1
|
Guo X, Zhou J, La Yan, Liu X, Yuan Y, Ye J, Zhang Z, Chen H, Ma Y, Zhong Z, Luo G, Chen H. Very long-chain fatty acids control peroxisome dynamics via a feedback loop in intestinal stem cells during gut regeneration. Dev Cell 2024; 59:3008-3024.e8. [PMID: 39047737 DOI: 10.1016/j.devcel.2024.06.020] [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: 10/13/2023] [Revised: 04/19/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024]
Abstract
Peroxisome dynamics are crucial for intestinal stem cell (ISC) differentiation and gut regeneration. However, the precise mechanisms that govern peroxisome dynamics within ISCs during gut regeneration remain unknown. Using mouse colitis and Drosophila intestine models, we have identified a negative-feedback control mechanism involving the transcription factors peroxisome proliferator-activated receptors (PPARs) and SOX21. This feedback mechanism effectively regulates peroxisome abundance during gut regeneration. Following gut injury, the released free very long-chain fatty acids (VLCFAs) increase peroxisome abundance by stimulating PPARs-PEX11s signaling. PPARs act to stimulate peroxisome fission and inhibit pexophagy. SOX21, which acts downstream of peroxisomes during ISC differentiation, induces peroxisome elimination through pexophagy while repressing PPAR expression. Hence, PPARs and SOX21 constitute a finely tuned negative-feedback loop that regulates peroxisome dynamics. These findings shed light on the complex molecular mechanisms underlying peroxisome regulation in ISCs, contributing to our understanding of gut renewal and repair.
Collapse
Affiliation(s)
- Xiaoxin Guo
- West China Centre of Excellence for Pancreatitis and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Juanyu Zhou
- West China Centre of Excellence for Pancreatitis and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - La Yan
- West China Centre of Excellence for Pancreatitis and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xingzhu Liu
- West China Centre of Excellence for Pancreatitis and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yu Yuan
- West China Centre of Excellence for Pancreatitis and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jinbao Ye
- West China Centre of Excellence for Pancreatitis and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zehong Zhang
- West China Centre of Excellence for Pancreatitis and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Haiou Chen
- West China Centre of Excellence for Pancreatitis and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yongxin Ma
- Department of Medical Genetics, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan 610041, China
| | - Zhendong Zhong
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Guanzheng Luo
- Key Laboratory of Gene Engineering of the Ministry of Education, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, Guangdong 510275, China
| | - Haiyang Chen
- West China Centre of Excellence for Pancreatitis and Laboratory of Stem Cell and Anti-Aging Research, National Clinical Research Center for Geriatrics and State Key Laboratory of Respiratory Health and Multimorbidity, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
2
|
Lei Y, Klionsky DJ. Transcriptional regulation of autophagy and its implications in human disease. Cell Death Differ 2023; 30:1416-1429. [PMID: 37045910 PMCID: PMC10244319 DOI: 10.1038/s41418-023-01162-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 03/27/2023] [Accepted: 03/29/2023] [Indexed: 04/14/2023] Open
Abstract
Macroautophagy/autophagy is a conserved catabolic pathway that is vital for maintaining cell homeostasis and promoting cell survival under stressful conditions. Dysregulation of autophagy is associated with a variety of human diseases, such as cancer, neurodegenerative diseases, and metabolic disorders. Therefore, this pathway must be precisely regulated at multiple levels, involving epigenetic, transcriptional, post-transcriptional, translational, and post-translational mechanisms, to prevent inappropriate autophagy activity. In this review, we focus on autophagy regulation at the transcriptional level, summarizing the transcription factors that control autophagy gene expression in both yeast and mammalian cells. Because the expression and/or subcellular localization of some autophagy transcription factors are altered in certain diseases, we also discuss how changes in transcriptional regulation of autophagy are associated with human pathophysiologies.
Collapse
Affiliation(s)
- Yuchen Lei
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Daniel J Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
3
|
Zhang R, Wu F, Cheng B, Wang C, Bai B, Chen J. Apelin-13 prevents the effects of oxygen-glucose deprivation/reperfusion on bEnd.3 cells by inhibiting AKT-mTOR signaling. Exp Biol Med (Maywood) 2023; 248:146-156. [PMID: 36573455 PMCID: PMC10041053 DOI: 10.1177/15353702221139186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Autophagy plays works by degrading misfolded proteins and dysfunctional organelles and maintains intracellular homeostasis. Apelin-13 has been investigated as an agent that might protect the blood-brain barrier (BBB) from cerebral ischemia/reperfusion (I/R) injury. In this study, we examined whether apelin-13 protects cerebral microvascular endothelial cells, important components of the BBB, from I/R injury by regulating autophagy. To mimic I/R injury, the mouse cerebral microvascular endothelia l cell line bEnd 3 undergoes the process of oxygen and glucose deprivation and re feeding in the process of culture. Cell viability was detected using a commercial kit, and cell migration was monitored by in vitro scratch assay. The tight junction (TJ) proteins ZO-1 and occludin; the autophagy markers LC3 II, beclin 1, and p62; and components of the AKT-mTOR signaling pathway were detected by Western blotting and immunofluorescence. To confirm the role of autophagy in OGD/R and the protective effect of apelin-13, we treated the cells with 3-methyladenine (3-MA), a pharmacological inhibitor of autophagy. Our results demonstrated that OGD/R increased autophagic activity but decreased viability, abundance of TJs, and migration. Viability and TJ abundance were further reduced when the OGD/R group was treated with 3-MA. These results indicated that bEnd.3 upregulates autophagy to ameliorate the effects of OGD/R injury on viability and TJs, but that the autophagy induced by OGD/R alone is not sufficient to protect against the effect on cell migration. Treatment of OGD/R samples with apelin-13 markedly increased viability, TJ abundance, and migration, as well as autophagic activity, whereas 3-MA inhibited this increase, suggesting that apelin-13 exerted its protective effects by upregulating autophagy.
Collapse
Affiliation(s)
- Rumin Zhang
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | - Fei Wu
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | - Baohua Cheng
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | - Chunmei Wang
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | - Bo Bai
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
| | - Jing Chen
- Neurobiology Key Laboratory of Jining Medical University, Jining 272067, China
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| |
Collapse
|
4
|
Xiong L, Zhang J, Shi H, Zhu G, Ji X, Li M, Zhu P, Luo K. Downregulation of TNFAIP1 alleviates OGD/R‑induced neuronal damage by suppressing Nrf2/GPX4‑mediated ferroptosis. Exp Ther Med 2022; 25:25. [PMID: 36561622 PMCID: PMC9748634 DOI: 10.3892/etm.2022.11724] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Accepted: 10/25/2022] [Indexed: 11/24/2022] Open
Abstract
TNFα-induced protein 1 (TNFAIP1) serve a role in neurovascular disease. However, the potential role and molecular mechanism of TNFAIP1 in cerebral ischemia-reperfusion (I/R) remains elusive. In the present study, reverse transcription-quantitative PCR and western blotting were used to assess TNFAIP1 mRNA and protein expression levels in PC12 cells. Furthermore, using Cell Counting Kit-8, flow cytometry and western blotting, cell viability and apoptosis were evaluated. Oxidative stress was evaluated using DCFH-DA staining and ELISA was used for assessment of inflammatory factors. Expression of components in the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway and ferroptosis were assessed using western blotting analysis and an iron assay kit. TNFAIP1 expression was significantly upregulated in oxygen glucose deprivation and reperfusion (OGD/R)-injured PC12 cells. However, knocking down TNFAIP1 expression restored PC12 cell viability and decreased apoptosis following OGD/R-challenge. Furthermore, TNFAIP1 silencing significantly suppressed OGD/R-induced oxidative stress and inflammatory damage in PC12 cells. TNFAIP1 knockdown inhibited ferroptosis via activation of the Nrf2 signaling pathway in OGD/R-injured PC12 cells. Erastin treatment reversed the beneficial effects of TNFAIP1 knockdown on PC12 cell viability, apoptosis alleviation, oxidative stress and inflammation following OGD/R treatment. These results suggested that TNFAIP1 knockdown could alleviate OGD/R-induced neuronal cell damage by suppressing Nrf2-mediated ferroptosis, which might lay the foundation for the investigation of targeted-therapy for cerebral I/R injury in clinic.
Collapse
Affiliation(s)
- Lie Xiong
- Central Laboratory, Zhejiang Chinese Medical University-Affiliated Jiaxing Traditional Chinese Medicine Hospital, Jiaxing, Zhejiang 314001, P.R. China
| | - Jingruo Zhang
- Department of Acupuncture, Zhejiang Chinese Medical University-Affiliated Jiaxing Traditional Chinese Medicine Hospital, Jiaxing, Zhejiang 314001, P.R. China
| | - Hanqiang Shi
- Central Laboratory, Zhejiang Chinese Medical University-Affiliated Jiaxing Traditional Chinese Medicine Hospital, Jiaxing, Zhejiang 314001, P.R. China
| | - Gaofeng Zhu
- Department of Acupuncture, Zhejiang Chinese Medical University-Affiliated Jiaxing Traditional Chinese Medicine Hospital, Jiaxing, Zhejiang 314001, P.R. China
| | - Xiaoyan Ji
- Department of Acupuncture, Zhejiang Chinese Medical University-Affiliated Jiaxing Traditional Chinese Medicine Hospital, Jiaxing, Zhejiang 314001, P.R. China
| | - Mengjiao Li
- Department of Acupuncture, Zhejiang Chinese Medical University-Affiliated Jiaxing Traditional Chinese Medicine Hospital, Jiaxing, Zhejiang 314001, P.R. China
| | - Ping Zhu
- Department of Neurosurgery, Zhejiang Chinese Medical University-Affiliated Jiaxing Traditional Chinese Medicine Hospital, Jiaxing, Zhejiang 314001, P.R. China
| | - Kaitao Luo
- Department of Acupuncture, Zhejiang Chinese Medical University-Affiliated Jiaxing Traditional Chinese Medicine Hospital, Jiaxing, Zhejiang 314001, P.R. China,Correspondence to: Dr Kaitao Luo, Department of Acupuncture, Zhejiang Chinese Medical University-Affiliated Jiaxing Traditional Chinese Medicine Hospital, 1501 East Zhongshan Road, Jiaxing, Zhejiang 314001, P.R. China
| |
Collapse
|
5
|
Yang CB, Liu J, Tong BCK, Wang ZY, Zhu Z, Su CF, Sreenivasmurthy SG, Wu JX, Iyaswamy A, Krishnamoorthi S, Huang SY, Cheung KH, Song JX, Tan JQ, Lu JH, Li M. TFEB, a master regulator of autophagy and biogenesis, unexpectedly promotes apoptosis in response to the cyclopentenone prostaglandin 15d-PGJ2. Acta Pharmacol Sin 2022; 43:1251-1263. [PMID: 34417577 PMCID: PMC9061728 DOI: 10.1038/s41401-021-00711-7] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 06/01/2021] [Indexed: 12/22/2022]
Abstract
Transcriptional factor EB (TFEB), a master regulator of autophagy and lysosomal biogenesis, is generally regarded as a pro-survival factor. Here, we identify that besides its effect on autophagy induction, TFEB exerts a pro-apoptotic effect in response to the cyclopentenone prostaglandin 15-deoxy-∆-12,14-prostaglandin J2 (15d-PGJ2). Specifically, 15d-PGJ2 promotes TFEB translocation from the cytoplasm into the nucleus to induce autophagy and lysosome biogenesis via reactive oxygen species (ROS) production rather than mTORC1 inactivation. Surprisingly, TFEB promotes rather than inhibits apoptosis in response to 15d-PGJ2. Mechanistically, ROS-mediated TFEB translocation into the nucleus transcriptionally upregulates the expression of ATF4, which is required for apoptosis elicited by 15d-PGJ2. Additionally, inhibition of TFEB activation by ROS scavenger N-acetyl cysteine or inhibition of protein synthesis by cycloheximide effectively compromises ATF4 upregulation and apoptosis in response to 15d-PGJ2. Collectively, these results indicate that ROS-induced TFEB activation exerts a novel role in promoting apoptosis besides its role in regulating autophagy in response to 15d-PGJ2. This work not only evidences how TFEB is activated by 15d-PGJ2, but also unveils a previously unexplored role of ROS-dependent activation of TFEB in modulating cell apoptosis in response to 15d-PGJ2.
Collapse
Affiliation(s)
- Chuan-bin Yang
- grid.263817.90000 0004 1773 1790Department of Geriatrics, Shenzhen People’s Hospital, (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 China ,grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jia Liu
- grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Benjamin Chun-Kit Tong
- grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Zi-ying Wang
- grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China ,grid.258164.c0000 0004 1790 3548Interdisciplinary Institute for Personalized Medicine in Brain Disorders, Jinan University, Guangzhou, 510632 China
| | - Zhou Zhu
- grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Cheng-fu Su
- grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Sravan Gopalkrishnashetty Sreenivasmurthy
- grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Jia-xi Wu
- grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ashok Iyaswamy
- grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Senthilkumar Krishnamoorthi
- grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Shi-ying Huang
- grid.263817.90000 0004 1773 1790Department of Geriatrics, Shenzhen People’s Hospital, (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020 China
| | - King-ho Cheung
- grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China
| | - Ju-xian Song
- grid.221309.b0000 0004 1764 5980Mr. and Mrs. Ko Chi Ming Centre for Parkinson’s Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China ,grid.411866.c0000 0000 8848 7685Medical College of Acupuncture-Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, 510006 China
| | - Jie-qiong Tan
- grid.216417.70000 0001 0379 7164Center for Medical Genetics and Hunan Key Laboratory of Animal Model for Human Diseases, School of Life Sciences, Central South University, Changsha, 410006 China
| | - Jia-hong Lu
- grid.437123.00000 0004 1794 8068State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Min Li
- Mr. and Mrs. Ko Chi Ming Centre for Parkinson's Disease Research, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR, China.
| |
Collapse
|
6
|
Faghfouri AH, Khajebishak Y, Payahoo L, Faghfuri E, Alivand M. PPAR-gamma agonists: Potential modulators of autophagy in obesity. Eur J Pharmacol 2021; 912:174562. [PMID: 34655597 DOI: 10.1016/j.ejphar.2021.174562] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2021] [Revised: 09/21/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022]
Abstract
Autophagy pathways are involved in the pathogenesis of some obesity related health problems. As obesity is a nutrient sufficiency condition, autophagy process can be altered in obesity through AMP activated protein kinase (AMPK) inhibition. Peroxisome proliferator-activated receptor-gamma (PPAR-gamma) as the main modulator of adipogenesis process can be effective in the regulation of obesity related phenotypes. As well, it has been revealed that PPAR-gamma and its agonists can regulate autophagy in different normal or cancer cells. However, their effects on autophagy modulation in obesity have been investigated in the limited number of studies. In the current comprehensive mechanistic review, we aimed to investigate the possible mechanisms of action of PPAR-gamma on the process of autophagy in obesity through narrating the effects of PPAR-gamma on autophagy in the non-obesity conditions. Moreover, mode of action of PPAR-gamma agonists on autophagy related implications comprehensively reviewed in the various studies. Understanding the different effects of PPAR-gamma agonists on autophagy in obesity can help to develop a new approach to management of obesity.
Collapse
Affiliation(s)
- Amir Hossein Faghfouri
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Department of Community Nutrition, Faculty of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yaser Khajebishak
- Department of Nutrition, Maragheh University of Medical Sciences, Maragheh, I.R., Iran
| | - Laleh Payahoo
- Department of Nutrition, Maragheh University of Medical Sciences, Maragheh, I.R., Iran
| | - Elnaz Faghfuri
- Digestive Disease Research Center, Ardabil University of Medical Sciences, Ardabil, Iran.
| | - Mohammadreza Alivand
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
7
|
Huang R, Zhang C, Wang X, Hu H. PPARγ in Ischemia-Reperfusion Injury: Overview of the Biology and Therapy. Front Pharmacol 2021; 12:600618. [PMID: 33995008 PMCID: PMC8117354 DOI: 10.3389/fphar.2021.600618] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 03/11/2021] [Indexed: 12/12/2022] Open
Abstract
Ischemia-reperfusion injury (IRI) is a complex pathophysiological process that is often characterized as a blood circulation disorder caused due to various factors (such as traumatic shock, surgery, organ transplantation, burn, and thrombus). Severe metabolic dysregulation and tissue structure destruction are observed upon restoration of blood flow to the ischemic tissue. Theoretically, IRI can occur in various tissues and organs, including the kidney, liver, myocardium, and brain, among others. The advances made in research regarding restoring tissue perfusion in ischemic areas have been inadequate with regard to decreasing the mortality and infarct size associated with IRI. Hence, the clinical treatment of patients with severe IRI remains a thorny issue. Peroxisome proliferator-activated receptor γ (PPARγ) is a member of a superfamily of nuclear transcription factors activated by agonists and is a promising therapeutic target for ameliorating IRI. Therefore, this review focuses on the role of PPARγ in IRI. The protective effects of PPARγ, such as attenuating oxidative stress, inhibiting inflammatory responses, and antagonizing apoptosis, are described, envisaging certain therapeutic perspectives.
Collapse
Affiliation(s)
- Ruizhen Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chiyu Zhang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xing Wang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Honglin Hu
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Rodríguez-Pascau L, Britti E, Calap-Quintana P, Dong YN, Vergara C, Delaspre F, Medina-Carbonero M, Tamarit J, Pallardó FV, Gonzalez-Cabo P, Ros J, Lynch DR, Martinell M, Pizcueta P. PPAR gamma agonist leriglitazone improves frataxin-loss impairments in cellular and animal models of Friedreich Ataxia. Neurobiol Dis 2021; 148:105162. [PMID: 33171227 DOI: 10.1016/j.nbd.2020.105162] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/14/2020] [Accepted: 11/03/2020] [Indexed: 01/08/2023] Open
Abstract
Friedreich ataxia (FRDA), the most common autosomal recessive ataxia, is characterized by degeneration of the large sensory neurons and spinocerebellar tracts, cardiomyopathy, and increased incidence in diabetes. The underlying pathophysiological mechanism of FRDA, driven by a significantly decreased expression of frataxin (FXN), involves increased oxidative stress, reduced activity of enzymes containing iron‑sulfur clusters (ISC), defective energy production, calcium dyshomeostasis, and impaired mitochondrial biogenesis, leading to mitochondrial dysfunction. The peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated transcriptional factor playing a key role in mitochondrial function and biogenesis, fatty acid storage, energy metabolism, and antioxidant defence. It has been previously shown that the PPARγ/PPARγ coactivator 1 alpha (PGC-1α) pathway is dysregulated when there is frataxin deficiency, thus contributing to FRDA pathogenesis and supporting the PPARγ pathway as a potential therapeutic target. Here we assess whether MIN-102 (INN: leriglitazone), a novel brain penetrant and orally bioavailable PPARγ agonist with an improved profile for central nervous system (CNS) diseases, rescues phenotypic features in cellular and animal models of FRDA. In frataxin-deficient dorsal root ganglia (DRG) neurons, leriglitazone increased frataxin protein levels, reduced neurite degeneration and α-fodrin cleavage mediated by calpain and caspase 3, and increased survival. Leriglitazone also restored mitochondrial membrane potential and partially reversed decreased levels of mitochondrial Na+/Ca2+ exchanger (NCLX), resulting in an improvement of mitochondrial functions and calcium homeostasis. In frataxin-deficient primary neonatal cardiomyocytes, leriglitazone prevented lipid droplet accumulation without increases in frataxin levels. Furthermore, leriglitazone improved motor function deficit in YG8sR mice, a FRDA mouse model. In agreement with the role of PPARγ in mitochondrial biogenesis, leriglitazone significantly increased markers of mitochondrial biogenesis in FRDA patient cells. Overall, these results suggest that targeting the PPARγ pathway by leriglitazone may provide an efficacious therapy for FRDA increasing the mitochondrial function and biogenesis that could increase frataxin levels in compromised frataxin-deficient DRG neurons. Alternately, leriglitazone improved the energy metabolism by increasing the fatty acid β-oxidation in frataxin-deficient cardiomyocytes without elevation of frataxin levels. This could be linked to a lack of significant mitochondrial biogenesis and cardiac hypertrophy. The results reinforced the different tissue requirement in FRDA and the pleiotropic effects of leriglitazone that could be a promising therapy for FRDA.
Collapse
Affiliation(s)
| | - Elena Britti
- Departament de Ciències Mèdiques Bàsiques, IRBLleida. Universitat de Lleida, Lleida 25198, Spain
| | - Pablo Calap-Quintana
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-INCLIVA, Valencia 46010, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Yi Na Dong
- Department of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Fabien Delaspre
- Departament de Ciències Mèdiques Bàsiques, IRBLleida. Universitat de Lleida, Lleida 25198, Spain
| | - Marta Medina-Carbonero
- Departament de Ciències Mèdiques Bàsiques, IRBLleida. Universitat de Lleida, Lleida 25198, Spain
| | - Jordi Tamarit
- Departament de Ciències Mèdiques Bàsiques, IRBLleida. Universitat de Lleida, Lleida 25198, Spain
| | - Federico V Pallardó
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-INCLIVA, Valencia 46010, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Pilar Gonzalez-Cabo
- Department of Physiology, Faculty of Medicine and Dentistry, University of Valencia-INCLIVA, Valencia 46010, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Joaquim Ros
- Departament de Ciències Mèdiques Bàsiques, IRBLleida. Universitat de Lleida, Lleida 25198, Spain
| | - David R Lynch
- Department of Pediatrics and Neurology, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Marc Martinell
- Minoryx Therapeutics SL., Mataró 08302, Barcelona, Spain; Minoryx Therapeutics BE., Gosselies 6041, Charleroi, Belgium
| | - Pilar Pizcueta
- Minoryx Therapeutics SL., Mataró 08302, Barcelona, Spain.
| |
Collapse
|
9
|
Feng C, Li D, Chen M, Jiang L, Liu X, Li Q, Geng C, Sun X, Yang G, Zhang L, Yao X. Citreoviridin induces myocardial apoptosis through PPAR-γ-mTORC2-mediated autophagic pathway and the protective effect of thiamine and selenium. Chem Biol Interact 2019; 311:108795. [PMID: 31419397 DOI: 10.1016/j.cbi.2019.108795] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 07/30/2019] [Accepted: 08/13/2019] [Indexed: 12/25/2022]
Abstract
Citreoviridin (CIT), a mycotoxin and ATP synthase inhibitor, is regarded as one of aetiology factors of cardiac beriberi and Keshan disease. Thiamine (VB1) and selenium (Se) improve the recovery of these two diseases respectively. The underlying mechanisms of cardiotoxic effect of CIT and cardioprotective effect of VB1 and Se have not been fully elucidated. In this study, we found that ectopic ATP synthase was more sensitive to CIT treatment than mitochondrial ATP synthase in H9c2 cardiomyocytes. CIT inhibited the transcriptional activity of peroxisome proliferator activated receptor gamma (PPAR-γ) in mice hearts and H9c2 cells. PPAR-γ agonist attenuated the inhibitory effect of CIT on mechanistic target of rapamycin complex 2 (mTORC2) and stimulatory effect of CIT on autophagy in cardiomyocytes. CIT induced apoptosis through lysosomal-mitochondrial axis in cardiomyocytes. PPAR-γ agonist and autophagy inhibitor alleviated CIT-induced apoptosis and accelerated cardiac biomarker. VB1 and Se accelerated the basal transcriptional activity of PPAR-γ in mice hearts and H9c2 cells. Furthermore, VB1 and Se reversed the effect of CIT on PPAR-γ, autophagy and apoptosis. Our findings defined PPAR-γ-mTORC2-autophagy pathway as the key link between CIT cardiotoxicity and cardioprotective effect of VB1 and Se. The present study would shed new light on the pathogenesis of cardiomyopathy and the cardioprotective mechanism of micronutrients.
Collapse
Affiliation(s)
- Chang Feng
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Dandan Li
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Min Chen
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Liping Jiang
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Xiaofang Liu
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Qiujuan Li
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Chengyan Geng
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Xiance Sun
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Guang Yang
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China
| | - Lianchun Zhang
- Department of Nursing, The Second Affiliated Hospital of Dalian Medical University, 467 Zhongshan Road, Dalian, 116023, China
| | - Xiaofeng Yao
- Department of Preventive Medicine, Dalian Medical University, 9 W Lushun South Road, Dalian, 116044, China.
| |
Collapse
|
10
|
Liu X, Zhang M, Zhang H, Zhao A, Sun J, Tang W. [Role of PPAR-γ-regulated autophagy in genistein-induced inhibition of hepatic stellate cell activation]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2019; 39:561-565. [PMID: 31140420 DOI: 10.12122/j.issn.1673-4254.2019.05.10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the inhibitory effect of genistein on activation of hepatic stellate cells (HSCs) in vitro and the role of the autophagy pathway regulated by PPAR-γ in mediating this effect. METHODS Cultured HSC-T6 cells were exposed to different concentrations of genistein for 48 h, and HSC activation was verified by detecting the expressions of -SMA and 1(I) collagen; autophagy activation in the cells was determined by detecting the expressions of LC3-II and p62 using Western blotting. The autophagy inhibitor 3-MA was used to confirm the role of autophagy in genistein-induced inhibition of HSC activation. A PPAR-γ inhibitor was used to explore the role of PPAR-γ in activating autophagy in the HSCs. RESULTS Genistein at concentrations of 5 and 50 μmol/L significantly inhibited the expressions of -SMA and 1(I) collagen (P < 0.05), markedly upregulated the expressions of PPAR-γ and the autophagy-related protein LC3-II (P < 0.05) and significantly down-regulated the expression of the ubiqutin-binding protein p62 (P < 0.05) in HSC-T6 cells. The cells pretreated with 3-MA prior to genistein treatment showed significantly increased protein expressions of -SMA and 1(I) collagen compared with the cells treated with genistein only (P < 0.05). Treatment with the PPAR-γ inhibitor obviously lowered the expression of LC3-II and enhanced the expression p62 in genistein-treated HSC-T6 cells, suggesting the activation of the autophagy pathway. CONCLUSIONS PPAR-γ- regulated autophagy plays an important role in mediating genistein-induced inhibition of HSC activation in vitro.
Collapse
Affiliation(s)
- Xipeng Liu
- Department of Clinical Nutrition, Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Meifang Zhang
- Department of Clinical Nutrition, Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Haifeng Zhang
- Department of Clinical Nutrition, Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Anda Zhao
- Department of Clinical Nutrition, Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Juan Sun
- Department of Clinical Nutrition, Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| | - Wen Tang
- Department of Clinical Nutrition, Ninth People's Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200011, China
| |
Collapse
|
11
|
Lu J, Liu LJ, Zhu JL, Shen Y, Zhuang ZW, Zhu CL. Hypothermic properties of dexmedetomidine provide neuroprotection in rats following cerebral ischemia-reperfusion injury. Exp Ther Med 2019; 18:817-825. [PMID: 31258715 DOI: 10.3892/etm.2019.7613] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 04/18/2019] [Indexed: 01/04/2023] Open
Abstract
Dexmedetomidine (Dex) is a sedative and analgesic agent that is widely administered to patients admitted to the intensive care unit, and has been demonstrated to result in hypothermia. Many patients have been revealed to benefit from therapeutic hypothermia, which can mitigate cerebral ischemia/reperfusion (I/R) injury following successful cardiopulmonary resuscitation. However, studies investigating the efficacy of Dex in I/R treatment is lacking. The present study aimed to investigate the efficacy of Dex in mitigating neuronal damage, and to determine the possible mechanism of its effects in a rat model of cardiac arrest (CA). CA was induced in Sprague-Dawley rats by asphyxiation for 5 min. Following successful resuscitation, the surviving rats were randomly divided into two treatment groups; one group was intraperitoneally administered with Dex (D group), whereas the control group was treated with normal saline (N group). Critical parameters, including core temperature and blood pressure were monitored following return of spontaneous circulation (ROSC). Arterial blood samples were collected at 10 min after surgery (baseline) 30 and 120 min post-ROSC; and neurological deficit scores (NDS) of the rats were taken 12 or 24 h after ROSC prior to euthanasia. The hippocampal tissue was then removed for analysis by histology, electron microscopy and western blotting. Rats in the D group exhibited a lower core temperature and higher NDS scores compared with the N group (P<0.05). In addition, Dex injection resulted in reduced expression of apoptotic and autophagy-associated factors in the hippocampus (P<0.05). Dex treatment induced hypothermia and improved neurological function in rats after ROSC following resuscitation from CA by inhibiting neuronal apoptosis and reducing autophagy, which suggested that Dex may be a potential therapy option for patients with CA.
Collapse
Affiliation(s)
- Jian Lu
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China.,Department of Emergency and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu 215002, P.R. China
| | - Li-Jun Liu
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Jian-Liang Zhu
- Department of Emergency and Critical Care Medicine, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215007, P.R. China
| | - Yi Shen
- Department of Emergency and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu 215002, P.R. China
| | - Zhi-Wei Zhuang
- Department of Emergency and Critical Care Medicine, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, Jiangsu 215002, P.R. China
| | - Chang-Lai Zhu
- Key Laboratory of Neuroregeneration, Nantong Medical College of Nantong University, Nantong, Jiangsu 226200, P.R. China
| |
Collapse
|
12
|
Sun B, Ou H, Ren F, Huan Y, Zhong T, Gao M, Cai H. Propofol inhibited autophagy through Ca 2+/CaMKKβ/AMPK/mTOR pathway in OGD/R-induced neuron injury. Mol Med 2018; 24:58. [PMID: 30470173 PMCID: PMC6251140 DOI: 10.1186/s10020-018-0054-1] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 09/24/2018] [Indexed: 11/22/2022] Open
Abstract
Background The neuroprotective role of propofol (PPF) in cerebral ischemia-reperfusion (I/R) has recently been highlighted. This study aimed to explore whether the neuroprotective mechanisms of PPF were linked to its regulation of Ca2+/CaMKKβ (calmodulin-dependent protein kinase kinase β)/AMPK (AMP-activated protein kinase)/mTOR (mammalian target of rapamycin)/autophagy pathway. Methods Cultured primary rat cerebral cortical neurons were treated with oxygen-glucose deprivation and re-oxygenation (OGD/R) to mimic cerebral I/R injury in vitro. Results Compared with the control neurons, OGD/R exposure successfully induced neuronal I/R injury. Furthermore, OGD/R exposure notably caused autophagy induction, reflected by augmented LC3-II/LC3-I ratio and Beclin 1 expression, decreased p62 expression, and increased LC3 puncta formation. Moreover, OGD/R exposure induced elevation of intracellular Ca2+ concentration ([Ca2+]i). However, PPF treatment significantly antagonized OGD/R-triggered cell injury, autophagy induction, and [Ca2+]i elevation. Further investigation revealed that both autophagy induction by rapamycin and [Ca2+]i elevation by the Ca2+ ionophore ionomycin significantly reversed the PPF-mediated amelioration of OGD/R-triggered cell injury. Importantly, ionomycin also significantly abrogated the PPF-mediated suppression of autophagy and CaMKKβ/AMPK/mTOR signaling in OGD/R-exposed neurons. Additionally, activation of CaMKKβ/AMPK/mTOR signaling abrogated the PPF-mediated autophagy suppression. Conclusion Our findings demonstrate that PPF antagonized OGD/R-triggered neuronal injury, which might be mediated, at least in part, via inhibition of autophagy through Ca2+/CaMKKβ/AMPK/mTOR pathway. Electronic supplementary material The online version of this article (10.1186/s10020-018-0054-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Bei Sun
- Department of Anesthesiology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, People's Republic of China
| | - Hao Ou
- Department of Emergency and Critical Care Medicine, The Third Xiangya Hospital, Central South University, 410013, Changsha, Hunan, People's Republic of China.,Translational Medicine Center of Sepsis, Department of Pathophysiology, The Third Xiangya Hospital, Central South University, 410013, Changsha, Hunan, People's Republic of China
| | - Fei Ren
- Department of Anesthesiology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, People's Republic of China
| | - Ye Huan
- Department of Anesthesiology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, People's Republic of China
| | - Tao Zhong
- Department of Anesthesiology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, People's Republic of China
| | - Min Gao
- Department of Emergency and Critical Care Medicine, The Third Xiangya Hospital, Central South University, 410013, Changsha, Hunan, People's Republic of China.,Translational Medicine Center of Sepsis, Department of Pathophysiology, The Third Xiangya Hospital, Central South University, 410013, Changsha, Hunan, People's Republic of China
| | - Hongwei Cai
- Department of Anesthesiology, Xiangya Hospital, Central South University, 410078, Changsha, Hunan, People's Republic of China.
| |
Collapse
|
13
|
Hydroxysafflor Yellow A Shows Protection against PPAR γ Inactivation in Nitrosative Neurons. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:9101740. [PMID: 30410641 PMCID: PMC6206554 DOI: 10.1155/2018/9101740] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Revised: 08/01/2018] [Accepted: 08/28/2018] [Indexed: 12/16/2022]
Abstract
Peroxynitrite-mediated nitrosative stress in the brain has been associated with various neurodegenerative disorders. Recent evidence highlights peroxisome proliferator-activated receptor γ (PPARγ) as a critical neuroprotective factor in neurodegenerative diseases. Here, we observed the effect of the herb hydroxysafflor yellow A (HSYA) during nitrosative stress in neurons and investigated the mechanism based on PPARγ protection. We found that a single exposure of primary neurons to peroxynitrite donor SIN-1 caused neuronal injury, which was accompanied by the increase of PPARγ nitration status and lack of activation of the receptor, as measured by PPARγ DNA-binding activity, by agonist (15d-PGJ2 or rosiglitazone) stimulation. The crucial role of PPARγ in neuronal defense against nitrosative stress was verified by showing that pretreatment with 15d-PGJ2 or rosiglitazone attenuated SIN-1-induced neuronal injury but pretreatment with GW9662, a PPARγ antagonist, aggravated SIN-1-induced neuronal injury. The addition of HSYA not only inhibited SIN-1-induced neuronal damage but prevented PPARγ nitrative modification and resumed PPARγ activity stimulated by either 15d-PGJ2 or rosiglitazone. Furthermore, HSYA also showed the ability to rescue the neuroprotective effect of 15d-PGJ2 or rosiglitazone when the agonists were coincubated with SIN-1. Finally, in vivo experiments demonstrated that the administration of HSYA also efficiently blocked PPARγ nitration and loss of activity in the SIN-1-injected hippocampus and reversed the increased neuronal susceptibility which was supported by the inhibition of Bcl-2 protein downregulation induced by SIN-1. The results suggest that HSYA protects neurons from nitrosative stress through keeping PPARγ as a functional receptor, allowing a more effective activation of this neuroprotective factor by the endogenous or exogenous agonist. Our findings provide new clues in understanding the role of the neuroprotective potential of the herbal HSYA.
Collapse
|
14
|
Yang G, Wang N, Seto SW, Chang D, Liang H. Hydroxysafflor yellow a protects brain microvascular endothelial cells against oxygen glucose deprivation/reoxygenation injury: Involvement of inhibiting autophagy via class I PI3K/Akt/mTOR signaling pathway. Brain Res Bull 2018; 140:243-257. [PMID: 29775658 DOI: 10.1016/j.brainresbull.2018.05.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Revised: 03/27/2018] [Accepted: 05/11/2018] [Indexed: 12/31/2022]
Abstract
The present study aimed to test whether Hydroxysafflor yellow A (HSYA) protects the brain microvascular endothelial cells (BMECs) injury induced by oxygen glucose deprivation/reoxygenation (OGD/R) via the PI3K/Akt/mTOR autophagy signaling pathway. Primary rat BMECs were cultured and identified by the expression of factor VIII-related antigen before being exposed to OGD/R to imitate ischemia/reperfusion (I/R) damage in vitro. The protective effect of HSYA was evaluated by assessing (1) cellular morphologic and ultrastructural changes; (2) cell viability and cytotoxicity; (3) transendothelial electrical resistance (TEER) of monolayer BMECs; (4) cell apoptosis; (5) fluorescence intensity of LC3B; (6) LC3 mRNA expression; (7) protein expressions of LC3, Beclin-1, Zonula occludens-1 (ZO-1), phospho-Akt (p-Akt), Akt, phospho-mTOR (p-mTOR) and mTOR. It was found that HSYA (20, 40, and 80 μM) and 3-MA effectively reversed the cellular morphological and ultrastructural changes, increased cell survival, normalized the permeability of BMECs, and suppressed apoptosis induced by OGD/R (2 h OGD followed by 24 h reoxygenation). Concurrently, HSYA and 3-MA also inhibited OGD/R-induced autophagy evidenced by the decreased number of autophagosomes and down-regulated levels of LC3 and Beclin-1 proteins and mRNAs. HSYA (80 μM), in combination with 3-MA showed a synergistic effect. Mechanistic studies revealed that HSYA (80 μM) markedly increased the levels of p-Akt and p-mTOR proteins. Blockade of PI3K activity by ZSTK474 abolished its anti-autophagic and pro-survival effect and lowered both Akt and mTOR phosphorylation levels. Taken together, these results suggest that HSYA protects BMECs against OGD/R-induced injury by inhibiting autophagy via the Class I PI3K/Akt/mTOR signaling pathway.
Collapse
Affiliation(s)
- Guang Yang
- Anhui University of Chinese Medicine, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China.
| | - Ning Wang
- Anhui University of Chinese Medicine, Hefei 230012, China; Key Laboratory of Xin'an Medicine, Ministry of Education, Hefei 230012, China; National Institute of Complementary Medicine, Western Sydney University, Penrith, NSW 2751, Australia.
| | - Sai Wang Seto
- National Institute of Complementary Medicine, Western Sydney University, Penrith, NSW 2751, Australia
| | - Dennis Chang
- National Institute of Complementary Medicine, Western Sydney University, Penrith, NSW 2751, Australia
| | - Huangzheng Liang
- School of Medical, Western Sydney University, Penrith, NSW 2751, Australia
| |
Collapse
|
15
|
Abstract
Hepatic ischemia-reperfusion injury is a common complication of liver surgery and an important cause of liver dysfunction after operation. The pathogenesis of liver ischemia-reperfusion injury is very complex, involving many factors. Autophagy is a lysosomal degradation pathway on which eukaryotic cells rely to maintain the cell homeostasis. Autophagy plays an important role in the process of liver ischemia-reperfusion injury. However, the specific role and mechanism of autophagy in liver ischemia-reperfusion injury are still controversial. In this paper, we reivew the role and mechanism of autophagy in hepatic ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Zi-Yi Li
- Department of General Surgery, People's Hospital of Daqing, Daqing 163316, Heilongjiang Province, China
| | - Li-Quan Tong
- Department of General Surgery, People's Hospital of Daqing, Daqing 163316, Heilongjiang Province, China
| |
Collapse
|
16
|
Yang Z, Wang J, Pan Z, Zhang Y. miR-143-3p regulates cell proliferation and apoptosis by targeting IGF1R and IGFBP5 and regulating the Ras/p38 MAPK signaling pathway in rheumatoid arthritis. Exp Ther Med 2018; 15:3781-3790. [PMID: 29581736 PMCID: PMC5863597 DOI: 10.3892/etm.2018.5907] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 01/12/2018] [Indexed: 01/16/2023] Open
Abstract
It has been demonstrated that the deregulation of microRNAs (miRNAs) affects the development of rheumatoid arthritis (RA). The primary objective of the current study was to determine the role of miR-143-3p in the progression of RA. The expression of miR-143-3p in synovium taken from patients with RA was assessed by reverse transcription-quantitative polymerase chain reaction. The expression of miR-143-3p was higher in synovium tissues of RA than that of osteoarthritis (OA). The decreased expression of miR-143-3p suppressed cell proliferation and promoted apoptosis in vitro. In addition, inhibition of miR-143-3p decreased levels of inflammatory cytokines, as determined by an enzyme-linked immunosorbent assay. IGF1R and IGFBP5 were found to be the target genes of miR-143-3p, and it was demonstrated that miR-143-3p regulated the proliferation and apoptosis of MH7A cells by targeting IGF1R and IGFBP5. Furthermore, TNF-α treatment stimulated the Ras/p38 mitogen activated protein kinase (MAPK) signaling pathway, whereas miR-143-3p inhibition suppressed it. The results of the current study indicate that miR-143-3p may regulate cell proliferation and apoptosis by targeting IGF1R and IGFBP5 expression and regulating the Ras/p38 MAPK signaling pathways. Therefore, miR-143-3p may be a novel therapeutic target in RA.
Collapse
Affiliation(s)
- Zhenguo Yang
- Department of Orthopaedics, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China.,Department of Orthopaedics, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250001, P.R. China
| | - Jifu Wang
- Department of Orthopaedics, East Courtyard Area of General Hospital of Shandong Yankuang Group, Zoucheng, Shandong 273500, P.R. China
| | - Zhuangzhuang Pan
- Department of Surgery, Lixia District People's Hospital, Jinan, Shandong 250014, P.R. China
| | - Yihang Zhang
- Graduate Student Education Center, Shandong Academy of Medical Science, Jinan, Shandong 250062, P.R. China
| |
Collapse
|
17
|
Liu YJ, Wang DY, Yang YJ, Lei WF. Effects and mechanism of dexmedetomidine on neuronal cell injury induced by hypoxia-ischemia. BMC Anesthesiol 2017; 17:117. [PMID: 28854873 PMCID: PMC5577810 DOI: 10.1186/s12871-017-0413-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 08/25/2017] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND The present study aims to investigate the protective effects of dexmedetomidine (DMED) on hypoxia ischemia injury induced by oxygen and glucose deprivation (OGD) in PC12 and primary neuronal cells. METHODS PC12 cells exposed to OGD was used to establish ischemia model. The OGD-induced cell injury was evaluated by alterations of cell viability, apoptosis and expressions of apoptosis-associated proteins. Oxidative stress and expressions of neurotrophic factors after OGD and DMED treatments were also explored. The activation of possible involved signaling pathways were studied after OGD and DMED treatments, along with the addition of inhibitors of these pathways. Finally, the effects of DMED on primary neuronal cells were verified according to the alterations of inflammatory cytokines release and oxidative stress. RESULTS DMED obviously increased cell viability and reduced cell apoptosis as well as ratio of Bax/Bcl-2 in OGD-treated PC12 cells. Then, the OGD-induced changes of LDH, MDA, SOD and GSH-Px as well as decreases of neurotrophic factors were all ameliorated by DMED treatment. Key kinases in Notch/NF-κB signaling pathway were up-regulated by OGD, whereas the up-regulations were decreased by DMED. In addition, inhibitor of Notch or NF-κB could augment the effects of DMED on OGD-induced cell injury. Finally, the protective effects of DMED were verified in primary neuronal cells. CONCLUSION DMED had protective effect on OGD-induced PC12 cell injury, depending on its anti-apoptotic, anti-oxidative activity and the inhibition of Notch/NF-κB activation. Our findings suggested that DMED could be used as a potential therapeutic drug for cerebral ischemia.
Collapse
Affiliation(s)
- Ya-Jun Liu
- Department of Anesthesiology, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Jinan, Shandong 250012 China
- Department of Anesthesiology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013 China
| | - Duan-Yu Wang
- Department of Anesthesiology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013 China
| | - Yong-Jian Yang
- Department of Anesthesiology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong 250013 China
| | - Wei-Fu Lei
- Department of Anesthesiology, Qilu Hospital of Shandong University, No. 107, Wenhua West Road, Jinan, Shandong 250012 China
| |
Collapse
|
18
|
Füllgrabe J, Ghislat G, Cho DH, Rubinsztein DC. Transcriptional regulation of mammalian autophagy at a glance. J Cell Sci 2017; 129:3059-66. [PMID: 27528206 DOI: 10.1242/jcs.188920] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Macroautophagy, hereafter referred to as autophagy, is a catabolic process that results in the lysosomal degradation of cytoplasmic contents ranging from abnormal proteins to damaged cell organelles. It is activated under diverse conditions, including nutrient deprivation and hypoxia. During autophagy, members of the core autophagy-related (ATG) family of proteins mediate membrane rearrangements, which lead to the engulfment and degradation of cytoplasmic cargo. Recently, the nuclear regulation of autophagy, especially by transcription factors and histone modifiers, has gained increased attention. These factors are not only involved in rapid responses to autophagic stimuli, but also regulate the long-term outcome of autophagy. Now there are more than 20 transcription factors that have been shown to be linked to the autophagic process. However, their interplay and timing appear enigmatic as several have been individually shown to act as major regulators of autophagy. This Cell Science at a Glance article and the accompanying poster highlights the main cellular regulators of transcription involved in mammalian autophagy and their target genes.
Collapse
Affiliation(s)
- Jens Füllgrabe
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Wellcome/MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - Ghita Ghislat
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Wellcome/MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - Dong-Hyung Cho
- Department of Gerontology, Graduate School of East-West Medical Science, Kyung Hee University, Yongin 17104, South Korea
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Wellcome/MRC Building, Addenbrooke's Hospital, Hills Road, Cambridge CB2 0XY, UK
| |
Collapse
|
19
|
Chen JL, Duan WJ, Luo S, Li S, Ma XH, Hou BN, Cheng SY, Fang SH, Wang Q, Huang SQ, Chen YB. Ferulic acid attenuates brain microvascular endothelial cells damage caused by oxygen-glucose deprivation via punctate-mitochondria-dependent mitophagy. Brain Res 2017; 1666:17-26. [DOI: 10.1016/j.brainres.2017.04.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 03/19/2017] [Accepted: 04/14/2017] [Indexed: 12/26/2022]
|
20
|
Liu T, Zhang Q, Mo W, Yu Q, Xu S, Li J, Li S, Feng J, Wu L, Lu X, Zhang R, Li L, Cheng K, Zhou Y, Zhou S, Kong R, Wang F, Dai W, Chen K, Xia Y, Lu J, Zhou Y, Zhao Y, Guo C. The protective effects of shikonin on hepatic ischemia/reperfusion injury are mediated by the activation of the PI3K/Akt pathway. Sci Rep 2017; 7:44785. [PMID: 28322249 PMCID: PMC5359611 DOI: 10.1038/srep44785] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Accepted: 02/13/2017] [Indexed: 12/20/2022] Open
Abstract
Hepatic ischemia/reperfusion (I/R) injury, which can result in severe liver injury and dysfunction, occurs in a variety of conditions such as liver transplantation, shock, and trauma. Cell death in hepatic I/R injury has been linked to apoptosis and autophagy. Shikonin plays a significant protective role in ischemia/reperfusion injury. The purpose of the present study was to investigate the protective effect of shikonin on hepatic I/R injury and explore the underlying mechanism. Mice were subjected to segmental (70%) hepatic warm ischemia to induce hepatic I/R injury. Two doses of shikonin (7.5 and 12.5 mg/kg) were administered 2 h before surgery. Balb/c mice were randomly divided into four groups: normal control, I/R, and shikonin preconditioning at two doses (7.5 and 12.5 mg/kg). The serum and liver tissues were collected at three time points (3, 6, and 24 h). Shikonin significantly reduced serum AST and ALT levels and improved pathological features. Shikonin affected the expression of Bcl-2, Bax, caspase 3, caspase 9, Beclin-1, and LC3, and upregulated PI3K and p-Akt compared with the levels in the I/R group. Shikonin attenuated hepatic I/R injury by inhibiting apoptosis and autophagy through a mechanism involving the activation of PI3K/Akt signaling.
Collapse
Affiliation(s)
- Tong Liu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - QingHui Zhang
- Department of Clinical Laboratory, Kunshan First People’s Hospital Affiliated to Jiangsu University, 215300, Kunshan, JiangSu, China
| | - Wenhui Mo
- Department of Gastroenterology, Minhang Hospital, Shanghai Medical School of Fudan University, Shanghai, 201100, China
| | - Qiang Yu
- Department of Gastroenterology, Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Shizan Xu
- Department of Gastroenterology, Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Jingjing Li
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Sainan Li
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jiao Feng
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Liwei Wu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Xiya Lu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Rong Zhang
- Department of Gastroenterology, Shanghai Tenth Hospital, School of Clinical Medicine of Nanjing Medical University, Shanghai 200072, China
| | - Linqiang Li
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Keran Cheng
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Yuqing Zhou
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Shunfeng Zhou
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Rui Kong
- The School of Medicine of Soochow University, Suzhou 215006, China
| | - Fan Wang
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Weiqi Dai
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Kan Chen
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yujing Xia
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Jie Lu
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yingqun Zhou
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Yan Zhao
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| | - Chuanyong Guo
- Department of Gastroenterology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai 200072, China
| |
Collapse
|
21
|
Physiological and Pathological Roles of 15-Deoxy-Δ12,14-Prostaglandin J2 in the Central Nervous System and Neurological Diseases. Mol Neurobiol 2017; 55:2227-2248. [PMID: 28299574 DOI: 10.1007/s12035-017-0435-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 02/03/2017] [Indexed: 12/29/2022]
|
22
|
Huang XP, Ding H, Yang XQ, Li JX, Tang B, Liu XD, Tang YH, Deng CQ. Synergism and mechanism of Astragaloside IV combined with Ginsenoside Rg1 against autophagic injury of PC12 cells induced by oxygen glucose deprivation/reoxygenation. Biomed Pharmacother 2017; 89:124-134. [PMID: 28219050 DOI: 10.1016/j.biopha.2017.02.015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/03/2017] [Accepted: 02/07/2017] [Indexed: 01/30/2023] Open
Abstract
The aim of this study was to explore the effect by which the combination of Astragaloside IV (AST IV) and Ginsenoside Rg1 (Rg1) resisted autophagic injury in PC12 cells induced by oxygen glucose deprivation/reoxygenation (OGD/R). We studied the nature of the interaction between AST IV and Rg1 that inhibited autophagy through the Isobologram method, and investigated the synergistic mechanism via the PI3K I/Akt/mTOR and PI3K III/Becline-1/Bcl-2 signaling pathways. Our results showed that, based on the 50% inhibiting concentration (IC50), AST IV combined with Rg1 at a 1:1 ratio resulted in a synergistic effect, whereas the combination of the two had an antagonistic effect on autophagy at ratios of 1:2 and 2:1. Meanwhile, AST IV and Rg1 alone increased cell survival and decreased lactate dehydrogenase (LDH) leakage induced by OGD/R, reduced autophagosomes and the LC3 II positive patch, down-regulated the LC3 II/LC3 I ratio and up-regulated the p62 protein; the 1:1 combination enhanced these effects. Mechanistic study showed that Rg1 and the 1:1 combination increased the phosphorylation of PI3K I, Akt and mTOR; the effects of the combination were greater than those of the drugs alone. AST IV and the 1:1 combination suppressed the expression of PI3K III and Becline-1, and the combination elevated Bcl-2 protein expression; the effects of the combination were better than those of the drugs alone. These results suggest that after 2 h-OGD followed by reoxygenation for 24h, PC12 cells suffer excessive autophagy and damage, which are blocked by AST IV or Rg1; moreover, the combination of AST IV and Rg1 at a 1:1 ratio of their IC50 concentrations has a synergistic inhibition on autophagic injury. The synergistic mechanism may be associated with the PI3K I/Akt/mTOR and PI3K III/Becline-1/Bcl-2 signaling pathways.
Collapse
Affiliation(s)
- Xiao-Ping Huang
- Molecular Pathology Laboratory, Hunan Provincial Key Laboratory for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Huang Ding
- Molecular Pathology Laboratory, Hunan Provincial Key Laboratory for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Xiao-Qian Yang
- Molecular Pathology Laboratory, Hunan Provincial Key Laboratory for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Jing-Xian Li
- Molecular Pathology Laboratory, Hunan Provincial Key Laboratory for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Biao Tang
- Hunan Education Department's Key Laboratory of Cell Biology and Molecular Technology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Xiao-Dan Liu
- Hunan Education Department's Key Laboratory of Cell Biology and Molecular Technology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Ying-Hong Tang
- Hunan Education Department's Key Laboratory of Cell Biology and Molecular Technology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Chang-Qing Deng
- Molecular Pathology Laboratory, Hunan Provincial Key Laboratory for Prevention and Treatment of Integrated Traditional Chinese and Western Medicine on Cardio-Cerebral Diseases, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China.
| |
Collapse
|
23
|
Liu Y, Lu Z, Cui M, Yang Q, Tang Y, Dong Q. Tissue kallikrein protects SH-SY5Y neuronal cells against oxygen and glucose deprivation-induced injury through bradykinin B2 receptor-dependent regulation of autophagy induction. J Neurochem 2016; 139:208-220. [PMID: 27248356 DOI: 10.1111/jnc.13690] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 05/21/2016] [Accepted: 05/24/2016] [Indexed: 12/22/2022]
Affiliation(s)
- Yanping Liu
- Department of Neurology; Huashan Hospital; State Key Laboratory of Medical Neurobiology; Fudan University; Shanghai China
| | - Zhengyu Lu
- Department of Neurology; Yueyang Hospital of Integrated Traditional Chinese and Western Medicine; Shanghai University of Traditional Chinese Medicine; Shanghai China
| | - Mei Cui
- Department of Neurology; Huashan Hospital; State Key Laboratory of Medical Neurobiology; Fudan University; Shanghai China
| | - Qi Yang
- Department of Neurology; Huashan Hospital; State Key Laboratory of Medical Neurobiology; Fudan University; Shanghai China
| | - Yuping Tang
- Department of Neurology; Huashan Hospital; State Key Laboratory of Medical Neurobiology; Fudan University; Shanghai China
| | - Qiang Dong
- Department of Neurology; Huashan Hospital; State Key Laboratory of Medical Neurobiology; Fudan University; Shanghai China
| |
Collapse
|
24
|
Salidroside protects cortical neurons against glutamate-induced cytotoxicity by inhibiting autophagy. Mol Cell Biochem 2016; 419:53-64. [PMID: 27357827 DOI: 10.1007/s11010-016-2749-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 06/18/2016] [Indexed: 12/20/2022]
Abstract
Recent evidence suggests that glutamate-induced cytotoxicity contributes to autophagic neuron death and is partially mediated by increased oxidative stress. Salidroside has been demonstrated to have neuroprotective effects in glutamate-induced neuronal damage. The precise mechanism of its regulatory role in neuronal autophagy is, however, poorly understood. This study aimed to probe the effects and mechanisms of salidroside in glutamate-induced autophagy activation in cultured rat cortical neurons. Cell viability assay, Western blotting, coimmunoprecipitation, and small interfering RNA were performed to analyze autophagy activities during glutamate-evoked oxidative injury. We found that salidroside protected neonatal neurons from glutamate-induced apoptotic cell death. Salidroside significantly attenuated the LC3-II/LC3-I ratio and expression of Beclin-1, but increased (SQSTM1)/p62 expression under glutamate exposure. Pretreatment with 3-methyladenine (3-MA), an autophagy inhibitor, decreased LC3-II/LC3-I ratio, attenuated glutamate-induced cell injury, and mimicked some of the protective effects of salidroside against glutamate-induced cell injury. Molecular analysis demonstrated that salidroside inhibited cortical neuron autophagy in response to glutamate exposure through p53 signaling by increasing the accumulation of cytoplasmic p53. Salidroside inhibited the glutamate-induced dissociation of the Bcl-2-Beclin-1 complex with minor affects on the PI3K/Akt/mTOR signaling pathways. These data demonstrate that the inhibition of autophagy could be responsible for the neuroprotective effects of salidroside on glutamate-induced neuronal injury.
Collapse
|
25
|
Xu YB, Zhang PJ, Liu Q, Mao XN, Wang CC. Role of autophagy related protein Beclin 1 in model of hepatic ischemia-reperfusion injury. Shijie Huaren Xiaohua Zazhi 2016; 24:209-214. [DOI: 10.11569/wcjd.v24.i2.209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Surgical resection is the optimal treatment for primary liver cancer, but surgery is often faced with recession of the liver function reserve, ischemia-reperfusion injury of the residual liver and other disadvantages. Autophagy is a form of programmed cell death after hepatic ischemia-reperfusion, and its role in ischemia-reperfusion injury is a hotspot of research in recent years. In the experimental research of simulated liver ischemia-reperfusion injury, the variation of autophagy related protein Beclin 1 is often detected, which suggests the change of autophagy activity. Many pretreatment methods have been found to be able to reduce the level of Beclin 1 and relieve the hepatic damage in the model of hepatic ischemia-reperfusion injury. Here we discuss the research progress in understanding the role of Beclin 1 in hepatic ischemia-reperfusion injury.
Collapse
|
26
|
Chang R, Zhou R, Qi X, Wang J, Wu F, Yang W, Zhang W, Sun T, Li Y, Yu J. Protective effects of aloin on oxygen and glucose deprivation-induced injury in PC12 cells. Brain Res Bull 2016; 121:75-83. [PMID: 26772628 DOI: 10.1016/j.brainresbull.2016.01.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 12/28/2015] [Accepted: 01/04/2016] [Indexed: 01/11/2023]
Abstract
The present study aims to determine whether aloin could protect cells from ischemic and reperfusion injury in vitro and to elucidate the related mechanisms. Oxygen and glucose deprivation model in PC12 cells was used in the present study. 2-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT), lactate dehydrogenase (LDH) assay and Hoechst 33342 nuclear staining were used to evaluate the protective effects of aloin, at concentrations of 10, 20, or 40 μg/mL in PC12 cells. PCR was applied to detect fluorescence caspase-3, Bax and Bcl-2 mRNA expression in PC12 cells. The contents of malondialdehyde (MDA), superoxide dismutase (SOD) activity were evaluated by biochemical method. The concentration of intracellular-free calcium [Ca(2+)]i, mitochondrial membrane potential (MMP) were determined to estimate the degree of neuronal damage. It was shown that aloin (10, 20, and 40 μg/mL) significantly attenuated PC12 cells damage with characteristics of an increased injured cells absorbance of MTT and releases of LDH, decreasing cell apoptosis, and antagonizing decreases in SOD activity and increase in MDA level induced by OGD-reoxygenation. Meanwhile pretreatment with aloin significantly reduced injury-induced intracellular ROS, increased MMP (P<0.01), but it inhibited [Ca(2+)]i (P<0.01) elevation in a dose-dependent manner. Furthermore, pre-treatment with aloin significantly up-regulated Bcl-2 mRNA expression, down-regulated Bax mRNA expression and consequently activated caspase-3 mRNA expression in a dose-dependent manner. The results indicated that the protection of aloin on OGD-induced apoptosis in PC12 cells is associated with its suppression on OGD-induced oxidative stress and protection on mitochondrial function and inhibition of caspase activity. Alion could be a promising candidate in the development of a novel class of anti-ischemic agent.
Collapse
Affiliation(s)
- Renyuan Chang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Ru Zhou
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Xue Qi
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Jing Wang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Fan Wu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Wenli Yang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Wannian Zhang
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China
| | - Tao Sun
- Ningxia Key Lab of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Yinchuan, China
| | - Yuxiang Li
- College of Nursing, Ningxia Medical University, Yinchuan, China.
| | - Jianqiang Yu
- Department of Pharmacology, College of Pharmacy, Ningxia Medical University, Yinchuan, China; Ningxia Hui Medicine Modern Engineering Research Center, Ningxia Medical University, Yinchuan, China.
| |
Collapse
|
27
|
15d-PGJ2 Reduced Microglia Activation and Alleviated Neurological Deficit of Ischemic Reperfusion in Diabetic Rat Model. BIOMED RESEARCH INTERNATIONAL 2015; 2015:864509. [PMID: 26844229 PMCID: PMC4710931 DOI: 10.1155/2015/864509] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/09/2015] [Accepted: 11/19/2015] [Indexed: 01/18/2023]
Abstract
To investigate the effect of PPARγ agonist 15d-PGJ2 treatment on the microglia activation and neurological deficit of ischemia reperfusion in diabetic rat model, adult Sprague-Dawley rats were sacrificed for the research. The rats were randomly categorized into four groups: (1) sham-operated group; (2) standard ischemia group; (3) diabetic ischemia group; (4) diabetic ischemia group with diabetes and treated with 15d-PGJ2. Compared to the sham-operated group, all the ischemic groups have significantly severer neurological deficits, more TNF-α and IL-1 expression, increased labeling of apoptotic cells, increased CD68 positive staining of brain lesion, and increased volume of infarct and cerebral edema in both 24 hours and 7 days after reperfusion. Interestingly, reduced neurological deficits, decreased TNF-α and IL-1 expression, less apoptotic cells and CD68 positive staining, and alleviated infarct and cerebral edema volume were observed when 15d-PGJ2 was intraperitoneally injected after reperfusion in diabetic ischemia group, suggesting its neuroprotective role in regulating microglia activation, which may have a therapeutic application in the future.
Collapse
|
28
|
Zhang Z, Guo M, Zhao S, Xu W, Shao J, Zhang F, Wu L, Lu Y, Zheng S. The update on transcriptional regulation of autophagy in normal and pathologic cells: A novel therapeutic target. Biomed Pharmacother 2015; 74:17-29. [DOI: 10.1016/j.biopha.2015.06.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2015] [Accepted: 06/15/2015] [Indexed: 02/08/2023] Open
|
29
|
Thouennon E, Cheng Y, Falahatian V, Cawley NX, Loh YP. Rosiglitazone-activated PPARγ induces neurotrophic factor-α1 transcription contributing to neuroprotection. J Neurochem 2015; 134:463-70. [PMID: 25940785 DOI: 10.1111/jnc.13152] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 03/04/2015] [Accepted: 04/02/2015] [Indexed: 12/20/2022]
Abstract
Brain peroxisome proliferator-activated receptor gamma (PPARγ), a member of the nuclear receptor superfamily of ligand-dependent transcription factors, is involved in neuroprotection. It is activated by the drug rosiglitazone, which then can increase the pro-survival protein B-cell lymphoma 2 (BCL-2), to mediate neuroprotection. However, the mechanism underlying this molecular cascade remains unknown. Here, we show that the neuroprotective protein neurotrophic factor-α1 (NF-α1), which also induces the expression of BCL-2, has a promoter that contains PPARγ-binding sites that are activated by rosiglitazone. Treatment of Neuro2a cells and primary hippocampal neurons with rosiglitazone increased endogenous NF-α1 expression and prevented H2 O2 -induced cytotoxicity. Concomitant with the increase in NF-α1, BCL-2 was also increased in these cells. When siRNA against NF-α1 was used, the induction of BCL-2 by rosiglitazone was prevented, and the neuroprotective effect of rosiglitazone was reduced. These results demonstrate that rosiglitazone-activated PPARγ directly induces the transcription of NF-α1, contributing to neuroprotection in neurons. We proposed the following cascade for neuroprotection against oxidative stress by rosiglitazone: Rosiglitazone enters the neuron and binds to peroxisome proliferator-activated receptor gamma (PPARγ) in the nucleus. The PPARγ-rosiglitazone complex binds to the neurotrophic factor-α1 (NF-α1) promoter and activates the transcription of NF-α1 mRNA which is then translated to the protein. NF-α1 is the secreted, binds to a cognate receptor and activates the extracellular signal-regulated kinases (ERK) pathway. This in turn enhances the expression of the pro-survival protein, B-cell lymphoma 2 (BCL-2) and inhibition of caspase 3 (Csp-3) to mediate neuroprotection under oxidative stress. Akt, protein kinase B (PKB).
Collapse
Affiliation(s)
- Erwan Thouennon
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Yong Cheng
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Vida Falahatian
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Niamh X Cawley
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Yoke Peng Loh
- Section on Cellular Neurobiology, Program on Developmental Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|