemic stroke targets were found with 92 overlapping targets, including hub genes such as TP53, AKT, PIK2CA, MAPK3, MMP9, and MMP2. Biological functions of Calculus Bovis are associated with protein hydrolyzation, phosphorylation of serine/threonine residues of protein substrates, peptide bond hydrolyzation of peptides and proteins, hydrolyzation of intracellular second messengers, antioxidation and reduction, RNA transcription, and other biological processes. CONCLUSION Calculus Bovis may play a role in ischemic stroke by activating PI3K-AKT and MAPK signaling pathways, which are involved in regulating inflammatory response, cell apoptosis, and proliferation.
Collapse
Affiliation(s)
- Fangchen Liu
- Department of Neurology, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
- Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ling Li
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Jian Chen
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Ying Wu
- Department of Neurology, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Yongbing Cao
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
| | - Ping Zhong
- Department of Neurology, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai 200082, China
- Department of Neurology, Shidong Hospital of Yangpu District, Shanghai 200090, China
| |
Collapse
|
36
|
Li YA, Liu ZG, Zhang YP, Hou HT, He GW, Xue LG, Yang Q, Liu XC. Differential expression profiles of circular RNAs in the rat hippocampus after deep hypothermic circulatory arrest. Artif Organs 2021; 45:866-880. [PMID: 33432632 DOI: 10.1111/aor.13910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/16/2020] [Accepted: 01/07/2021] [Indexed: 11/28/2022]
Abstract
Neurological dysfunction commonly occurs after cardiac surgery with deep hypothermic circulatory arrest (DHCA). The mechanisms underlying DHCA-associated brain injury remain poorly understood. This study determined the changes in expression profiles of circular RNAs (circRNAs) in the hippocampus in rats that underwent DHCA, with an attempt to explore the potential role of circRNAs in the brain injury associated with DHCA. Adult male Sprague Dawley rats were subjected to cardiopulmonary bypass with DHCA. Brain injury was evaluated by neurological severity scores and histological as well as transmission electron microscope examinations. The expression profiles of circRNAs in the hippocampal tissues were screened by microarray. Quantitative real-time PCR (RT-qPCR) was used to validate the reliability of the microarray results. Bioinformatic algorithms were applied to construct a competing endogenous RNA (ceRNA) network, and Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses were performed to explore the potential biological roles of the circRNAs. Out of 14 145 circRNAs screened, 56 were differentially expressed in the hippocampus between the DHCA and sham-operated rats, including 30 upregulated and 26 downregulated circRNAs. The expression changes of six selected circRNAs (upregulated: rno_circRNA_011190, rno_circRNA_012988, rno_circRNA_000544; downregulated: rno_circRNA_010393, rno_circRNA_012043, rno_circRNA_015149) were further confirmed by RT-qPCR. Bioinformatics analysis showed the enrichment of these confirmed circRNAs and their potential target mRNAs in several KEGG pathways including histidine metabolism, adipocytokine signaling, and cAMP signaling. By revealing the change expression profiles of circRNAs in the brain after DHCA, this study indicates possible involvements of these dysregulated circRNAs in brain injury and suggests a potential of targeting circRNAs for prevention and treatment of neurological dysfunction associated with DHCA.
Collapse
Affiliation(s)
- Yi-Ai Li
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Graduate School of Peking Union Medical College, Tianjin, China
| | - Zhi-Gang Liu
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Graduate School of Peking Union Medical College, Tianjin, China
| | - You-Peng Zhang
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Graduate School of Peking Union Medical College, Tianjin, China
| | - Hai-Tao Hou
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Graduate School of Peking Union Medical College, Tianjin, China
| | - Guo-Wei He
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Graduate School of Peking Union Medical College, Tianjin, China.,Department of Cardiac Surgery, The First Affiliated Hospital, Zhejiang University, Hangzhou, China.,School of Pharmacy, Wannan Medical College, Wuhu, China.,Department of Surgery, Oregon Health and Science University, Portland, OR, USA
| | - Lan-Gang Xue
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Graduate School of Peking Union Medical College, Tianjin, China
| | - Qin Yang
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Graduate School of Peking Union Medical College, Tianjin, China
| | - Xiao-Cheng Liu
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Graduate School of Peking Union Medical College, Tianjin, China
| |
Collapse
|
37
|
Xu X, Sun M, Luo X, Zhang Z, Su L, Cui L, Zhu Z, Lu X, Wang R, Han F, Qian X, Yang Y. One-electron reduction triggered nitric oxide release for ischemia-reperfusion protection. Free Radic Biol Med 2021; 164:13-19. [PMID: 33418107 DOI: 10.1016/j.freeradbiomed.2020.12.443] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Revised: 12/23/2020] [Accepted: 12/29/2020] [Indexed: 12/25/2022]
Abstract
Nitric oxide donors (NODs) are indispensable in biological research and disease treatment. NODs had been utilized to treat cardiovascular diseases in clinic and many others are under trial. Thiols are typically required for these donors to release NO. Yet, their mechanism is complex and often lead to resistance. Herein, we reported that N-nitrosated electron-deficient dyes are capable of NO release with one-electron reduction. A fluorophore is generated simultaneously, whose fluorescence is harnessed to monitor the profile of NO release. Through electrochemical and spectral studies, NOD f3 was found to exhibit good biocompatibility and high reduction efficiency and its potentials in cell-protection in oxygen and glucose deprivation (OGD) models were showcased with endothelial cells. This work aims at offering a new approach to design reduction-triggered NOD, which have therapeutic potentials in ischemia-reperfusion.
Collapse
Affiliation(s)
- Xiu Xu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Meiling Sun
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xiao Luo
- School of Chemistry and Molecular Engineering, East China Normal University, Dongchuan Road 500, Shanghai, 200241, China
| | - Ziqian Zhang
- Guangxi Scientific Research Center of Traditional Chinese Medicine, Guangxi University of Chinese Medicine, Nanning, 530200, China
| | - Lin Su
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Lingfei Cui
- School of Chemistry and Molecular Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Zhihui Zhu
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Xicun Lu
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Rui Wang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
| | - Xuhong Qian
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Youjun Yang
- State Key Laboratory of Bioreactor Engineering, Shanghai Key Laboratory of Chemical Biology, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
| |
Collapse
|
38
|
Tan LL, Jiang XL, Xu LX, Li G, Feng CX, Ding X, Sun B, Qin ZH, Zhang ZB, Feng X, Li M. TP53-induced glycolysis and apoptosis regulator alleviates hypoxia/ischemia-induced microglial pyroptosis and ischemic brain damage. Neural Regen Res 2021; 16:1037-1043. [PMID: 33269748 PMCID: PMC8224121 DOI: 10.4103/1673-5374.300453] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Our previous studies have demonstrated that TP53-induced glycolysis and apoptosis regulator (TIGAR) can protect neurons after cerebral ischemia/reperfusion. However, the role of TIGAR in neonatal hypoxic-ischemic brain damage (HIBD) remains unknown. In the present study, 7-day-old Sprague-Dawley rat models of HIBD were established by permanent occlusion of the left common carotid artery followed by 2-hour hypoxia. At 6 days before induction of HIBD, a lentiviral vector containing short hairpin RNA of either TIGAR or gasdermin D (LV-sh_TIGAR or LV-sh_GSDMD) was injected into the left lateral ventricle and striatum. Highly aggressively proliferating immortalized (HAPI) microglial cell models of in vitro HIBD were established by 2-hour oxygen/glucose deprivation followed by 24-hour reoxygenation. Three days before in vitro HIBD induction, HAPI microglial cells were transfected with LV-sh_TIGAR or LV-sh_GSDMD. Our results showed that TIGAR expression was increased in the neonatal rat cortex after HIBD and in HAPI microglial cells after oxygen/glucose deprivation/reoxygenation. Lentivirus-mediated TIGAR knockdown in rats markedly worsened pyroptosis and brain damage after hypoxia/ischemia in vivo and in vitro. Application of exogenous nicotinamide adenine dinucleotide phosphate (NADPH) increased the NADPH level and the glutathione/oxidized glutathione ratio and decreased reactive oxygen species levels in HAPI microglial cells after oxygen/glucose deprivation/reoxygenation. Additionally, exogenous NADPH blocked the effects of TIGAR knockdown in neonatal HIBD in vivo and in vitro. These findings show that TIGAR can inhibit microglial pyroptosis and play a protective role in neonatal HIBD. The study was approved by the Animal Ethics Committee of Soochow University of China (approval No. 2017LW003) in 2017.
Collapse
Affiliation(s)
- Lan-Lan Tan
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiao-Lu Jiang
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Li-Xiao Xu
- Department of Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Gen Li
- Department of Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Chen-Xi Feng
- Department of Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xin Ding
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Bin Sun
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Zheng-Hong Qin
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu Province, China
| | - Zu-Bin Zhang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, Jiangsu Province, China
| | - Xing Feng
- Department of Neonatology, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Mei Li
- Department of Pediatrics Research Institute, Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| |
Collapse
|
39
|
TP53-induced glycolysis and apoptosis regulator (TIGAR) ameliorates lysosomal damage in the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine-mediated mouse model of Parkinson's disease. Toxicol Lett 2020; 339:60-69. [PMID: 33359019 DOI: 10.1016/j.toxlet.2020.12.011] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/03/2020] [Accepted: 12/18/2020] [Indexed: 01/13/2023]
Abstract
The progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNpc) correlates with rupture of lysosome in Parkinson's disease (PD). It has been found that TP53-induced glycolysis and apoptosis regulator (TIGAR) has been attributed to the regulation of metabolic pathways and neuroprotective effect. In the present study, we showed in a mouse model that 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) caused lysosomal damage and DA neurons loss in the SNpc. MPTP only induced SP1-mediated TIGAR upregulation in the early stage of neurotoxin-induced pathology, and this compensatory mechanism was not enough to maintain normal lysosomal function. MPTP significantly decreased the levels of NADPH and GSH, and the effects were ameliorated by the expression of exogenous TIGAR but execerbated by knockdown of TIAGR. TIGAR or NADPH alleviated oxidative stress, rescued lysosomal dysfunction and attenuated DA neurons degeneration. Overexpression of TIGAR or NADPH supplement inhibited MPP+-mediated reactive oxygen species (ROS), lysosomal membrane permeabilization (LMP) and autophagic flux impairment in PC12 cells. Together, these findings suggest that TIGAR reduces MPTP-mediated oxidative stress, lysosomal depletion and DA neuron damage.
Collapse
|
40
|
Chen L, Xia YF, Shen SF, Tang J, Chen JL, Qian K, Chen Z, Qin ZH, Sheng R. Syntaxin 17 inhibits ischemic neuronal injury by resuming autophagy flux and ameliorating endoplasmic reticulum stress. Free Radic Biol Med 2020; 160:319-333. [PMID: 32828953 DOI: 10.1016/j.freeradbiomed.2020.08.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023]
Abstract
Previous studies have shown that syntaxin 17 (STX17) is involved in mediating the fusion of autophagosomes and lysosomes. This study aimed to investigate the role and mechanism of STX17 in neuronal injury following cerebral ischemia/reperfusion. The ischemia/reperfusion (I/R) models were established by transient middle cerebral artery occlusion (tMCAO) in mice and oxygen glucose deprivation/reperfusion (O/R) in primary cultured cortical neurons and HT22 cells. Cerebral ischemia/reperfusion significantly up-regulated the expression of STX17 in neurons. Lentivirus mediated knockdown of STX17 in neurons reduced neuronal viability and increased LDH leakage. Injection of AAV9-shSTX17 into the brain of mice then subjected to tMCAO also significantly augmented the infarct area and exacerbated neurobehavioral deficits and mortality. Depletion of STX17 caused accumulation of autophagic marker/substrate LC3 II and p62, blockade of the autophagic flux, and the accumulation of dysfunctional lysosomes. Knockdown of STX17 also aggravated endoplasmic reticulum (ER) stress-dependent neuronal apoptosis induced by ischemia/reperfusion. Importantly, induction of autophagy-lysosomal pathway and alleviation of ER stress partially rescued STX17 knockdown-induced neuronal damage. These results suggest that STX17 may ameliorate ischemia/reperfusion-induced neuronal damage by enhancing autophagy flux and reducing ER stress-dependent neuronal apoptosis.
Collapse
Affiliation(s)
- Lei Chen
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Yun-Fei Xia
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Shu-Fang Shen
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Jie Tang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Jia-Li Chen
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Ke Qian
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Zhong Chen
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China
| | - Rui Sheng
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences of Soochow University, Suzhou, China.
| |
Collapse
|
41
|
Eriksson SE, Ceder S, Bykov VJN, Wiman KG. p53 as a hub in cellular redox regulation and therapeutic target in cancer. J Mol Cell Biol 2020; 11:330-341. [PMID: 30892598 PMCID: PMC6734141 DOI: 10.1093/jmcb/mjz005] [Citation(s) in RCA: 81] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 12/21/2018] [Accepted: 02/11/2019] [Indexed: 12/25/2022] Open
Abstract
The TP53 tumor suppressor gene encodes a DNA-binding transcription factor that regulates multiple cellular processes including cell growth and cell death. The ability of p53 to bind to DNA and activate transcription is tightly regulated by post-translational modifications and is dependent on a reducing cellular environment. Some p53 transcriptional target genes are involved in regulation of the cellular redox homeostasis, e.g. TIGAR and GLS2. A large fraction of human tumors carry TP53 mutations, most commonly missense mutations that lead to single amino acid substitutions in the core domain. Mutant p53 proteins can acquire so called gain-of-function activities and influence the cellular redox balance in various ways, for instance by binding of the Nrf2 transcription factor, a major regulator of cellular redox state. The DNA-binding core domain of p53 has 10 cysteine residues, three of which participate in holding a zinc atom that is critical for p53 structure and function. Several novel compounds that refold and reactivate missense mutant p53 bind to specific p53 cysteine residues. These compounds can also react with other thiols and target components of the cellular redox system, such as glutathione. Dual targeting of mutant p53 and redox homeostasis may allow more efficient treatment of cancer.
Collapse
Affiliation(s)
- Sofi E Eriksson
- Karolinska Institutet, Department of Oncology-Pathology, BioClinicum, Stockholm, Sweden
| | - Sophia Ceder
- Karolinska Institutet, Department of Oncology-Pathology, BioClinicum, Stockholm, Sweden
| | - Vladimir J N Bykov
- Karolinska Institutet, Department of Oncology-Pathology, BioClinicum, Stockholm, Sweden
| | - Klas G Wiman
- Karolinska Institutet, Department of Oncology-Pathology, BioClinicum, Stockholm, Sweden
| |
Collapse
|
42
|
Liu ZQ, Liu N, Huang SS, Lin MM, Qin S, Wu JC, Liang ZQ, Qin ZH, Wang Y. NADPH protects against kainic acid-induced excitotoxicity via autophagy-lysosome pathway in rat striatum and primary cortical neurons. Toxicology 2020; 435:152408. [PMID: 32057834 DOI: 10.1016/j.tox.2020.152408] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/24/2020] [Accepted: 02/10/2020] [Indexed: 12/20/2022]
Abstract
PURPOSE To investigate the effects and mechanisms of NADPH on Kainic acid (KA)-induced excitotoxicity. METHODS KA, a non-N-methyl-d-aspartate glutamate receptor agonist, was exposed to adult SD rats via intrastriatal injection and rat primary cortical neurons to establish excitotoxic models in vivo and in vitro, respectively. To determine the effects of NADPH on KA-induced excitotoxicity, neuronal survival, neurologically behavioral score and oxidative stress were evaluated. To explore the mechanisms of neuroprotective effects of NADPH, the autophagy-lysosome pathway related proteins were detected. RESULTS In vivo, NADPH (1 mg/kg or 2 mg/kg) diminished KA (2.5 nmol)-induced enlargement of lesion size in striatum, improved KA-induced dyskinesia and reversed KA-induced activation of glial cells. Nevertheless, the neuroprotective effect of NADPH was not significant under the condition of autophagy activation. NADPH (2 mg/kg) inhibited KA (2.5 nmol)-induced down-regulation of TP-53 induced glycolysis and apoptosis regulator (TIGAR) and p62, and up-regulation of the protein levels of LC3-II/LC3-I, Beclin-1 and Atg5. In vitro, the excitotoxic neuronal injury was induced after KA (50 μM, 100 μM or 200 μM) treatment as demonstrated by decreased cell viability. Moreover, KA (100 μM) increased the intracellular levels of calcium and reactive oxygen species (ROS) and declined the levels of the reduced form of glutathione (GSH). Pretreatment of NADPH (10 μM) effectively reversed these changes. Meanwhile NADPH (10 μM) inhibited KA (100 μM)-induced down-regulation of TIGAR and p62, and up-regulation of the ratio of LC3-II/LC3-I, Beclin-1, Atg5, active-cathepsin B and active-cathepsin D. CONCLUSIONS Our data provide a possible mechanism that NADPH ameliorates KA-induced excitotoxicity by blocking the autophagy-lysosome pathway and up-regulating TIGAR along with its antioxidant properties.
Collapse
Affiliation(s)
- Zi-Qi Liu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Na Liu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Si-Si Huang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Miao-Miao Lin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Shu Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Jun-Chao Wu
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zhong-Qin Liang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Zheng-Hong Qin
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yan Wang
- Department of Pharmacology and Laboratory of Aging and Nervous Diseases and Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
43
|
Cheung EC, DeNicola GM, Nixon C, Blyth K, Labuschagne CF, Tuveson DA, Vousden KH. Dynamic ROS Control by TIGAR Regulates the Initiation and Progression of Pancreatic Cancer. Cancer Cell 2020; 37:168-182.e4. [PMID: 31983610 PMCID: PMC7008247 DOI: 10.1016/j.ccell.2019.12.012] [Citation(s) in RCA: 166] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Revised: 10/02/2019] [Accepted: 12/21/2019] [Indexed: 12/20/2022]
Abstract
The TIGAR protein has antioxidant activity that supports intestinal tissue repair and adenoma development. Using a pancreatic ductal adenocarcinoma (PDAC) model, we show that reactive oxygen species (ROS) regulation by TIGAR supports premalignant tumor initiation while restricting metastasis. Increased ROS in PDAC cells drives a phenotypic switch that increases migration, invasion, and metastatic capacity. This switch is dependent on increased activation of MAPK signaling and can be reverted by antioxidant treatment. In mouse and human, TIGAR expression is modulated during PDAC development, with higher TIGAR levels in premalignant lesions and lower TIGAR levels in metastasizing tumors. Our study indicates that temporal, dynamic control of ROS underpins full malignant progression and helps to rationalize conflicting reports of pro- and anti-tumor effects of antioxidant treatment.
Collapse
Affiliation(s)
| | - Gina M DeNicola
- Department of Cancer Physiology, Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Colin Nixon
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Karen Blyth
- Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK; Institute of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, UK
| | | | - David A Tuveson
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724, USA
| | | |
Collapse
|
44
|
Camacho-Jiménez L, Felix-Portillo M, Nuñez-Hernandez DM, Yepiz-Plascencia G. Molecular cloning and modeling of the Tp53-induced glycolysis and apoptotic regulator (TIGAR) from the Pacific white shrimp Litopenaeus vannamei and its expression in response to hypoxia. FISH & SHELLFISH IMMUNOLOGY 2019; 93:484-491. [PMID: 31377432 DOI: 10.1016/j.fsi.2019.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 07/31/2019] [Accepted: 08/01/2019] [Indexed: 06/10/2023]
Abstract
Hypoxia is a common stressor for aquaculture species. The Pacific white shrimp Litopenaeus vannamei survives low dissolved oxygen (DO) conditions by adjusting its energy metabolism. In vertebrates, the transcription factor p53 regulates glucose metabolism under stress through diverse target genes like the Tp53-induced glycolysis and apoptotic regulator (TIGAR), a protein similar to fructose-2,6-bisphosphatase that has a pro-survival role in cells participating in the defense against oxidative damage. Until now, TIGAR has been not reported in any invertebrate species, including crustaceans. In this work, we report the molecular cloning of the white shrimp TIGAR. The cDNA sequence is 765 bp encoding a 254 amino acid protein. Bioinformatics analyses predicted that although the overall sequence identities of L. vannamei TIGAR and vertebrate proteins are not very high (33.61%-35.34%), they have a remarkable predicted structural similarity with full conservation of catalytic residues, secondary and three-dimensional structures. Gene expression analysis by RT-qPCR revealed that the mRNA abundance of TIGAR in white shrimp is tissue-specific under normal oxygen conditions, with higher expression in gills than hepatopancreas and muscle. Also, gene expression in gills and hepatopancreas is modified by environmental hypoxia, suggesting that TIGAR participates in the cellular tolerance of L. vannamei to this stressor.
Collapse
Affiliation(s)
- Laura Camacho-Jiménez
- Centro de Investigación en Alimentación y Desarrollo (CIAD), A.C, Carretera Gustavo Enrique Astiazarán Rosas, no. 46, col La Victoria, Hermosillo, Sonora, C.P. 83304, Mexico
| | - Monserrath Felix-Portillo
- Facultad de Zootecnia y Ecología, Universidad Autónoma de Chihuahua, Periférico Francisco R. Almada, Km 1, Chihuahua, Chihuahua, 33820, Mexico
| | - Dahlia M Nuñez-Hernandez
- Centro de Investigación en Alimentación y Desarrollo (CIAD), A.C, Carretera Gustavo Enrique Astiazarán Rosas, no. 46, col La Victoria, Hermosillo, Sonora, C.P. 83304, Mexico
| | - Gloria Yepiz-Plascencia
- Centro de Investigación en Alimentación y Desarrollo (CIAD), A.C, Carretera Gustavo Enrique Astiazarán Rosas, no. 46, col La Victoria, Hermosillo, Sonora, C.P. 83304, Mexico.
| |
Collapse
|
45
|
Zhou W, Yao Y, Li J, Wu D, Zhao M, Yan Z, Pang A, Kong L. TIGAR Attenuates High Glucose-Induced Neuronal Apoptosis via an Autophagy Pathway. Front Mol Neurosci 2019; 12:193. [PMID: 31456661 PMCID: PMC6700368 DOI: 10.3389/fnmol.2019.00193] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 07/25/2019] [Indexed: 01/21/2023] Open
Abstract
Hyperglycemia-induced neuronal apoptosis is one of the important reasons for diabetic neuropathy. Long-time exposure to high glucose accelerates many aberrant glucose metabolic pathways and eventually leads to neuronal injury. However, the underlying mechanisms of metabolic alterations remain unknown. TP53-inducible glycolysis and apoptosis regulator (TIGAR) is an endogenous inhibitor of glycolysis and increases the flux of pentose phosphate pathway (PPP) by regulating glucose 6-phosphate dehydrogenase (G6PD). TIGAR is highly expressed in neurons, but its role in hyperglycemia-induced neuronal injury is still unclear. In this study, we observed that TIGAR and G6PD are decreased in the hippocampus of streptozotocin (STZ)-induced diabetic mice. Correspondingly, in cultured primary neurons and Neuro-2a cell line, stimulation with high glucose induced significant neuronal apoptosis and down-regulation of TIGAR expression. Overexpression of TIGAR reduced the number of TUNEL-positive neurons and prevented the activation of Caspase-3 in cultured neurons. Furthermore, enhancing the expression of TIGAR rescued high glucose-induced autophagy impairment and the decrease of G6PD. Nitric oxide synthase 1 (NOS1), a negative regulator of autophagy, is also inhibited by overexpression of TIGAR. Inhibition of autophagy abolished the protective effect of TIGAR in neuronal apoptosis in Neuro-2a. Importantly, overexpression of TIGAR in the hippocampus ameliorated STZ-induced cognitive impairment in mice. Therefore, our data demonstrated that TIGAR may have an anti-apoptosis effect via up-regulation of autophagy in diabetic neuropathy.
Collapse
Affiliation(s)
- Wenjuan Zhou
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Yuan Yao
- Department of Physical Education, Shanghai Normal University, Shanghai, China
| | - Jinxing Li
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dong Wu
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Man Zhao
- Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong Provincial Key Laboratory of Mental Disorders, Department of Human Anatomy and Histoembryology, School of Basic Medical Sciences, Shandong University, Jinan, China
| | - Zongting Yan
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Aimei Pang
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Liang Kong
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
46
|
Li Z, Shao Z, Chen S, Huang D, Peng Y, Chen S, Ma K. TIGAR impedes compression-induced intervertebral disc degeneration by suppressing nucleus pulposus cell apoptosis and autophagy. J Cell Physiol 2019; 235:1780-1794. [PMID: 31317559 DOI: 10.1002/jcp.29097] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Accepted: 06/27/2019] [Indexed: 12/19/2022]
Abstract
To investigate whether TP53-induced glycolysis and apoptosis regulator (TIGAR) participates in compression-induced intervertebral disc (IVD) degeneration, and to determine the regulatory effect of TIGAR on nucleus pulposus (NP) cell autophagy and apoptosis following compression-induced injuries. IVD tissues were collected from human patients undergoing surgery (n = 20) and skeletally mature Sprague-Dawley rats (n = 15). Initially, the effect of compression on the expression of TIGAR was evaluated with in vivo and in vitro models. In addition, TIGAR was silenced to investigate the regulatory effect of TIGAR on compression-induced intracellular reactive oxygen species (ROS) levels, autophagy, and apoptosis in rat NP cells. Furthermore, the P53 inhibitor pifithrin-α (PFTα) and SP1 inhibitor mithramycin A were employed to detect expression level changes of TIGAR and autophagy-associated target molecules. TIGAR expression of NP cells increased gradually in human degenerative IVDs and in rat NP cells under compression both in vivo and in vitro. TIGAR knockdown enhanced compression-induced intracellular ROS generation and the NADPH/NADP+ and GSH/GSSG ratios. Moreover, TIGAR knockdown amplified the compression-induced caspase-3 activation and the apoptosis rate of rat NP cells. Likewise, knockdown of TIGAR significantly accelerated LC3B expression and autophagosome formation in rat NP cells during compression-induced injuries. The results also established that mithramycin A could inhibit TIGAR expression and autophagy levels in NP cells under compression conditions, while PFTα had no similar effect. Our data demonstrated that TIGAR acted as an important endogenous negative regulator of ROS levels, which might inhibit compression-induced apoptosis and autophagy through SP1-dependent mechanisms.
Collapse
Affiliation(s)
- Zhiliang Li
- Department of Orthopaedics, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zengwu Shao
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Songfeng Chen
- Department of Orthopaedics, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Donghua Huang
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yizhong Peng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Sheng Chen
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kaige Ma
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
47
|
Tang Z, He Z. TIGAR promotes growth, survival and metastasis through oxidation resistance and AKT activation in glioblastoma. Oncol Lett 2019; 18:2509-2517. [PMID: 31402948 PMCID: PMC6676722 DOI: 10.3892/ol.2019.10574] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 06/12/2019] [Indexed: 12/20/2022] Open
Abstract
Glioblastoma has a poor prognosis and is one of the most lethal types of cancer in the world. TP53 induced glycolysis regulatory phosphatase (TIGAR) is upregulated in various types of cancer. Therefore, the present study investigated the role of TIGAR in glioblastoma. TIGAR expression was measured in glioma samples and cell lines using immunohistochemistry and western blotting. Reduced nicotinamide adenine dinucleotide phosphate (NADPH), glutathione, malondialdehyde and intracellular reactive oxygen species levels were detected to measure oxidative stress in U-87MG cells following short hairpin RNA (shRNA)-mediated knockdown of TIGAR. Cell viability was determined using an MTT assay for TIGAR-overexpression vector- and TIGAR-shRNA-transfected U-87MG cells. Apoptosis was assessed to evaluate whether TIGAR knockdown sensitized cells to the antitumor effects of temozolomide (TMZ). Migration, invasion and epithelial-mesenchymal transition (EMT) were further assessed using Transwell and western blotting assays. A co-immunoprecipitation assay was used to detect the interaction between TIGAR and protein kinase B (AKT). The results of the present study revealed that TIGAR was positively associated with poor survival and was upregulated in glioblastoma. TIGAR knockdown significantly increased oxidative stress, decreased cell proliferation and exacerbated TMZ-induced apoptosis in U-87MG cells. Additionally, TIGAR knockdown decreased migration, invasion and EMT, and treatment of TIGAR-shRNA-transfected cells with NADPH had no effect on metastasis. In addition, TIGAR promoted AKT activation and bound to AKT. In conclusion, the present study demonstrated that TIGAR may promote glioblastoma growth and progression through oxidation resistance and AKT activation.
Collapse
Affiliation(s)
- Zhi Tang
- Department of Neurosurgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| | - Zhengwen He
- Department of Neurosurgery, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
48
|
Lespay-Rebolledo C, Tapia-Bustos A, Bustamante D, Morales P, Herrera-Marschitz M. The Long-Term Impairment in Redox Homeostasis Observed in the Hippocampus of Rats Subjected to Global Perinatal Asphyxia (PA) Implies Changes in Glutathione-Dependent Antioxidant Enzymes and TIGAR-Dependent Shift Towards the Pentose Phosphate Pathways: Effect of Nicotinamide. Neurotox Res 2019; 36:472-490. [PMID: 31187430 DOI: 10.1007/s12640-019-00064-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 05/08/2019] [Accepted: 05/09/2019] [Indexed: 12/18/2022]
Abstract
We have recently reported that global perinatal asphyxia (PA) induces a regionally sustained increase in oxidized glutathione (GSSG) levels and GSSG/GSH ratio, a decrease in tissue-reducing capacity, a decrease in catalase activity, and an increase in apoptotic caspase-3-dependent cell death in rat neonatal brain up to 14 postnatal days, indicating a long-term impairment in redox homeostasis. In the present study, we evaluated whether the increase in GSSG/GSH ratio observed in hippocampus involves changes in glutathione reductase (GR) and glutathione peroxidase (GPx) activity, the enzymes reducing glutathione disulfide (GSSG) and hydroperoxides, respectively, as well as catalase, the enzyme protecting against peroxidation. The study also evaluated whether there is a shift in the metabolism towards the penthose phosphate pathway (PPP), by measuring TIGAR, the TP53-inducible glycolysis and apoptosis regulator, associated with delayed cell death, further monitoring calpain activity, involved in bax-dependent cell death, and XRCC1, a scaffolding protein interacting with genome sentinel proteins. Global PA was induced by immersing fetus-containing uterine horns removed by a cesarean section from on term rat dams into a water bath at 37 °C for 21 min. Asphyxia-exposed and sibling cesarean-delivered fetuses were manually resuscitated and nurtured by surrogate dams. Animals were euthanized at postnatal (P) days 1 or 14, dissecting samples from hippocampus to be assayed for glutathione, GR, GPx (all by spectrophotometry), catalase (Western blots and ELISA), TIGAR (Western blots), calpain (fluorescence), and XRCC1 (Western blots). One hour after delivery, asphyxia-exposed and control neonates were injected with either 100 μl saline or 0.8 mmol/kg nicotinamide, i.p., shown to protect from the short- and long-term consequences of PA. It was found that global PA produced (i) a sustained increase of GSSG levels and GSSG/GSH ratio at P1 and P14; (ii) a decrease of GR, GPx, and catalase activity at P1 and P14; (iii) a decrease at P1, followed by an increase at P14 of TIGAR levels; (iv) an increase of calpain activity at P14; and (v) an increase of XRCC1 levels, but only at P1. (vi) Nicotinamide prevented the effect of PA on GSSG levels and GSSG/GSH ratio, and on GR, GPx, and catalase activity, also on increased TIGAR levels and calpain activity observed at P14. The present study demonstrates that the long-term impaired redox homeostasis observed in the hippocampus of rats subjected to global PA implies changes in GR, GPx, and catalase, and a shift towards PPP, as indicated by an increase of TIGAR levels at P14.
Collapse
Affiliation(s)
- C Lespay-Rebolledo
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia, 1027, Santiago, Chile
| | - A Tapia-Bustos
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia, 1027, Santiago, Chile
| | - D Bustamante
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia, 1027, Santiago, Chile
| | - P Morales
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia, 1027, Santiago, Chile. .,Department of Neuroscience, Medical Faculty, University of Chile, Av. Independencia, 1027, Santiago, Chile.
| | - M Herrera-Marschitz
- Programme of Molecular & Clinical Pharmacology, ICBM, Medical Faculty, University of Chile, Av. Independencia, 1027, Santiago, Chile.
| |
Collapse
|
49
|
Zhang DM, Zhang T, Wang MM, Wang XX, Qin YY, Wu J, Han R, Sheng R, Wang Y, Chen Z, Han F, Ding Y, Li M, Qin ZH. TIGAR alleviates ischemia/reperfusion-induced autophagy and ischemic brain injury. Free Radic Biol Med 2019; 137:13-23. [PMID: 30978385 DOI: 10.1016/j.freeradbiomed.2019.04.002] [Citation(s) in RCA: 76] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/12/2019] [Accepted: 04/01/2019] [Indexed: 12/29/2022]
Abstract
Autophagy has been reported to play protective and pathogenetic roles in cerebral ischemia/reperfusion (I/R)-induced neuronal injury. Our previous studies have shown that TP53-induced glycolysis and apoptosis regulator (TIGAR) ameliorates I/R-induced brain injury and reduces anti-cancer drug-induced autophagy activation. However, if TIGAR plays a regulatory role on autophagy in cerebral I/R injury is still unclear. The purpose of the present study is to investigate the role of TIGAR on I/R-induced autophagy activation and ischemic neuronal injury in vivo and in vitro stroke models using TIGAR-transgenic (tg-TIGAR) mice and TIGAR-knockout (ko-TIGAR) mice. The present study confirmed that autophagy was activated after I/R. Overexpression of TIGAR in tg-TIGAR mice significantly reduced I/R-induced autophagy activation and alleviated brain damage, while knockout of TIGAR in ko-TIGAR mice enhanced I/R-induced autophagy activation and exacerbated brain injury in vivo and in vitro. The different activity of autophagy in tg-TIGAR and ko-TIGAR primary neurons after OGD/R were largely reversed by knockdown or re-expression of TIGAR in these neurons. The autophagy inhibitor 3-methyladenine (3-MA) partly prevented exacerbation of brain damage induced by ko-TIGAR, whereas the autophagy inducer rapamycin partially abolished the neuroprotective effect of tg-TIGAR. Knockout of TIGAR reduced the levels of phosphorylated mTOR and S6KP70, which were blocked by 3-MA and NADPH after I/R and OGD/R in vivo and in vitro, respectively. Overexpression of TIGAR increased the levels of phosphorylated mTOR and S6KP70 under OGD/R condition, this enhancement effect was suppressed by rapamycin. In conclusion, our current data suggest that TIGAR protected against neuronal injury partly through inhibiting autophagy by regulating the mTOR-S6KP70 signaling pathway.
Collapse
Affiliation(s)
- Ding-Mei Zhang
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Tian Zhang
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Ming-Ming Wang
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Xin-Xin Wang
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Yuan-Yuan Qin
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Junchao Wu
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Rong Han
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Rui Sheng
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Yan Wang
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China
| | - Zhong Chen
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of the Ministry of Health of China, Zhejiang Province Key Laboratory of Neurobiology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Feng Han
- College of Pharmaceutical Science, Nanjing Medical University, Nanjing, 210029, China
| | - Yuqiang Ding
- Institutes of Brain Sciences, State Key Laboratory of Medical Neurobiology, Fudan University, Shanghai, 200032, China
| | - Mei Li
- Institute of Pediatric Research, Children's Hospital of Soochow University, Suzhou, 215025, China.
| | - Zheng-Hong Qin
- Department of Pharmacology, Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Neuropsychiatric Diseases, College of Pharmaceutical Science, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
50
|
Chen C, Mei Q, Wang L, Feng X, Tao X, Qiu C, Zhu J. TIGAR suppresses seizures induced by kainic acid through inhibiting oxidative stress and neuronal apoptosis. Biochem Biophys Res Commun 2019; 515:436-441. [PMID: 31160088 DOI: 10.1016/j.bbrc.2019.05.156] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 05/25/2019] [Indexed: 01/29/2023]
Abstract
TP53-induced glycolysis and apoptosis regulator (TIGAR) activates the pentose phosphate pathway (PPP), which feeds reduced nicotinamide adenine dinucleotide phosphate (NADPH) to the antioxidant glutathione pathway. Oxidative stress-induced neuronal apoptosis is the pathological basis of several neurological disorders, including epilepsy. To determine the potential anti-epileptic action TIGAR in a rodent kainic acid (KA)-induced seizure model. Seizures were induced by the intra-cerebroventricular injection of KA, followed by injection of empty or TIGAR-expressing lentiviral vectors. Immunofluorescence was used to detect the localization of TIGAR in the cortices and hippocampi, and the expression levels of relevant proteins were determined by Western blotting. Oxidative stress-related markers were detected using commercially available kits. Neuronal apoptosis was detected by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining. TIGAR were mainly expressed in the neurons and rarely located in the astrocytes, and increased in the cortices and hippocampi of KA-treated rats in a time-dependent manner. Lentivirus-mediated TIGAR overexpression significantly decreased the oxidative stress and neuronal apoptosis induced by KA, resulting in prolonged seizure latency and lower Racine scores. Our findings indicate that TIGAR has anti-epileptic, anti-oxidant and anti-apoptotic effects, and is therefore a promising therapeutictarget for epilepsy.
Collapse
Affiliation(s)
- Chunyou Chen
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, 317500, China.
| | - Qin Mei
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, 317500, China.
| | - Linlin Wang
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, 317500, China.
| | - Xuewen Feng
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, 317500, China.
| | - Xiaoxiao Tao
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, 317500, China.
| | - Chenfeng Qiu
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, 317500, China.
| | - Jingang Zhu
- Department of Neurology, The First People's Hospital of Wenling, Taizhou, 317500, China.
| |
Collapse
|