1
|
Wang Y, Li D, Zhang L, Yin Z, Han Z, Ge X, Li M, Zhao J, Zhang S, Zuo Y, Xiong X, Gao H, Liu Q, Chen F, Lei P. Exosomes derived from microglia overexpressing miR-124-3p alleviate neuronal endoplasmic reticulum stress damage after repetitive mild traumatic brain injury. Neural Regen Res 2024; 19:2010-2018. [PMID: 38227530 DOI: 10.4103/1673-5374.391189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 09/18/2023] [Indexed: 01/17/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202409000-00033/figure1/v/2024-01-16T170235Z/r/image-tiff We previously reported that miR-124-3p is markedly upregulated in microglia-derived exosomes following repetitive mild traumatic brain injury. However, its impact on neuronal endoplasmic reticulum stress following repetitive mild traumatic brain injury remains unclear. In this study, we first used an HT22 scratch injury model to mimic traumatic brain injury, then co-cultured the HT22 cells with BV2 microglia expressing high levels of miR-124-3p. We found that exosomes containing high levels of miR-124-3p attenuated apoptosis and endoplasmic reticulum stress. Furthermore, luciferase reporter assay analysis confirmed that miR-124-3p bound specifically to the endoplasmic reticulum stress-related protein IRE1α, while an IRE1α functional salvage experiment confirmed that miR-124-3p targeted IRE1α and reduced its expression, thereby inhibiting endoplasmic reticulum stress in injured neurons. Finally, we delivered microglia-derived exosomes containing miR-124-3p intranasally to a mouse model of repetitive mild traumatic brain injury and found that endoplasmic reticulum stress and apoptosis levels in hippocampal neurons were significantly reduced. These findings suggest that, after repetitive mild traumatic brain injury, miR-124-3 can be transferred from microglia-derived exosomes to injured neurons, where it exerts a neuroprotective effect by inhibiting endoplasmic reticulum stress. Therefore, microglia-derived exosomes containing miR-124-3p may represent a novel therapeutic strategy for repetitive mild traumatic brain injury.
Collapse
Affiliation(s)
- Yan Wang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Dai Li
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Lan Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhenyu Yin
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhaoli Han
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xintong Ge
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Meimei Li
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Jing Zhao
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Shishuang Zhang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Zuo
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiangyang Xiong
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Han Gao
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Qiang Liu
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Fanglian Chen
- Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
2
|
Wang Y, Wang Y, Yin H, Xiao Z, Ren Z, Ma X, Zhang J, Fu X, Zhang F, Zeng L. BI1 Activates Autophagy and Mediates TDP43 to Regulate ALS Pathogenesis. Mol Neurobiol 2024:10.1007/s12035-024-04313-2. [PMID: 38954254 DOI: 10.1007/s12035-024-04313-2] [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: 03/01/2024] [Accepted: 06/16/2024] [Indexed: 07/04/2024]
Abstract
Amyotrophic lateral sclerosis (ALS) is the most prevalent motor neuron disease in adults. Currently, there are no known drugs or clinical approaches that have demonstrated efficacy in treating ALS. Mitochondrial function and autophagy have been identified as crucial mechanisms in the development of ALS. While Bax inhibitor 1 (BI1) has been implicated in neurodegenerative diseases, its exact mechanism remains unknown. This study investigates the therapeutic impact of BI1 overexpression on ALS both in vivo and in vitro, revealing its ability to mitigate SOD1G93A-induced apoptosis, nuclear damage, mitochondrial dysfunction, and axonal degeneration of motor neurons. At the same time, BI1 prolongs onset time and lifespan of ALS mice, improves motor function, and alleviates neuronal damage, muscle damage, neuromuscular junction damage among other aspects. The findings indicate that BI1 can inhibit pathological TDP43 morphology and initially stimulate autophagy through interaction with TDP43. This study establishes a solid theoretical foundation for understanding the regulation of autophagy by BI1 and TDP43 while shedding light on the pathogenesis of ALS through their interaction - offering new concepts and targets for clinical implementation and drug development.
Collapse
Affiliation(s)
- Yu Wang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun, 130012, China
| | - Yuxiang Wang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun, 130012, China
| | - Hanlan Yin
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun, 130012, China
| | - Zixuan Xiao
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun, 130012, China
| | - Zhichao Ren
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun, 130012, China
| | - Xueting Ma
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun, 130012, China
| | - Jingtian Zhang
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin, 130118, China
| | - Xueqi Fu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun, 130012, China
| | - Fuqiang Zhang
- Scientific Research Centre of China-Japan Union Hospital, Jilin University, Changchun, 130033, China.
| | - Linlin Zeng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Science, Jilin University, Changchun, 130012, China.
| |
Collapse
|
3
|
Fang F, Liu P, Huang H, Feng X, Li L, Sun Y, Kaufman RJ, Hu Y. RGC-specific ATF4 and/or CHOP deletion rescues glaucomatous neurodegeneration and visual function. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:286-295. [PMID: 37547290 PMCID: PMC10400881 DOI: 10.1016/j.omtn.2023.07.015] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 07/11/2023] [Indexed: 08/08/2023]
Abstract
Endoplasmic reticulum (ER) stress has been linked with various acute and chronic neurodegenerative diseases. We previously found that optic nerve (ON) injury and diseases induce neuronal ER stress in retinal ganglion cells (RGCs). We further demonstrated that germline deletion of CHOP preserves the structure and function of both RGC somata and axons in mouse glaucoma models. Here we report that RGC-specific deletion of CHOP and/or its upstream regulator ATF4 synergistically promotes RGC and ON survival and preserves visual function in mouse ON crush and silicone oil-induced ocular hypertension (SOHU) glaucoma models. Consistently, topical application of the ATF4/CHOP chemical inhibitor ISRIB or RGC-specific CRISPR-mediated knockdown of the ATF4 downstream effector Gadd45a also delivers significant neuroprotection in the SOHU glaucoma model. These studies suggest that blocking the neuronal intrinsic ATF4/CHOP axis of ER stress is a promising neuroprotection strategy for neurodegeneration.
Collapse
Affiliation(s)
- Fang Fang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Pingting Liu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Haoliang Huang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Xue Feng
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Liang Li
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| | - Randal J. Kaufman
- Degenerative Diseases Program, Center for Genetic Disorders and Aging Research, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto, CA 94304, USA
| |
Collapse
|
4
|
Faulkner MB, Rizk M, Bazzi Z, Dysko RC, Zhang Z. Sex-Specific Effects of Buprenorphine on Endoplasmic Reticulum Stress, Abnormal Protein Accumulation, and Cell Loss After Pediatric Mild Traumatic Brain Injury in Mice. Neurotrauma Rep 2023; 4:573-585. [PMID: 37752926 PMCID: PMC10518695 DOI: 10.1089/neur.2023.0051] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2023] Open
Abstract
Traumatic brain injury (TBI) in children often leads to poor developmental outcomes attributable to progressive cell loss caused by secondary injuries, including endoplasmic reticulum (ER) stress. Buprenorphine (BPN) is commonly used in children for pain management; however, the effects of BPN on ER stress in the pediatric population are still inconclusive. This study investigated the sex-specific effects of BPN on ER stress, abnormal protein accumulation, and cell loss in a mouse impact acceleration model of pediatric TBI. On post-natal day 20-21 (P20-21), male and female littermates were randomized into sham, TBI + saline and TBI + BPN groups. BPN (0.075 mg/kg) was administered to TBI + BPN mice at 30 min after injury and then every 6-12 h for 2 days. The impact of BPN was evaluated at 1, 3, and 7 days post-injury. We found that TBI induced more prominent ER stress pathway activation at 1 and 3 days post-injury in males, compared to females, whereas abnormal protein accumulation and cell loss were more severe in females at 7 days post-injury, compared with males. Although BPN partially ameliorated abnormal protein accumulation and cell loss in both males and females, BPN only decreased ER stress pathway activation in males, not in females. In conclusion, BPN exhibits sex-specific effects on ER stress, abnormal protein accumulation, and cell loss in a time-dependent manner at the acute phase after pediatric TBI, which provides the rationale to assess the potential effects of BPN on long-term outcomes after pediatric TBI in both males and females.
Collapse
Affiliation(s)
- Megan B. Faulkner
- Department of Natural Sciences, University of Michigan–Dearborn, Dearborn, Michigan, USA
| | - Mariam Rizk
- Department of Natural Sciences, University of Michigan–Dearborn, Dearborn, Michigan, USA
| | - Zahraa Bazzi
- Department of Natural Sciences, University of Michigan–Dearborn, Dearborn, Michigan, USA
| | - Robert C. Dysko
- Unit for Laboratory Animal Medicine, University of Michigan–Ann Arbor, Ann Arbor, Michigan, USA
| | - Zhi Zhang
- Department of Natural Sciences, University of Michigan–Dearborn, Dearborn, Michigan, USA
| |
Collapse
|
5
|
Targeting autophagy, oxidative stress, and ER stress for neurodegenerative diseases treatment. J Control Release 2022; 345:147-175. [DOI: 10.1016/j.jconrel.2022.03.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/27/2022] [Accepted: 03/01/2022] [Indexed: 12/13/2022]
|
6
|
Chen K, Yang LN, Lai C, Liu D, Zhu LQ. Role of Grina/Nmdara1 in the Central Nervous System Diseases. Curr Neuropharmacol 2021; 18:861-867. [PMID: 32124700 PMCID: PMC7569322 DOI: 10.2174/1570159x18666200303104235] [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: 12/10/2019] [Revised: 01/11/2020] [Accepted: 02/13/2020] [Indexed: 12/24/2022] Open
Abstract
Glutamate receptor, ionotropic, N-methyl-D-aspartate associated protein 1 (GRINA) is a member of the NMDA receptors (NMDARs) and is involved in several neurological diseases, which governs the key processes of neuronal cell death or the release of neurotransmitters. Upregulation of GRINA has been reported in multiple diseases in human beings, such as major depressive disorder (MDD) and schizophrenia (SCZ), with which the underlying mechanisms remain elusive. In this review, we provide a general overview of the expression and physiological function of GRINA in the central nervous system (CNS) diseases, including stroke, depression, epilepsy, SCZ, and Alzheimer’s disease (AD).
Collapse
Affiliation(s)
- Kai Chen
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine,
Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People’s Republic of China,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, People’s Republic of China
| | - Liu Nan Yang
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine,
Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People’s Republic of China,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, People’s Republic of China
| | - Chuan Lai
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science
and Technology, Wuhan 430030, People’s Republic of China,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, People’s Republic of China
| | - Dan Liu
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science
and Technology, Wuhan 430030, People’s Republic of China,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, People’s Republic of China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine,
Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People’s Republic of China,The Institute for Brain Research, Collaborative Innovation Center for Brain Science, Huazhong University of Science and Technology, Wuhan 430030, People’s Republic of China
| |
Collapse
|
7
|
Alswady-Hoff M, Erdem JS, Phuyal S, Knittelfelder O, Sharma A, Fonseca DDM, Skare Ø, Slupphaug G, Zienolddiny S. Long-Term Exposure to Nanosized TiO 2 Triggers Stress Responses and Cell Death Pathways in Pulmonary Epithelial Cells. Int J Mol Sci 2021; 22:ijms22105349. [PMID: 34069552 PMCID: PMC8161419 DOI: 10.3390/ijms22105349] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/14/2021] [Accepted: 05/17/2021] [Indexed: 02/03/2023] Open
Abstract
There is little in vitro data available on long-term effects of TiO2 exposure. Such data are important for improving the understanding of underlying mechanisms of adverse health effects of TiO2. Here, we exposed pulmonary epithelial cells to two doses (0.96 and 1.92 µg/cm2) of TiO2 for 13 weeks and effects on cell cycle and cell death mechanisms, i.e., apoptosis and autophagy were determined after 4, 8 and 13 weeks of exposure. Changes in telomere length, cellular protein levels and lipid classes were also analyzed at 13 weeks of exposure. We observed that the TiO2 exposure increased the fraction of cells in G1-phase and reduced the fraction of cells in G2-phase, which was accompanied by an increase in the fraction of late apoptotic/necrotic cells. This corresponded with an induced expression of key apoptotic proteins i.e., BAD and BAX, and an accumulation of several lipid classes involved in cellular stress and apoptosis. These findings were further supported by quantitative proteome profiling data showing an increase in proteins involved in cell stress and genomic maintenance pathways following TiO2 exposure. Altogether, we suggest that cell stress response and cell death pathways may be important molecular events in long-term health effects of TiO2.
Collapse
Affiliation(s)
- Mayes Alswady-Hoff
- National Institute of Occupational Health, NO-0033 Oslo, Norway; (M.A.-H.); (J.S.E.); (S.P.); (Ø.S.)
| | - Johanna Samulin Erdem
- National Institute of Occupational Health, NO-0033 Oslo, Norway; (M.A.-H.); (J.S.E.); (S.P.); (Ø.S.)
| | - Santosh Phuyal
- National Institute of Occupational Health, NO-0033 Oslo, Norway; (M.A.-H.); (J.S.E.); (S.P.); (Ø.S.)
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, NO-0316 Oslo, Norway
| | | | - Animesh Sharma
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (A.S.); (D.d.M.F.); (G.S.)
- Proteomics and Metabolomics Core Facility (PROMEC), Norwegian University of Science and Technology and the Central Norway Regional Health Authority, NO-7491 Trondheim, Norway
| | - Davi de Miranda Fonseca
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (A.S.); (D.d.M.F.); (G.S.)
- Proteomics and Metabolomics Core Facility (PROMEC), Norwegian University of Science and Technology and the Central Norway Regional Health Authority, NO-7491 Trondheim, Norway
| | - Øivind Skare
- National Institute of Occupational Health, NO-0033 Oslo, Norway; (M.A.-H.); (J.S.E.); (S.P.); (Ø.S.)
| | - Geir Slupphaug
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, NO-7491 Trondheim, Norway; (A.S.); (D.d.M.F.); (G.S.)
- Proteomics and Metabolomics Core Facility (PROMEC), Norwegian University of Science and Technology and the Central Norway Regional Health Authority, NO-7491 Trondheim, Norway
| | - Shanbeh Zienolddiny
- National Institute of Occupational Health, NO-0033 Oslo, Norway; (M.A.-H.); (J.S.E.); (S.P.); (Ø.S.)
- Correspondence: ; Tel.: +47-23195284
| |
Collapse
|
8
|
Exploring the dermotoxicity of the mycotoxin deoxynivalenol: combined morphologic and proteomic profiling of human epidermal cells reveals alteration of lipid biosynthesis machinery and membrane structural integrity relevant for skin barrier function. Arch Toxicol 2021; 95:2201-2221. [PMID: 33890134 PMCID: PMC8166681 DOI: 10.1007/s00204-021-03042-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 04/01/2021] [Indexed: 12/26/2022]
Abstract
Deoxynivalenol (vomitoxin, DON) is a secondary metabolite produced by Fusarium spp. fungi and it is one of the most prevalent mycotoxins worldwide. Crop infestation results not only in food and feed contamination, but also in direct dermal exposure, especially during harvest and food processing. To investigate the potential dermotoxicity of DON, epidermoid squamous cell carcinoma cells A431 were compared to primary human neonatal keratinocytes (HEKn) cells via proteome/phosphoproteome profiling. In A431 cells, 10 µM DON significantly down-regulated ribosomal proteins, as well as mitochondrial respiratory chain elements (OXPHOS regulation) and transport proteins (TOMM22; TOMM40; TOMM70A). Mitochondrial impairment was reflected in altered metabolic competence, apparently combined with interference of the lipid biosynthesis machinery. Functional effects on the cell membrane were confirmed by live cell imaging and membrane fluidity assays (0.1–10 µM DON). Moreover, a common denominator for both A431 and HEKn cells was a significant downregulation of the squalene synthase (FDFT1). In sum, proteome alterations could be traced back to the transcription factor Klf4, a crucial regulator of skin barrier function. Overall, these results describe decisive molecular events sustaining the capability of DON to impair skin barrier function. Proteome data generated in the study are fully accessible via ProteomeXchange with the accession numbers PXD011474 and PXD013613.
Collapse
|
9
|
Han Q, Wang J, Luo H, Li L, Lu X, Liu A, Deng Y, Jiang Y. TMBIM6, a potential virus target protein identified by integrated multiomics data analysis in SARS-CoV-2-infected host cells. Aging (Albany NY) 2021; 13:9160-9185. [PMID: 33744846 PMCID: PMC8064151 DOI: 10.18632/aging.202718] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 02/16/2021] [Indexed: 12/21/2022]
Abstract
Coronavirus disease 2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). In this study, we collected open access data to analyze the mechanisms associated with SARS-CoV-2 infection. Gene set enrichment analysis (GSEA) revealed that apoptosis-related pathways were enriched in the cells after SARS-CoV-2 infection, and the results of differential expression analysis showed that biological functions related to endoplasmic reticulum stress (ERS) and lipid metabolism were disordered. TMBIM6 was identified as a potential target for SARS-CoV-2 in host cells through weighted gene coexpression network analysis (WGCNA) of the time course of expression of host and viral proteins. The expression and related functions of TMBIM6 were subsequently analyzed to illuminate how viral proteins interfere with the physiological function of host cells. The potential function of viral proteins was further analyzed by GEne Network Inference with Ensemble of trees (GENIE3). This study identified TMBIM6 as a target protein associated with the pathogenesis of SARS-CoV-2, which might provide a novel therapeutic approach for COVID-19 in the future.
Collapse
Affiliation(s)
- Qizheng Han
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Junhao Wang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Haihua Luo
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Lei Li
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Xinya Lu
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Aihua Liu
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yongqiang Deng
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| | - Yong Jiang
- Guangdong Provincial Key Laboratory of Proteomics, State Key Laboratory of Organ Failure Research, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
10
|
Drouin A, Wallbillich N, Theberge M, Liu S, Katz J, Bellovoda K, Se Yun Cheon S, Gootkind F, Bierman E, Zavras J, Berberich MJ, Kalocsay M, Guastaldi F, Salvadori N, Troulis M, Fusco DN. Impact of Zika virus on the human type I interferon osteoimmune response. Cytokine 2021; 137:155342. [PMID: 33130337 DOI: 10.1016/j.cyto.2020.155342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/25/2020] [Accepted: 10/08/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND The developing field of osteoimmunology supports importance of an interferon (IFN) response pathway in osteoblasts. Clarifying osteoblast-IFN interactions is important because IFN is used as salvage anti-tumor therapy but systemic toxicity is high with variable clinical results. In addition, osteoblast response to systemic bursts and disruptions of IFN pathways induced by viral infection may influence bone remodeling. ZIKA virus (ZIKV) infection impacts bone development in humans and IFN response in vitro. Consistently, initial evidence of permissivity to ZIKV has been reported in human osteoblasts. HYPOTHESIS Osteoblast-like Saos-2 cells are permissive to ZIKV and responsive to IFN. METHODS Multiple approaches were used to assess whether Saos-2 cells are permissive to ZIKV infection and exhibit IFN-mediated ZIKV suppression. Proteomic methods were used to evaluate impact of ZIKV and IFN on Saos-2 cells. RESULTS Evidence is presented confirming Saos-2 cells are permissive to ZIKV and support IFN-mediated suppression of ZIKV. ZIKV and IFN differentially impact the Saos-2 proteome, exemplified by HELZ2 protein which is upregulated by IFN but non responsive to ZIKV. Both ZIKV and IFN suppress proteins associated with microcephaly/pseudo-TORCH syndrome (BI1, KI20A and UBP18), and ZIKV induces potential entry factor PLVAP. CONCLUSIONS Transient ZIKV infection influences osteoimmune state, and IFN and ZIKV activate distinct proteomes in Saos-2 cells, which could inform therapeutic, engineered, disruptions.
Collapse
Affiliation(s)
- Arnaud Drouin
- Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70114, United States; Department of Pathology, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70114, United States
| | - Nicholas Wallbillich
- Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70114, United States
| | - Marc Theberge
- Tulane University, 6823 St Charles Ave, New Orleans, LA 70118, United States
| | - Sharon Liu
- Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70114, United States
| | - Joshua Katz
- Tulane University, 6823 St Charles Ave, New Orleans, LA 70118, United States
| | - Kamela Bellovoda
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, United States
| | - Scarlett Se Yun Cheon
- Department of Medicine, Massachusetts General Hospital, 55 Fruit Street, Boston, MA 02114, United States
| | - Frederick Gootkind
- Department of Oral & Maxillofacial Surgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, United States
| | - Emily Bierman
- Department of Oral & Maxillofacial Surgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, United States
| | - Jason Zavras
- Department of Oral & Maxillofacial Surgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, United States
| | - Matthew J Berberich
- Laboratory of Systems Pharmacology, Harvard Medical School, Armenise Building, 200 Longwood, Ave, Boston, MA 02115, United States
| | - Marian Kalocsay
- Laboratory of Systems Pharmacology, Harvard Medical School, Armenise Building, 200 Longwood, Ave, Boston, MA 02115, United States
| | - Fernando Guastaldi
- Department of Oral & Maxillofacial Surgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, United States
| | - Nicolas Salvadori
- Institut de recherche pour le développement (IRD)-PHPT, Marseille, France; Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Maria Troulis
- Department of Oral & Maxillofacial Surgery, Massachusetts General Hospital, 55 Fruit Street, Boston, MA, 02114, United States
| | - Dahlene N Fusco
- Department of Medicine, Tulane University School of Medicine, 1430 Tulane Avenue, New Orleans, LA, 70114, United States.
| |
Collapse
|
11
|
Verchot J, Pajerowska-Mukhtar KM. UPR signaling at the nexus of plant viral, bacterial, and fungal defenses. Curr Opin Virol 2020; 47:9-17. [PMID: 33360330 DOI: 10.1016/j.coviro.2020.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 11/13/2020] [Accepted: 11/15/2020] [Indexed: 12/24/2022]
Abstract
In recent years there have been significant advances in our understanding of the ER stress responses in plants that are associated with virus infection, as well as bacterial and fungal diseases. In plants, ER stress induced by virus infection includes several signaling pathways that include the unfolded protein response (UPR) to promote the expression of chaperone proteins for proper protein folding. Understanding how facets of ER stress signaling broadly engage in pathogen responses, as well as those that are specific to virus infection is important to distinguishing features essential for broad cellular defenses and processes that may be specifically linked to viral infectivity and disease.
Collapse
Affiliation(s)
- Jeanmarie Verchot
- Department of Plant Pathology and Microbiology, Texas A&M University, College Station, TX 77845, USA..
| | | |
Collapse
|
12
|
Vanka R, Nakka VP, Kumar SP, Baruah UK, Babu PP. Molecular targets in cerebral malaria for developing novel therapeutic strategies. Brain Res Bull 2020; 157:100-107. [PMID: 32006570 DOI: 10.1016/j.brainresbull.2020.01.020] [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: 01/23/2020] [Accepted: 01/27/2020] [Indexed: 10/25/2022]
Abstract
Cerebral malaria (CM) is the severe neurological complication associated with Plasmodium falciparum infection. In clinical settings CM is predominantly characterized by fever, epileptic seizures, and asexual forms of parasite on blood smears, coma and even death. Cognitive impairment in the children and adults even after survival is one of the striking consequences of CM. Poor diagnosis often leads to inappropriate malaria therapy which in turn progress into a severe form of disease. Activation of multiple cell death pathways such as Inflammation, oxidative stress, apoptosis and disruption of blood brain barrier (BBB) plays critical role in the pathogenesis of CM and secondary brain damage. Thus, understanding such mechanisms of neuronal cell death might help to identify potential molecular targets for CM. Mitigation strategies for mortality rate and long-term cognitive deficits caused by existing anti-malarial drugs still remains a valid research question to ask. In this review, we discuss in detail about critical neuronal cell death mechanisms and the overall significance of adjunctive therapy with recent trends, which provides better insight towards establishing newer therapeutic strategies for CM.
Collapse
Affiliation(s)
- Ravisankar Vanka
- Department of Pharmaceutics, Aditya Pharmacy College, Suramaplem, Gandepalli Mandal, East Godavari, Andhra Pradesh, 533437, India
| | - Venkata Prasuja Nakka
- Department of Biochemistry, Acharya Nagarjuna University, Nagarjuna Nagar, Guntur, Andhra Pradesh, 522510, India
| | - Simhadri Praveen Kumar
- Department of Biotechnology and Bioinformatics, School of life Sciences, University of Hyderabad, Hyderabad, Telangana, 500046, India
| | - Uday Krishna Baruah
- Department of Pharmaceutics, JSS College of Pharmacy, Ooty, Tamil Nadu 643001, India
| | - Phanithi Prakash Babu
- Department of Biotechnology and Bioinformatics, School of life Sciences, University of Hyderabad, Hyderabad, 500046, Telangana, India.
| |
Collapse
|
13
|
Lebeaupin C, Blanc M, Vallée D, Keller H, Bailly-Maitre B. BAX inhibitor-1: between stress and survival. FEBS J 2020; 287:1722-1736. [PMID: 31841271 DOI: 10.1111/febs.15179] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/18/2019] [Accepted: 12/13/2019] [Indexed: 12/13/2022]
Abstract
Cellular gatekeepers are essential to maintain order within a cell and anticipate signals of stress to promote survival. BCL2 associated X, apoptosis regulator (BAX) inhibitor-1 (BI-1), also named transmembrane BAX inhibitor motif containing-6, is a highly conserved endoplasmic reticulum (ER) transmembrane protein. Originally identified as an inhibitor of BAX-induced apoptosis, its pro-survival properties have been expanded to include functions targeted against ER stress, calcium imbalance, reactive oxygen species accumulation, and metabolic dysregulation. Nevertheless, the structural biology and biochemical mechanism of action of BI-1 are still under debate. BI-1 has been implicated in several diseases, including chronic liver disease, diabetes, ischemia/reperfusion injury, neurodegeneration, and cancer. While most studies have demonstrated a beneficial role for BI-1 in the ubiquitous maintenance of cellular homeostasis, its expression in cancer cells seems most often to contribute to tumorigenesis and metastasis. Here, we summarize what is known about BI-1 and encourage future studies on BI-1's contribution to cellular life and death decisions to advocate its potential as a target for drug development and other therapeutic strategies.
Collapse
Affiliation(s)
- Cynthia Lebeaupin
- INSERM U1065, C3M, Université Côte d'Azur, Nice, France.,Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Marina Blanc
- INSERM U1065, C3M, Université Côte d'Azur, Nice, France
| | | | - Harald Keller
- INRA1355-CNRS7254, Université Côte d'Azur, Sophia Antipolis, France
| | | |
Collapse
|
14
|
Arc silence aggravates traumatic neuronal injury via mGluR1-mediated ER stress and necroptosis. Cell Death Dis 2020; 11:4. [PMID: 31919348 PMCID: PMC6952410 DOI: 10.1038/s41419-019-2198-5] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 12/07/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022]
Abstract
Delayed neuronal death is associated with neurological deficits and mortality after traumatic brain injury (TBI), where post-synaptic density (PSD) proteins are thought to play key roles. The immediate-early gene (IEG) coded protein Arc is a brain-specific PSD protein that controls synaptic plasticity and learning behaviors. In this study, we investigated the expression and biological function of Arc in neuronal death after TBI in an in vitro model mimicked by traumatic neuronal injury (TNI) in cortical neurons. TNI caused a temporal increase of Arc expression at 3 and 6 h. Knockdown of Arc expression using small interfering RNA (Si-Arc-3) promoted TNI-induced cytotoxicity and apoptosis. The results of western blot showed that Si-Arc-3 transfection further enhanced the activation of endoplasmic reticulum (ER) stress-associated factors, including glucose-regulated protein 78 (GRP78), C/EBP homologous protein (CHOP) and caspase-12 after TNI. In addition, knockdown of Arc significantly increased expression of (receptor-interacting protein kinase 1) RIP1 and the number of necroptotic cells, which were apparently prevented by necrostatin-1 (Nec-1). The results of immunostaining and western blot showed that knockdown of Arc activated the metabotropic glutamate receptor 1 (mGluR1) and intracellular Ca2+ release in neurons. Mechanistically, the Si-Arc-3-induced activation of ER stress-associated factors, RIP1 expression, apoptosis, and necroptosis were partially reversed by the mGluR1 antagonist AIDA. In summary, our data suggest that silence of Arc expression aggravates neuronal death after TNI by promoting apoptosis and necroptosis. These data support for the first time that Arc may represent a novel candidate for therapies against TBI.
Collapse
|
15
|
Doycheva D, Xu N, Kaur H, Malaguit J, McBride DW, Tang J, Zhang JH. Adenoviral TMBIM6 vector attenuates ER-stress-induced apoptosis in a neonatal hypoxic-ischemic rat model. Dis Model Mech 2019; 12:dmm040352. [PMID: 31636086 PMCID: PMC6898997 DOI: 10.1242/dmm.040352] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 10/14/2019] [Indexed: 12/31/2022] Open
Abstract
Endoplasmic reticulum (ER) stress is a major pathology encountered after hypoxic-ischemic (HI) injury. Accumulation of unfolded proteins triggers the unfolded protein response (UPR), resulting in the activation of pro-apoptotic cascades that lead to cell death. Here, we identified Bax inhibitor 1 (BI-1), an evolutionarily conserved protein encoded by the transmembrane BAX inhibitor motif-containing 6 (TMBIM6) gene, as a novel modulator of ER-stress-induced apoptosis after HI brain injury in a neonatal rat pup. The main objective of our study was to overexpress BI-1, via viral-mediated gene delivery of human adenoviral-TMBIM6 (Ad-TMBIM6) vector, to investigate its anti-apoptotic effects as well as to elucidate its signaling pathways in an in vivo neonatal HI rat model and in vitro oxygen-glucose deprivation (OGD) model. Ten-day-old unsexed Sprague Dawley rat pups underwent right common carotid artery ligation followed by 1.5 h of hypoxia. Rat pups injected with Ad-TMBIM6 vector, 48 h pre-HI, showed a reduction in relative infarcted area size, attenuated neuronal degeneration and improved long-term neurological outcomes. Furthermore, silencing of BI-1 or further activating the IRE1α branch of the UPR, using a CRISPR activation plasmid, was shown to reverse the protective effects of BI-1. Based on our in vivo and in vitro data, the protective effects of BI-1 are mediated via inhibition of IRE1α signaling and in part via inhibition of the second stress sensor receptor, PERK. Overall, this study showed a novel role for BI-1 and ER stress in the pathophysiology of HI and could provide a basis for BI-1 as a potential therapeutic target.
Collapse
Affiliation(s)
- Desislava Doycheva
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Ningbo Xu
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
- Department of Interventional Radiology, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Harpreet Kaur
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Jay Malaguit
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - Devin William McBride
- The Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Jiping Tang
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| | - John H Zhang
- Center for Neuroscience Research, Department of Physiology and Pharmacology, Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
- Departments of Anesthesiology, Neurosurgery and Neurology, Loma Linda University School of Medicine, Loma Linda, CA 92354, USA
| |
Collapse
|
16
|
Kumar V, Mesentier-Louro LA, Oh AJ, Heng K, Shariati MA, Huang H, Hu Y, Liao YJ. Increased ER Stress After Experimental Ischemic Optic Neuropathy and Improved RGC and Oligodendrocyte Survival After Treatment With Chemical Chaperon. Invest Ophthalmol Vis Sci 2019; 60:1953-1966. [PMID: 31060051 PMCID: PMC6735778 DOI: 10.1167/iovs.18-24890] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Purpose Increased endoplasmic reticulum (ER) stress is one of the earliest subcellular changes in neuro-ophthalmic diseases. In this study, we investigated the expression of key molecules in the ER stress pathways following nonarteritic anterior ischemic optic neuropathy (AION), the most common acute optic neuropathy in adults over 50, and assessed the impact of chemical chaperon 4-phenylbutyric acid (4-PBA) in vivo. Methods We induced AION using photochemical thrombosis in adult mice and performed histologic analyses of key molecules in the ER stress pathway in the retina and optic nerve. We also assessed the effects of daily intraperitoneal injections of 4-PBA after AION. Results In the retina at baseline, there was low proapoptotic transcriptional regulator C/EBP homologous protein (CHOP) and high prosurvival chaperon glucose-regulated protein 78 (GRP78) expression in retinal ganglion cells (RGCs). One day after AION, there was significantly increased CHOP and reduced GRP78 expressions in the ganglion cell layer. In the optic nerve at baseline, there was little CHOP and high GRP78 expression. One day after AION, there was significantly increased CHOP and no change in GRP78 expression. Treatment immediately after AION using daily intraperitoneal injection of chemical chaperone 4-PBA for 19 days significantly rescued Brn3A+ RGCs and Olig2+ optic nerve oligodendrocytes. Conclusions We showed for the first time that acute AION resulted in increased ER stress and differential expression of ER stress markers CHOP and GRP78 in the retina and optic nerve. Rescue of RGCs and oligodendrocytes with 4-PBA provides support for ER stress reduction as possible treatment for AION.
Collapse
Affiliation(s)
- Varun Kumar
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States
| | | | - Angela Jinsook Oh
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States
| | - Kathleen Heng
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States
| | - Mohammad Ali Shariati
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States
| | - Haoliang Huang
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States
| | - Yang Hu
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States
| | - Yaping Joyce Liao
- Department of Ophthalmology, Stanford University, School of Medicine, Stanford, California, United States.,Department of Neurology, Stanford University, School of Medicine, Stanford, California, United States
| |
Collapse
|
17
|
Guo G, Xu M, Chang Y, Luyten T, Seitaj B, Liu W, Zhu P, Bultynck G, Shi L, Quick M, Liu Q. Ion and pH Sensitivity of a TMBIM Ca 2+ Channel. Structure 2019; 27:1013-1021.e3. [PMID: 30930064 PMCID: PMC6560632 DOI: 10.1016/j.str.2019.03.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/25/2019] [Accepted: 02/28/2019] [Indexed: 12/12/2022]
Abstract
The anti-apoptotic transmembrane Bax inhibitor motif (TMBIM) containing protein family regulates Ca2+ homeostasis, cell death, and the progression of diseases including cancers. The recent crystal structures of the TMBIM homolog BsYetJ reveal a conserved Asp171-Asp195 dyad that is proposed in regulating a pH-dependent Ca2+ translocation. Here we show that BsYetJ mediates Ca2+ fluxes in permeabilized mammalian cells, and its interaction with Ca2+ is sensitive to protons and other cations. We report crystal structures of BsYetJ in additional states, revealing the flexibility of the dyad in a closed state and a pore-opening mechanism. Functional studies show that the dyad is responsible for both Ca2+ affinity and pH dependence. Computational simulations suggest that protonation of Asp171 weakens its interaction with Arg60, leading to an open state. Our integrated analysis provides insights into the regulation of the BsYetJ Ca2+ channel that may inform understanding of human TMBIM proteins regarding their roles in cell death and diseases.
Collapse
Affiliation(s)
- Gongrui Guo
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973, USA; NSLS-II, Brookhaven National Laboratory, Upton, NY 11973, USA
| | - Min Xu
- Computational Chemistry and Molecular Biophysics Unit, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MA 21224, USA
| | - Yanqi Chang
- New York Consortium on Membrane Protein Structure, New York Structural Biology Center, New York, NY 10027, USA
| | - Tomas Luyten
- Laboratory Molecular and Cellular Signaling, Department Cellular and Molecular Medicine, Campus Gasthuisberg O/N-I Bus 802, Herestraat 49 3000 Leuven, Belgium
| | - Bruno Seitaj
- Laboratory Molecular and Cellular Signaling, Department Cellular and Molecular Medicine, Campus Gasthuisberg O/N-I Bus 802, Herestraat 49 3000 Leuven, Belgium
| | - Wu Liu
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973, USA; NSLS-II, Brookhaven National Laboratory, Upton, NY 11973, USA
| | - Ping Zhu
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973, USA
| | - Geert Bultynck
- Laboratory Molecular and Cellular Signaling, Department Cellular and Molecular Medicine, Campus Gasthuisberg O/N-I Bus 802, Herestraat 49 3000 Leuven, Belgium
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Unit, National Institute on Drug Abuse - Intramural Research Program, National Institutes of Health, Baltimore, MA 21224, USA.
| | - Matthias Quick
- Department of Psychiatry, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Division of Molecular Therapeutics, New York State Psychiatric Institute, New York, NY 10032, USA.
| | - Qun Liu
- Biology Department, Brookhaven National Laboratory, Upton, NY 11973, USA; NSLS-II, Brookhaven National Laboratory, Upton, NY 11973, USA.
| |
Collapse
|
18
|
Zhou J, Song J, Wu S. Autophagic degradation of stromal interaction molecule 2 mediates disruption of neuronal dendrites by endoplasmic reticulum stress. J Neurochem 2019; 151:351-369. [PMID: 31038732 DOI: 10.1111/jnc.14712] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Revised: 03/07/2019] [Accepted: 04/26/2019] [Indexed: 10/26/2022]
Abstract
Endoplasmic reticulum (ER) stress has been highlighted as one of the factors involved in axon/dendrite degeneration, which is an early event in Alzheimer's, Parkinson's diseases as well as in acute disorders such as ischemia and axotomy-induced retinal ganglion cell degeneration. These lines of evidence suggest critical roles of ER stress at the early stage of neurodegeneration, but the relevant mechanism is rarely exploited. In this study, we report that treatment with sublethal level of ER stressors, tunicamycin or brefeldin A, in primary rat neuronal cultures, significantly reduced dendrite arbor. Under the same treatment, either stressor reduced store-operated calcium entry (SOCE) and cytosolic calcium, [Ca2+ ]i , which were associated with autophagic degradation of stromal interaction molecule 2 (STIM2). Knockdown of ATG7 or activating transcription factor 4 completely reversed the reduction of STIM2 and significantly reversed the inhibition of SOCE under ER stress. Overexpression of STIM2 in neurons significantly prevented the ER stress-induced disruption of dendrite arbor. Altogether, our data reveal an unprecedented mechanism by which ER stress induces dendrite degeneration, that is, ER stress induces autophagic degradation of STIM2, leading to ensued SOCE inhibition and reduced [Ca2+ ]i , resulting in trimming effect on dendrites.
Collapse
Affiliation(s)
- Jing Zhou
- School of Optometry and Ophthalmology and the Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China.,State Key Laboratory of Ophthalmology, Optometry, and Visual Science, Wenzhou, Zhejiang, P.R. China
| | - Juan Song
- School of Optometry and Ophthalmology and the Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China.,State Key Laboratory of Ophthalmology, Optometry, and Visual Science, Wenzhou, Zhejiang, P.R. China
| | - Shengzhou Wu
- School of Optometry and Ophthalmology and the Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China.,State Key Laboratory of Ophthalmology, Optometry, and Visual Science, Wenzhou, Zhejiang, P.R. China
| |
Collapse
|
19
|
Doycheva D, Kaur H, Tang J, Zhang JH. The characteristics of the ancient cell death suppressor, TMBIM6, and its related signaling pathways after endoplasmic reticulum stress. J Neurosci Res 2019; 98:77-86. [PMID: 31044452 DOI: 10.1002/jnr.24434] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Revised: 03/27/2019] [Accepted: 04/10/2019] [Indexed: 12/27/2022]
Abstract
Activation of the unfolded protein response in combination with generation of reactive oxygen species, from cytochrome P450 members and NADPH-P450 reductases, are two major consequences of Endoplasmic Reticulum (ER) stress that cause oxidative damage and cell death. Herein, we reviewed the role of Bax Inhibitor-1 (BI-1), an evolutionarily conserved protein encoded by the Transmembrane Bax inhibitor Motif Containing 6 gene, in protection from ER stress. As BI-1 has multimodal properties that can target a wide array of pathophysiological consequences after injury, our main objective was to explore BI-1's protective role in ER stress and its potential signaling pathways.
Collapse
Affiliation(s)
- Desislava Doycheva
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California
| | - Harpreet Kaur
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California
| | - Jiping Tang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California
| | - John H Zhang
- Department of Physiology and Pharmacology, Basic Sciences, School of Medicine, Loma Linda University, Loma Linda, California.,Department of Anesthesiology, School of Medicine, Loma Linda University, Loma Linda, California.,Department of Neurosurgery, School of Medicine, Loma Linda University, Loma Linda, California.,Department of Neurology, School of Medicine, Loma Linda University, Loma Linda, California
| |
Collapse
|
20
|
Glotfelty EJ, Delgado TE, Tovar-y-Romo LB, Luo Y, Hoffer BJ, Olson L, Karlsson TE, Mattson MP, Harvey BK, Tweedie D, Li Y, Greig NH. Incretin Mimetics as Rational Candidates for the Treatment of Traumatic Brain Injury. ACS Pharmacol Transl Sci 2019; 2:66-91. [PMID: 31396586 PMCID: PMC6687335 DOI: 10.1021/acsptsci.9b00003] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Indexed: 12/17/2022]
Abstract
Traumatic brain injury (TBI) is becoming an increasing public health issue. With an annually estimated 1.7 million TBIs in the United States (U.S) and nearly 70 million worldwide, the injury, isolated or compounded with others, is a major cause of short- and long-term disability and mortality. This, along with no specific treatment, has made exploration of TBI therapies a priority of the health system. Age and sex differences create a spectrum of vulnerability to TBI, with highest prevalence among younger and older populations. Increased public interest in the long-term effects and prevention of TBI have recently reached peaks, with media attention bringing heightened awareness to sport and war related head injuries. Along with short-term issues, TBI can increase the likelihood for development of long-term neurodegenerative disorders. A growing body of literature supports the use of glucagon-like peptide-1 (GLP-1), glucose-dependent insulinotropic peptide (GIP), and glucagon (Gcg) receptor (R) agonists, along with unimolecular combinations of these therapies, for their potent neurotrophic/neuroprotective activities across a variety of cellular and animal models of chronic neurodegenerative diseases (Alzheimer's and Parkinson's diseases) and acute cerebrovascular disorders (stroke). Mild or moderate TBI shares many of the hallmarks of these conditions; recent work provides evidence that use of these compounds is an effective strategy for its treatment. Safety and efficacy of many incretin-based therapies (GLP-1 and GIP) have been demonstrated in humans for the treatment of type 2 diabetes mellitus (T2DM), making these compounds ideal for rapid evaluation in clinical trials of mild and moderate TBI.
Collapse
Affiliation(s)
- Elliot J. Glotfelty
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
- Department
of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | - Thomas E. Delgado
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Luis B. Tovar-y-Romo
- Division
of Neuroscience, Institute of Cellular Physiology, Universidad Nacional Autónoma de México, Mexico City, Mexico
| | - Yu Luo
- Department
of Molecular Genetics, University of Cincinnati, Cincinnati, Ohio 45221, United States
| | - Barry J. Hoffer
- Department
of Neurosurgery, Case Western Reserve University
School of Medicine, Cleveland, Ohio 44106, United States
| | - Lars Olson
- Department
of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | | - Mark P. Mattson
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Brandon K. Harvey
- Molecular
Mechanisms of Cellular Stress and Inflammation Unit, Integrative Neuroscience
Department, National Institute on Drug Abuse,
National Institutes of Health, Baltimore, Maryland 21224, United States
| | - David Tweedie
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Yazhou Li
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| | - Nigel H. Greig
- Translational
Gerontology Branch, and Laboratory of Neurosciences, Intramural
Research Program, National Institute on
Aging, National Institutes of Health, Baltimore, Maryland 21224, United States
| |
Collapse
|
21
|
Curcumin mitigates axonal injury and neuronal cell apoptosis through the PERK/Nrf2 signaling pathway following diffuse axonal injury. Neuroreport 2019; 29:661-677. [PMID: 29570500 PMCID: PMC5959262 DOI: 10.1097/wnr.0000000000001015] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Diffuse axonal injury (DAI) accounts for more than 50% of all traumatic brain injury. In response to the mechanical damage associated with DAI, the abnormal proteins produced in the neurons and axons, namely, β-APP and p-tau, induce endoplasmic reticulum (ER) stress. Curcumin, a major component extracted from the rhizome of Curcuma longa, has shown potent anti-inflammatory, antioxidant, anti-infection, and antitumor activity in previous studies. Moreover, curcumin is an activator of nuclear factor-erythroid 2-related factor 2 (Nrf2) and promotes its nuclear translocation. In this study, we evaluated the therapeutic potential of curcumin for the treatment of DAI and investigated the mechanisms underlying the protective effects of curcumin against neural cell death and axonal injury after DAI. Rats subjected to a model of DAI by head rotational acceleration were treated with vehicle or curcumin to evaluate the effect of curcumin on neuronal and axonal injury. We observed that curcumin (20 mg/kg intraperitoneal) administered 1 h after DAI induction alleviated the aggregation of p-tau and β-APP in neurons, reduced ER-stress-related cell apoptosis, and ameliorated neurological deficits. Further investigation showed that the protective effect of curcumin in DAI was mediated by the PERK/Nrf2 pathway. Curcumin promoted PERK phosphorylation, and then Nrf2 dissociated from Keap1 and was translocated to the nucleus, which activated ATF4, an important bZIP transcription factor that maintains intracellular homeostasis, but inhibited the CHOP, a hallmark of ER stress and ER-associated programmed cell death. In summary, we demonstrate for the first time that curcumin confers protection against abnormal proteins and neuronal apoptosis after DAI, that the process is mediated by strengthening of the unfolded protein response to overcome ER stress, and that the protective effect of curcumin against DAI is dependent on the activation of Nrf2.
Collapse
|
22
|
Abstract
The brain undergoes several changes at structural, molecular, and cellular levels leading to alteration in its functions and these processes are primarily maintained by proteostasis in cells. However, an imbalance in proteostasis due to the abnormal accumulation of protein aggregates induces endoplasmic reticulum (ER) stress. This event, in turn, activate the unfolded protein response; however, in most neurodegenerative conditions and brain injury, an uncontrolled unfolded protein response elicits memory dysfunction. Although the underlying signaling mechanism for impairment of memory function following induction of ER stress remains elusive, recent studies have highlighted that inactivation of a transcription factor, CREB, which is essential for synaptic function and memory formation, plays an essential role for ER stress-induced synaptic and memory dysfunction. In this review, current studies and most updated view on how ER stress affects memory function in both physiological and pathological conditions will be highlighted.
Collapse
Affiliation(s)
- Nilkantha Sen
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
23
|
Liu J, Zhou S, Zhang Y, Li X, Qian X, Tao W, Jin L, Zhao J. Bax inhibitor-1 suppresses early brain injury following experimental subarachnoid hemorrhage in rats. Int J Mol Med 2018; 42:2891-2902. [PMID: 30226536 DOI: 10.3892/ijmm.2018.3858] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Accepted: 09/04/2018] [Indexed: 11/05/2022] Open
Abstract
Early brain injury (EBI) following subarachnoid hemorrhage (SAH) is an important cause of high mortality and poor prognosis in SAH. B‑cell lymphoma 2‑associated X protein inhibitor‑1 (BI‑1) is an evolutionarily conserved antiapoptotic protein that is primarily located in the membranes of endoplasmic reticulum (ER). BI‑1 has been studied in certain nervous system‑associated diseases, but the role of this protein in SAH remains unclear. In the present study, the role of BI‑1 in EBI following SAH was investigated in rat models and its associated mechanisms were examined. The SAH rat model was generated by inserting nylon cords into the internal carotid artery from the external carotid artery. Samples were assessed using neurological scores, brain water content measurements, hematoxylin and eosin (H&E) staining, blood‑brain barrier (BBB) permeability, terminal deoxynucleotidyl transferase‑mediated dUTP nick‑end labeling and quantitative polymerase chain reaction assays, and western blot analyses. It was identified that the mRNA and protein levels of BI‑1 decreased markedly and were lowest at 24 h after SAH. BI‑1 overexpression and small hairpin RNA (shRNA)‑mediated silencing markedly suppressed or severely exacerbated EBI following SAH, respectively. BI‑1 overexpression in the SAH model improved neurological scores and decreased the brain water content, BBB permeability and levels of apoptosis compared with the control and sham groups following SAH. BI‑1 shRNA in the SAH model demonstrated contrary results. In addition, the mRNA or protein expression levels of ER stress‑associated genes (glucose regulated protein, 78 kDa, C/EBP homologous protein, Serine/threonine‑protein kinase/endoribonuclease IRE1, c‑Jun N terminal kinases and apoptotic signaling kinase‑1) were markedly suppressed or increased following BI‑1 overexpression and shRNA‑mediated silencing, respectively. The present study suggested that BI‑1 serves a neuroprotective role in EBI following SAH by attenuating BBB disruption, brain edema and apoptosis mediated by ER stress.
Collapse
Affiliation(s)
- Jiaxin Liu
- Department of Environmental Science, Faculty of Environmental Science and Engineering, Kunming University of Science and Technology, Kunming, Yunnan 650504, P.R. China
| | - Shuai Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Yueting Zhang
- Very Important Person Ward, The Second Affiliated Hospital of Kunming Medical College, Kunming, Yunnan 650032, P.R. China
| | - Xiuying Li
- Department of Pharmacology, Medical School, Kunming University of Science and Technology, Kunming, Yunnan 650504, P.R. China
| | - Xiying Qian
- Department of Neurosurgery, The First Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Weihua Tao
- Department of Neurosurgery, The First Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Lide Jin
- Department of Neurosurgery, The First Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| | - Jianhua Zhao
- Department of Neurosurgery, The First Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650032, P.R. China
| |
Collapse
|
24
|
Shi L, Wang Z, Liu X, Li M, Zhang S, Song X. Bax inhibitor-1 is required for resisting the Early Brain Injury induced by subarachnoid hemorrhage through regulating IRE1-JNK pathway. Neurol Res 2018; 40:189-196. [PMID: 29334839 DOI: 10.1080/01616412.2018.1424699] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Lei Shi
- Department of Neurosurgery, Zhumadian Central Hospital, Zhumadian, China
| | - Zaizong Wang
- Department of Neurosurgery, Zhumadian Central Hospital, Zhumadian, China
| | - Xianjin Liu
- Department of Neurosurgery, Zhumadian Central Hospital, Zhumadian, China
| | - Mao Li
- Department of Neurosurgery, Zhumadian Central Hospital, Zhumadian, China
| | - Shangfei Zhang
- Department of Neurosurgery, Zhumadian Central Hospital, Zhumadian, China
| | - Xiaobin Song
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical College, Kunming, China
| |
Collapse
|
25
|
Kulbe JR, Hall ED. Chronic traumatic encephalopathy-integration of canonical traumatic brain injury secondary injury mechanisms with tau pathology. Prog Neurobiol 2017; 158:15-44. [PMID: 28851546 PMCID: PMC5671903 DOI: 10.1016/j.pneurobio.2017.08.003] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Revised: 08/09/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022]
Abstract
In recent years, a new neurodegenerative tauopathy labeled Chronic Traumatic Encephalopathy (CTE), has been identified that is believed to be primarily a sequela of repeated mild traumatic brain injury (TBI), often referred to as concussion, that occurs in athletes participating in contact sports (e.g. boxing, American football, Australian football, rugby, soccer, ice hockey) or in military combatants, especially after blast-induced injuries. Since the identification of CTE, and its neuropathological finding of deposits of hyperphosphorylated tau protein, mechanistic attention has been on lumping the disorder together with various other non-traumatic neurodegenerative tauopathies. Indeed, brains from suspected CTE cases that have come to autopsy have been confirmed to have deposits of hyperphosphorylated tau in locations that make its anatomical distribution distinct for other tauopathies. The fact that these individuals experienced repetitive TBI episodes during their athletic or military careers suggests that the secondary injury mechanisms that have been extensively characterized in acute TBI preclinical models, and in TBI patients, including glutamate excitotoxicity, intracellular calcium overload, mitochondrial dysfunction, free radical-induced oxidative damage and neuroinflammation, may contribute to the brain damage associated with CTE. Thus, the current review begins with an in depth analysis of what is known about the tau protein and its functions and dysfunctions followed by a discussion of the major TBI secondary injury mechanisms, and how the latter have been shown to contribute to tau pathology. The value of this review is that it might lead to improved neuroprotective strategies for either prophylactically attenuating the development of CTE or slowing its progression.
Collapse
Affiliation(s)
- Jacqueline R Kulbe
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States
| | - Edward D Hall
- Spinal Cord & Brain Injury Research Center, University of Kentucky College of Medicine, United States; Department of Neuroscience, University of Kentucky College of Medicine, United States.
| |
Collapse
|
26
|
Huang H, Miao L, Liang F, Liu X, Xu L, Teng X, Wang Q, Ridder WH, Shindler KS, Sun Y, Hu Y. Neuroprotection by eIF2α-CHOP inhibition and XBP-1 activation in EAE/optic neuritiss. Cell Death Dis 2017; 8:e2936. [PMID: 28726788 PMCID: PMC5550873 DOI: 10.1038/cddis.2017.329] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/05/2017] [Accepted: 06/13/2017] [Indexed: 12/22/2022]
Abstract
No therapies exist to prevent neuronal deficits in multiple sclerosis (MS), because the molecular mechanism responsible for the progressive neurodegeneration is unknown. We previously showed that axon injury-induced neuronal endoplasmic reticulum (ER) stress plays an important role in retinal ganglion cell (RGC) death and optic nerve degeneration in traumatic and glaucomatous optic neuropathies. Optic neuritis, one of the most common clinical manifestations of MS, is readily modeled by experimental autoimmune encephalomyelitis (EAE) in mouse. Using this in vivo model, we now show that ER stress is induced early in EAE and that modulation of ER stress by inhibition of eIF2α-CHOP and activation of XBP-1 in RGC specifically, protects RGC somata and axons and preserves visual function. This finding adds to the evidence that ER stress is a general upstream mechanism for neurodegeneration and suggests that targeting ER stress molecules is a promising therapeutic strategy for neuroprotection in MS.
Collapse
Affiliation(s)
- Haoliang Huang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto CA 94304, USA
| | - Linqing Miao
- Shriners Center for Neural Repair and Rehabilitation, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Feisi Liang
- Shriners Center for Neural Repair and Rehabilitation, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Xiaodong Liu
- Shriners Center for Neural Repair and Rehabilitation, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Lin Xu
- Shriners Center for Neural Repair and Rehabilitation, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Xiuyin Teng
- Shriners Center for Neural Repair and Rehabilitation, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | - Qizhao Wang
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto CA 94304, USA
| | - William H Ridder
- Southern California College of Optometry, Marshall B. Ketchum University, Fullerton, CA 92831, USA
| | - Kenneth S Shindler
- Scheie Eye Institute and F.M. Kirby Center for Molecular Ophthalmology, Departments of Ophthalmology and Neurology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yang Sun
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto CA 94304, USA
| | - Yang Hu
- Department of Ophthalmology, Stanford University School of Medicine, Palo Alto CA 94304, USA
| |
Collapse
|
27
|
M'Angale PG, Staveley BE. Bax-inhibitor-1 knockdown phenotypes are suppressed by Buffy and exacerbate degeneration in a Drosophila model of Parkinson disease. PeerJ 2017; 5:e2974. [PMID: 28243526 PMCID: PMC5322759 DOI: 10.7717/peerj.2974] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Accepted: 01/10/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Bax inhibitor-1 (BI-1) is an evolutionarily conserved cytoprotective transmembrane protein that acts as a suppressor of Bax-induced apoptosis by regulation of endoplasmic reticulum stress-induced cell death. We knocked down BI-1 in the sensitive dopa decarboxylase (Ddc) expressing neurons of Drosophila melanogaster to investigate its neuroprotective functions. We additionally sought to rescue the BI-1-induced phenotypes by co-expression with the pro-survival Buffy and determined the effect of BI-1 knockdown on the neurodegenerative α-synuclein-induced Parkinson disease (PD) model. METHODS We used organismal assays to assess longevity of the flies to determine the effect of the altered expression of BI-1 in the Ddc-Gal4-expressing neurons by employing two RNAi transgenic fly lines. We measured the locomotor ability of these RNAi lines by computing the climbing indices of the climbing ability and compared them to a control line that expresses the lacZ transgene. Finally, we performed biometric analysis of the developing eye, where we counted the number of ommatidia and calculated the area of ommatidial disruption. RESULTS The knockdown of BI-1 in these neurons was achieved under the direction of the Ddc-Gal4 transgene and resulted in shortened lifespan and precocious loss of locomotor ability. The co-expression of Buffy, the Drosophila anti-apoptotic Bcl-2 homologue, with BI-1-RNAi resulted in suppression of the reduced lifespan and impaired climbing ability. Expression of human α-synuclein in Drosophila dopaminergic neurons results in neuronal degeneration, accompanied by the age-dependent loss in climbing ability. We exploited this neurotoxic system to investigate possible BI-1 neuroprotective function. The co-expression of α-synuclein with BI-1-RNAi results in a slight decrease in lifespan coupled with an impairment in climbing ability. In supportive experiments, we employed the neuron-rich Drosophila compound eye to investigate subtle phenotypes that result from altered gene expression. The knockdown of BI-1 in the Drosophila developing eye under the direction of the GMR-Gal4 transgene results in reduced ommatidia number and increased disruption of the ommatidial array. Similarly, the co-expression of BI-1-RNAi with Buffy results in the suppression of the eye phenotypes. The expression of α-synuclein along with the knockdown of BI-1 resulted in reduction of ommatidia number and more disruption of the ommatidial array. CONCLUSION Knockdown of BI-1 in the dopaminergic neurons of Drosophila results in a shortened lifespan and premature loss in climbing ability, phenotypes that appear to be strongly associated with models of PD in Drosophila, and which are suppressed upon overexpression of Buffy and worsened by co-expression with α-synuclein. This suggests that BI-1 is neuroprotective and its knockdown can be counteracted by the overexpression of the pro-survival Bcl-2 homologue.
Collapse
Affiliation(s)
- P Githure M'Angale
- Department of Biology, Memorial University of Newfoundland , St. John's, NL , Canada
| | - Brian E Staveley
- Department of Biology, Memorial University of Newfoundland , St. John's, NL , Canada
| |
Collapse
|
28
|
Mollereau B, Rzechorzek NM, Roussel BD, Sedru M, Van den Brink DM, Bailly-Maitre B, Palladino F, Medinas DB, Domingos PM, Hunot S, Chandran S, Birman S, Baron T, Vivien D, Duarte CB, Ryoo HD, Steller H, Urano F, Chevet E, Kroemer G, Ciechanover A, Calabrese EJ, Kaufman RJ, Hetz C. Adaptive preconditioning in neurological diseases - therapeutic insights from proteostatic perturbations. Brain Res 2016; 1648:603-616. [PMID: 26923166 PMCID: PMC5010532 DOI: 10.1016/j.brainres.2016.02.033] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 02/16/2016] [Indexed: 02/06/2023]
Abstract
In neurological disorders, both acute and chronic neural stress can disrupt cellular proteostasis, resulting in the generation of pathological protein. However in most cases, neurons adapt to these proteostatic perturbations by activating a range of cellular protective and repair responses, thus maintaining cell function. These interconnected adaptive mechanisms comprise a 'proteostasis network' and include the unfolded protein response, the ubiquitin proteasome system and autophagy. Interestingly, several recent studies have shown that these adaptive responses can be stimulated by preconditioning treatments, which confer resistance to a subsequent toxic challenge - the phenomenon known as hormesis. In this review we discuss the impact of adaptive stress responses stimulated in diverse human neuropathologies including Parkinson׳s disease, Wolfram syndrome, brain ischemia, and brain cancer. Further, we examine how these responses and the molecular pathways they recruit might be exploited for therapeutic gain. This article is part of a Special Issue entitled SI:ER stress.
Collapse
Affiliation(s)
- B Mollereau
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France.
| | - N M Rzechorzek
- Centre for Clinical Brain Sciences, Chancellor's Building, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom; Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Roslin, Midlothian EH25 9RG, United Kingdom
| | - B D Roussel
- Inserm, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, 14000 Caen, France
| | - M Sedru
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France
| | - D M Van den Brink
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France
| | - B Bailly-Maitre
- INSERM U1065, C3M, Team 8 (Hepatic Complications in Obesity), Nice, France
| | - F Palladino
- Univ Lyon, ENS de Lyon, Univ Claude Bernard, CNRS UMR5239, INSERM U1210, Laboratory of Biology and Modelling of the Cell, F-69007, Lyon, France
| | - D B Medinas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Molecular Studies of the Cell, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Faculty of Medicine, University of Chile, Santiago, Chile
| | - P M Domingos
- ITQB-UNL, Av. da Republica, EAN, 2780-157 Oeiras, Portugal
| | - S Hunot
- Inserm, U 1127, F-75013 Paris, France; CNRS, UMR 7225, F-75013 Paris, France; Sorbonne Universités, UPMC Univ Paris 06, UMR S 1127, F-75013 Paris, France; Institut du Cerveau et de la Moelle épinière, ICM, F-75013 Paris, France
| | - S Chandran
- Centre for Clinical Brain Sciences, Chancellor's Building, University of Edinburgh, Edinburgh EH16 4SB, United Kingdom
| | - S Birman
- Genes Circuits Rhythms and Neuropathology, Brain Plasticity Unit, CNRS UMR 8249, ESPCI ParisTech, PSL Research University, 75005 Paris, France
| | - T Baron
- ANSES, French Agency for Food, Environmental and Occupational Health & Safety, Neurodegenerative Diseases Unit, 31, avenue Tony Garnier, 69364 Lyon Cedex 07, France
| | - D Vivien
- Inserm, UMR-S U919 Serine Proteases and Pathophysiology of the Neurovascular Unit, 14000 Caen, France
| | - C B Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Faculty of Medicine, Rua Larga, and Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| | - H D Ryoo
- Department of Cell Biology, New York University School of Medicine, New York, NY, USA
| | - H Steller
- Howard Medical Institute, The Rockefeller University, 1230 York Avenue, New York, NY 10021, USA
| | - F Urano
- Washington University School of Medicine, Department of Internal Medicine, St. Louis, MO 63110 USA
| | - E Chevet
- Inserm ERL440 "Oncogenesis, Stress, Signaling", Université de Rennes 1, Rennes, France; Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - G Kroemer
- Equipe 11 labellisée par la Ligue contre le Cancer, Centre de Recherche des Cordeliers, Paris, France; Cell Biology and Metabolomics platforms, Gustave Roussy Comprehensive Cancer Center, Villejuif, France; INSERM, U1138, Paris, France; Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Université Pierre et Marie Curie, Paris, France; Pôle de Biologie, Hôpital Européen Georges Pompidou, AP-HP, Paris, France; Karolinska Institute, Department of Women׳s and Children׳s Health, Karolinska University Hospital, Stockholm, Sweden
| | - A Ciechanover
- The Polak Cancer and Vascular Biology Research Center, The Rappaport Faculty of Medicine and Research Institute, Technion-Israel Institute of Technology, Haifa 30196, Israel
| | - E J Calabrese
- Department of Environmental Health Sciences, University of Massachusetts, Morrill I, N344, Amherst, MA 01003, USA
| | - R J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, 10901 N. Torrey Pines Rd., La Jolla, CA 92037, USA
| | - C Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Molecular Studies of the Cell, Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Faculty of Medicine, University of Chile, Santiago, Chile; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|
29
|
McCarthy FP, Adetoba A, Gill C, Bramham K, Bertolaccini M, Burton GJ, Girardi G, Seed PT, Poston L, Chappell LC. Urinary congophilia in women with hypertensive disorders of pregnancy and preexisting proteinuria or hypertension. Am J Obstet Gynecol 2016; 215:464.e1-7. [PMID: 27133010 DOI: 10.1016/j.ajog.2016.04.041] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Revised: 04/15/2016] [Accepted: 04/21/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND Congophilia indicates the presence of amyloid protein, which is an aggregate of misfolded proteins, that is implicated in the pathophysiologic condition of preeclampsia. Recently, urinary congophilia has been proposed as a test for the diagnosis and prediction of preeclampsia. OBJECTIVES The purpose of this study was to determine whether urine congophilia is present in a cohort of women with preeclampsia and in pregnant and nonpregnant women with renal disease. STUDY DESIGN With the use of a preeclampsia, chronic hypertension, renal disease, and systemic lupus erythematosus cohort, we analyzed urine samples from healthy pregnant control subjects (n = 31) and pregnant women with preeclampsia (n = 23), gestational hypertension (n = 10), chronic hypertension (n = 14), chronic kidney disease; n = 28), chronic kidney disease with superimposed preeclampsia (n = 5), and chronic hypertension and superimposed preeclampsia (n = 12). Samples from nonpregnant control subjects (n = 10) and nonpregnant women with either systemic lupus erythematosus with (n = 25) and without (n = 14) lupus nephritis were analyzed. For each sample, protein concentration was standardized before it was mixed with Congo Red, spotted to nitrocellulose membrane, and rinsed with methanol. The optical density of the residual Congo Red stain was determined; Congo red stain retention was calculated, and groups were compared with the use of the Mann-Whitney test or Kruskal-Wallis analysis of Variance test, as appropriate. RESULTS Congophilia was increased in urine from women with preeclampsia (median Congo red stain retention, 47%; interquartile range, 22-68%) compared with healthy pregnant control subjects (Congo red stain retention: 16%; interquartile range, 13-21%; P = .002), women with gestational hypertension (Congo red stain retention, 20%; interquartile range, 13-27%; P = .008), or women with chronic hypertension (Congo red stain retention, 17%; interquartile range, 12-28%; P = .01). There were no differences in Congo red retention between pregnant women with chronic hypertension and normal pregnant control subjects (Congo red stain retention, 17% [interquartile range, 12-28%] vs 16% [interquartile range, 13-21%], respectively; P = .72). Congophilia was present in pregnant women with chronic kidney disease (Congo red stain retention, 32%; interquartile range, 14-57%), being similar to values found in women with preeclampsia (P = .22) and for women with chronic kidney disease and superimposed preeclampsia (Congo red stain retention, 57%; [interquartile range, 29-71%; P = .18). Nonpregnant women with lupus nephritis had higher congophilia levels compared with nonpregnant female control subjects (Congo red stain retention, 38% [interquartile range, 17-73%] vs 9% [7-11%], respectively; P < .001) and nonpregnant women with systemic lupus erythematosus without nephritis (Congo red stain retention, 38% [interquartile range, 17-73%] vs 13% [interquartile range, 11-17%], respectively; P = .001). A significant positive correlation was observed between congophilia and protein:creatinine ratio (Spearman rank correlations, 0.702; 95% confidence interval, 0.618-0.770; P < .001). CONCLUSION This study confirms that women with preeclampsia and chronic kidney disease without preeclampsia have elevated urine congophilia levels compared with healthy pregnant women. Nonpregnant women with lupus nephritis also have elevated urine congophilia levels compared with healthy control subjects. An elevated Congo Red stain retention may not be able to differentiate between these conditions; further research is required to explore the use of congophilia in clinical practice.
Collapse
|
30
|
Chen Y, Holstein DM, Aime S, Bollo M, Lechleiter JD. Calcineurin β protects brain after injury by activating the unfolded protein response. Neurobiol Dis 2016; 94:139-56. [PMID: 27334877 PMCID: PMC4983525 DOI: 10.1016/j.nbd.2016.06.011] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Revised: 06/15/2016] [Accepted: 06/17/2016] [Indexed: 11/26/2022] Open
Abstract
The Ca2+-dependent phosphatase, calcineurin (CN) is thought to play a detrimental role in damaged neurons; however, its role in astrocytes is unclear. In cultured astrocytes, CNβ expression increased after treatment with a sarco/endoplasmic reticulum Ca2+-ATPase inhibitor, thapsigargin, and with oxygen and glucose deprivation, an in vitro model of ischemia. Similarly, CNβ was induced in astrocytes in vivo in two different mouse models of brain injury - photothrombotic stroke and traumatic brain injury (TBI). Immunoprecipitation and chemical activation dimerization methods pointed to physical interaction of CNβ with the unfolded protein response (UPR) sensor, protein kinase RNA-like endoplasmic reticulum kinase (PERK). In accordance, induction of CNβ resulted in oligomerization and activation of PERK. Strikingly, the presence of a phosphatase inhibitor did not interfere with CNβ-mediated activation of PERK, suggesting a hitherto undiscovered non-enzymatic role for CNβ. Importantly, the cytoprotective function of CNβ was PERK-dependent both in vitro and in vivo. Loss of CNβ in vivo resulted in a significant increase in cerebral damage, and correlated with a decrease in astrocyte size, PERK activity and glial fibrillary acidic protein (GFAP) expression. Taken together, these data reveal a critical role for the CNβ-PERK axis in not only prolonging astrocyte cell survival but also in modulating astrogliosis after brain injury.
Collapse
Affiliation(s)
- Yanan Chen
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, TX, USA
| | - Deborah M Holstein
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, TX, USA
| | - Sofia Aime
- Instituto de Investigación Médica M y M Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - Mariana Bollo
- Instituto de Investigación Médica M y M Ferreyra, INIMEC-CONICET, Universidad Nacional de Córdoba, Córdoba, Argentina
| | - James D Lechleiter
- Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, TX, USA; Center for Biomedical Neuroscience, University of Texas Health Science Center at San Antonio, TX, USA.
| |
Collapse
|
31
|
Injury to the nervous system: A look into the ER. Brain Res 2016; 1648:617-625. [PMID: 27117870 DOI: 10.1016/j.brainres.2016.04.053] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 12/12/2022]
Abstract
Injury to the central or peripheral nervous systems leads to the loss of cognitive and/or sensorimotor capabilities that still lack an effective treatment. Although injury to the nervous system involves multiple and complex molecular factors, alteration to protein homeostasis is emerging as a relevant pathological mechanism. In particular, chronic endoplasmic reticulum (ER) stress is proposed as a possible driver of neuronal dysfunction in conditions such as spinal cord injury, stroke and damage to peripheral nerves. Importantly, manipulation of the unfolded protein response (UPR), a homeostatic pathway engaged by ER stress, has proved effective in improving cognitive and motor recovery after nervous system injury. Here we provide an overview on recent findings depicting a functional role of the UPR to the functional recovery after injury in the peripheral and central nervous systems. This article is part of a Special Issue entitled SI:ER stress.
Collapse
|
32
|
Ingberg E, Dock H, Theodorsson E, Theodorsson A, Ström JO. Method parameters' impact on mortality and variability in mouse stroke experiments: a meta-analysis. Sci Rep 2016; 6:21086. [PMID: 26876353 PMCID: PMC4753409 DOI: 10.1038/srep21086] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 01/13/2016] [Indexed: 12/17/2022] Open
Abstract
Although hundreds of promising substances have been tested in clinical trials,
thrombolysis currently remains the only specific pharmacological treatment for
ischemic stroke. Poor quality, e.g. low statistical power, in the preclinical
studies has been suggested to play an important role in these failures. Therefore,
it would be attractive to use animal models optimized to minimize unnecessary
mortality and outcome variability, or at least to be able to power studies more
exactly by predicting variability and mortality given a certain experimental setup.
The possible combinations of methodological parameters are innumerous, and an
experimental comparison of them all is therefore not feasible. As an alternative
approach, we extracted data from 334 experimental mouse stroke articles and, using a
hypothesis-driven meta-analysis, investigated the method parameters’
impact on infarct size variability and mortality. The use of Swiss and C57BL6 mice
as well as permanent occlusion of the middle cerebral artery rendered the lowest
variability of the infarct size while the emboli methods increased variability. The
use of Swiss mice increased mortality. Our study offers guidance for researchers
striving to optimize mouse stroke models.
Collapse
Affiliation(s)
- Edvin Ingberg
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Hua Dock
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Elvar Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden
| | - Annette Theodorsson
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Division of Neuro and Inflammation Science, Department of Clinical and Experimental Medicine, Linköping University, Department of Neurosurgery, Anaesthetics, Operations and Specialty Surgery Center, Region Östergötland, Sweden
| | - Jakob O Ström
- Division of Microbiology and Molecular Medicine, Department of Clinical and Experimental Medicine, Linköping University, Department of Clinical Chemistry, Center for Diagnostics, Region Östergötland, Sweden.,Vårdvetenskapligt Forskningscentrum/Centre for Health Sciences, Örebro University Hospital, County Council of Örebro, Örebro, Sweden.,School of Health and Medical Sciences, Örebro University, Örebro, Sweden
| |
Collapse
|
33
|
Nakka VP, Prakash-Babu P, Vemuganti R. Crosstalk Between Endoplasmic Reticulum Stress, Oxidative Stress, and Autophagy: Potential Therapeutic Targets for Acute CNS Injuries. Mol Neurobiol 2016; 53:532-544. [PMID: 25482050 PMCID: PMC4461562 DOI: 10.1007/s12035-014-9029-6] [Citation(s) in RCA: 185] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2014] [Accepted: 11/30/2014] [Indexed: 01/06/2023]
Abstract
Endoplasmic reticulum (ER) stress induces a variety of neuronal cell death pathways that play a critical role in the pathophysiology of stroke. ER stress occurs when unfolded/misfolded proteins accumulate and the folding capacity of ER chaperones exceeds the capacity of ER lumen to facilitate their disposal. As a consequence, a complex set of signaling pathways will be induced that transmit from ER to cytosol and nucleus to compensate damage and to restore the normal cellular homeostasis, collectively known as unfolded protein response (UPR). However, failure of UPR due to severe or prolonged stress leads to cell death. Following acute CNS injuries, chronic disturbances in protein folding and oxidative stress prolong ER stress leading to sustained ER dysfunction and neuronal cell death. While ER stress responses have been well studied after stroke, there is an emerging need to study the association of ER stress with other cell pathways that exacerbate neuronal death after an injury. In this review, we summarize the current understanding of the role for ER stress in acute brain injuries, highlighting the diverse molecular mechanisms associated with ER stress and its relation to oxidative stress and autophagy. We also discussed the existing and developing therapeutic options aimed to reduce ER stress to protect the CNS after acute injuries.
Collapse
Affiliation(s)
- Venkata Prasuja Nakka
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA
- Department of Biotechnology & Bioinformatics, School of Life sciences, University of Hyderabad, Hyderabad, India
| | - Phanithi Prakash-Babu
- Department of Biotechnology & Bioinformatics, School of Life sciences, University of Hyderabad, Hyderabad, India
| | - Raghu Vemuganti
- Department of Neurological Surgery, School of Medicine and Public Health, University of Wisconsin, Madison, WI, 53792, USA.
| |
Collapse
|
34
|
Activation of cerebral sodium-glucose transporter type 1 function mediated by post-ischemic hyperglycemia exacerbates the development of cerebral ischemia. Neuroscience 2015; 310:674-85. [PMID: 26454021 DOI: 10.1016/j.neuroscience.2015.10.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 09/15/2015] [Accepted: 10/03/2015] [Indexed: 12/21/2022]
Abstract
The regulation of post-ischemic hyperglycemia plays an important role in suppressing neuronal damage in therapeutic strategies for cerebral ischemia. We previously reported that the cerebral sodium-glucose transporter (SGLT) was involved in the post-ischemic hyperglycemia-induced exacerbation of cerebral ischemic neuronal damage. Cortical SGLT-1, one of the cerebral SGLT isoforms, is dramatically increased by focal cerebral ischemia. In this study, we focused on the involvement of cerebral SGLT-1 in the development of cerebral ischemic neuronal damage. It was previously reported that activation of 5'-adenosine monophosphate-activated protein kinase (AMPK) increases SGLT-1 expression. Moreover, ischemic stress-induced activation of AMPK exacerbates cerebral ischemic neuronal damage. Therefore, we directly confirmed the relationship between cerebral SGLT-1 and cerebral AMPK activation using in vitro primary culture of mouse cortical neurons. An in vivo mouse model of focal cerebral ischemia was generated using a middle cerebral artery occlusion (MCAO). The development of infarct volume and behavioral abnormalities on day 3 after MCAO were ameliorated in cerebral SGLT-1 knock down mice. Cortical and striatal SGLT-1 expression levels were significantly increased at 12h after MCAO. Immunofluorescence revealed that SGLT-1 and the neuronal nuclear antigen (NeuN) were co-localized in the cortex and striatum of MCAO mice. In the in vitro study, primary cortical neurons were cultured for five days before each treatment with reagents. Concomitant treatment with hydrogen peroxide and glucose induced the elevation of SGLT-1 and phosphorylated AMPK/AMPK ratio, and this elevation was suppressed by compound C, an AMPK inhibitor in primary cortical neurons. Moreover, compound C suppressed neuronal cell death induced by concomitant hydrogen peroxide/glucose treatment in primary cortical neurons. Therefore, we concluded that enhanced cerebral SGLT-1 function mediated by post-ischemic hyperglycemia exacerbates the development of cerebral ischemic neuronal damage. One of the mechanisms of cerebral SGLT-1 up-regulation may be involved in the AMPK activation after cerebral ischemia.
Collapse
|
35
|
Image analysis algorithms for immunohistochemical assessment of cell death. Methods Mol Biol 2014. [PMID: 25431066 DOI: 10.1007/978-1-4939-2152-2_14] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
Light microscopy allows for the inexpensive and fast detection of neuronal /glial cell demise and estimation of infarct and traumatic lesion volumes; the direct correlates of cell death. Quantitative assessment of brain tissue damage following stroke, traumatic brain injury (TBI ) or neurodegenerative diseases, and recovery after therapeutic intervention has been facilitated by recent developments in computer-assisted image analysis technologies that enable more objective and accurate morphometric quantification of cell injury in whole brain sections. In this chapter, the proposed workflow describes what tasks need to be fulfilled to visualize and gauge cell death characterization by histological stains and immunohistochemical markers.
Collapse
|
36
|
Lucke-Wold BP, Turner RC, Logsdon AF, Bailes JE, Huber JD, Rosen CL. Linking traumatic brain injury to chronic traumatic encephalopathy: identification of potential mechanisms leading to neurofibrillary tangle development. J Neurotrauma 2014; 31:1129-38. [PMID: 24499307 PMCID: PMC4089022 DOI: 10.1089/neu.2013.3303] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Significant attention has recently been drawn to the potential link between head trauma and the development of neurodegenerative disease, namely chronic traumatic encephalopathy (CTE). The acute neurotrauma associated with sports-related concussions in athletes and blast-induced traumatic brain injury in soldiers elevates the risk for future development of chronic neurodegenerative diseases such as CTE. CTE is a progressive disease distinguished by characteristic tau neurofibrillary tangles (NFTs) and, occasionally, transactive response DNA binding protein 43 (TDP43) oligomers, both of which have a predilection for perivascular and subcortical areas near reactive astrocytes and microglia. The disease is currently only diagnosed postmortem by neuropathological identification of NFTs. A recent workshop sponsored by National Institute of Neurological Disorders and Stroke emphasized the need for premortem diagnosis, to better understand disease pathophysiology and to develop targeted treatments. In order to accomplish this objective, it is necessary to discover the mechanistic link between acute neurotrauma and the development of chronic neurodegenerative and neuropsychiatric disorders such as CTE. In this review, we briefly summarize what is currently known about CTE development and pathophysiology, and subsequently discuss injury-induced pathways that warrant further investigation. Understanding the mechanistic link between acute brain injury and chronic neurodegeneration will facilitate the development of appropriate diagnostic and therapeutic options for CTE and other related disorders.
Collapse
Affiliation(s)
- Brandon Peter Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, West Virginia
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Ryan Coddington Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, West Virginia
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Aric Flint Logsdon
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Julian Edwin Bailes
- Department of Neurosurgery, NorthShore University Health System, University of Chicago Pritzker School of Medicine, Evanston, Illinois
| | - Jason Delwyn Huber
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Medicine, Morgantown, West Virginia
| | - Charles Lee Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, West Virginia
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, West Virginia
| |
Collapse
|
37
|
Otellin VA, Khozhai LI, Shishko TT. Reactions of neural elements of neocortex to action of hypoxia at the early neonatal period in rats. J EVOL BIOCHEM PHYS+ 2014. [DOI: 10.1134/s0022093014020094] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
38
|
Lee GH, Lee HY, Li B, Kim HR, Chae HJ. Bax inhibitor-1-mediated inhibition of mitochondrial Ca2+ intake regulates mitochondrial permeability transition pore opening and cell death. Sci Rep 2014; 4:5194. [PMID: 24899098 PMCID: PMC4046133 DOI: 10.1038/srep05194] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 05/20/2014] [Indexed: 12/13/2022] Open
Abstract
A recently studied endoplasmic reticulum (ER) stress regulator, Bax inhibitor-1 (BI-1) plays a regulatory role in mitochondrial Ca2+ levels. In this study, we identified ER-resident and mitochondria-associated ER membrane (MAM)-resident populations of BI-1. ER stress increased mitochondrial Ca2+ to a lesser extent in BI-1–overexpressing cells (HT1080/BI-1) than in control cells, most likely as a result of impaired mitochondrial Ca2+ intake ability and lower basal levels of intra-ER Ca2+. Moreover, opening of the Ca2+-induced mitochondrial permeability transition pore (PTP) and cytochrome c release were regulated by BI-1. In HT1080/BI-1, the basal mitochondrial membrane potential was low and also resistant to Ca2+ compared with control cells. The activity of the mitochondrial membrane potential-dependent mitochondrial Ca2+ intake pore, the Ca2+ uniporter, was reduced in the presence of BI-1. This study also showed that instead of Ca2+, other cations including K+ enter the mitochondria of HT1080/BI-1 through mitochondrial Ca2+-dependent ion channels, providing a possible mechanism by which mitochondrial Ca2+ intake is reduced, leading to cell protection. We propose a model in which BI-1–mediated sequential regulation of the mitochondrial Ca2+ uniporter and Ca2+-dependent K+ channel opening inhibits mitochondrial Ca2+ intake, thereby inhibiting PTP function and leading to cell protection.
Collapse
Affiliation(s)
- Geum-Hwa Lee
- Department of Pharmacology and Cardiovascular Research Institute, Medical School, Chonbuk National University, Jeonju, 561-181, Republic of Korea
| | - Hwa-Young Lee
- Department of Pharmacology and Cardiovascular Research Institute, Medical School, Chonbuk National University, Jeonju, 561-181, Republic of Korea
| | - Bo Li
- Department of Pharmacology and Cardiovascular Research Institute, Medical School, Chonbuk National University, Jeonju, 561-181, Republic of Korea
| | - Hyung-Ryong Kim
- Department of Dental Pharmacology and Wonkwang Dental Research Institute, School of Dentistry, Wonkwang University, Iksan, 570-749, Republic of Korea
| | - Han-Jung Chae
- Department of Pharmacology and Cardiovascular Research Institute, Medical School, Chonbuk National University, Jeonju, 561-181, Republic of Korea
| |
Collapse
|
39
|
Docosahexaenoic acid reduces ER stress and abnormal protein accumulation and improves neuronal function following traumatic brain injury. J Neurosci 2014; 34:3743-55. [PMID: 24599472 DOI: 10.1523/jneurosci.2872-13.2014] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In this study, we investigated the development of endoplasmic reticulum (ER) stress after traumatic brain injury (TBI) and the efficacy of post-TBI administration of docosahexaenoic acid (DHA) in reducing ER stress. TBI was induced by cortical contusion injury in Sprague-Dawley rats. Either DHA (16 mg/kg in DMSO) or vehicle DMSO (1 ml/kg) was administered intraperitoneally at 5 min after TBI, followed by a daily dose for 3-21 d. TBI triggered sustained expression of the ER stress marker proteins including phosphorylated eukaryotic initiation factor-2α, activating transcription factor 4, inositol requiring kinase 1, and C/EBP homologous protein in the ipsilateral cortex at 3-21 d after TBI. The prolonged ER stress was accompanied with an accumulation of abnormal ubiquitin aggregates and increased expression of amyloid precursor protein (APP) and phosphorylated tau (p-Tau) in the frontal cortex after TBI. The ER stress marker proteins were colocalized with APP accumulation in the soma. Interestingly, administration of DHA attenuated all ER stress marker proteins and reduced the accumulation of both ubiquitinated proteins and APP/p-Tau proteins. In addition, the DHA-treated animals exhibited early recovery of their sensorimotor function after TBI. In summary, our study demonstrated that TBI induces a prolonged ER stress, which is positively correlated with abnormal APP accumulation. The sustained ER stress may play a role in chronic neuronal damage after TBI. Our findings illustrate that post-TBI administration of DHA has therapeutic potentials in reducing ER stress, abnormal protein accumulation, and neurological deficits.
Collapse
|
40
|
TMBIM protein family: ancestral regulators of cell death. Oncogene 2014; 34:269-80. [DOI: 10.1038/onc.2014.6] [Citation(s) in RCA: 75] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 12/27/2013] [Accepted: 01/02/2014] [Indexed: 12/13/2022]
|
41
|
B L, R.K Y, G.S J, H.-R K, H.-J C. The characteristics of Bax inhibitor-1 and its related diseases. Curr Mol Med 2014; 14:603-15. [PMID: 24894176 PMCID: PMC4083451 DOI: 10.2174/1566524014666140603101113] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2013] [Revised: 10/01/2013] [Accepted: 11/24/2013] [Indexed: 11/28/2022]
Abstract
Bax inhibitor-1 (BI-1) is an evolutionarily-conserved endoplasmic reticulum protein. The expression of BI-1 in mammalian cells suppresses apoptosis induced by Bax, a pro-apoptotic member of the Bcl-2 family. BI-1 has been shown to be associated with calcium (Ca(2+)) levels, reactive oxygen species (ROS) production, cytosolic acidification, and autophagy as well as endoplasmic reticulum stress signaling pathways. According to both in vitro and clinical studies, BI-1 promotes the characteristics of cancers. In other diseases, BI-1 has also been shown to regulate insulin resistance, adipocyte differentiation, hepatic dysfunction and depression. However, the roles of BI-1 in these disease conditions are not fully consistent among studies. Until now, the molecular mechanisms of BI-1 have not directly explained with regard to how these conditions can be regulated. Therefore, this review investigates the physiological role of BI-1 through molecular mechanism studies and its application in various diseases.
Collapse
Affiliation(s)
- Li B
- Department of Pharmacology, Medical School, Chonbuk National University, Jeonju, 561-181, Republic of Korea
| | - Yadav R.K
- Department of Pharmacology, Medical School, Chonbuk National University, Jeonju, 561-181, Republic of Korea
| | - Jeong G.S
- Department of Pharmacology, Medical School, Chonbuk National University, Jeonju, 561-181, Republic of Korea
| | - Kim H.-R
- Department of Dental Pharmacology and Wonkwang Dental Research Institute, School of Dentistry, Wonkwang University, Iksan, 570-749, Republic of Korea
| | - Chae H.-J
- Department of Pharmacology, Medical School, Chonbuk National University, Jeonju, 561-181, Republic of Korea
| |
Collapse
|
42
|
The protective role of Bax inhibitor-1 against chronic mild stress through the inhibition of monoamine oxidase A. Sci Rep 2013; 3:3398. [PMID: 24292328 PMCID: PMC3844965 DOI: 10.1038/srep03398] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 11/15/2013] [Indexed: 11/08/2022] Open
Abstract
The anti-apoptotic protein Bax inhibitor-1 (BI-1) is a regulator of apoptosis linked to endoplasmic reticulum (ER) stress. It has been hypothesized that BI-1 protects against neuron degenerative diseases. In this study, BI-1⁻/⁻ mice showed increased vulnerability to chronic mild stress accompanied by alterations in the size and morphology of the hippocampi, enhanced ROS accumulation and an ER stress response compared with BI-1⁺/⁺ mice. BI-1⁻/⁻ mice exposed to chronic mild stress showed significant activation of monoamine oxidase A (MAO-A), but not MAO-B, compared with BI-1⁺/⁺ mice. To examine the involvement of BI-1 in the Ca²⁺-sensitive MAO activity, thapsigargin-induced Ca²⁺ release and MAO activity were analyzed in neuronal cells overexpressing BI-1. The in vitro study showed that BI-1 regulates Ca²⁺ release and related MAO-A activity. This study indicates an endogenous protective role of BI-1 under conditions of chronic mild stress that is primarily mediated through Ca²⁺-associated MAO-A regulation.
Collapse
|
43
|
Begum G, Harvey L, Dixon CE, Sun D. ER stress and effects of DHA as an ER stress inhibitor. Transl Stroke Res 2013; 4:635-42. [PMID: 24323417 PMCID: PMC3864671 DOI: 10.1007/s12975-013-0282-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Revised: 08/01/2013] [Accepted: 08/06/2013] [Indexed: 11/30/2022]
Abstract
The endoplasmic reticulum (ER) functions in the synthesis, folding, modification, and transport of newly synthesized transmembrane and secretory proteins. The ER also has important roles in the storage of intracellular Ca(2+) and regulation of Ca(2+) homeostasis. The integrity of the Ca(2+) homeostasis in the ER lumen is vital for proper folding of proteins. Dysregulation of ER Ca(2+) could result in an increase in unfolded or misfolded proteins and ER stress. ER stress triggers activation of the unfolded protein response (UPR), which is a fundamentally adaptive cell response and functions as a cytoprotective mechanism by over-expression of relevant chaperones and the global shutdown of protein synthesis. UPR activation occurs when three key ER membrane-sensor proteins detect an accumulation of aberrant proteins. The UPR acts to alleviate ER stress, but if the stress is too severe or prolonged, apoptosis will be triggered. In this review, we focused on ER stress and the effects of docosahexaenoic acid (DHA) on ER stress. DHA and its bioactive compounds, such as protectins and resolvins, provide neuroprotection against oxidative stress and apoptosis and have the ability to resolve inflammation in neurological diseases. New studies reveal that DHA blocks inositol trisphosphate receptor (IP3R)-mediated ER Ca(2+) depletion and ER stress. The administration of DHA post-traumatic brain injury (TBI) reduces ER stress, aberrant protein accumulation, and neurological deficits. Therefore, DHA presents therapeutic potentials for TBI via its pleiotropic effects including inhibition of ER stress.
Collapse
Affiliation(s)
- Gulnaz Begum
- Dept. of Neurology, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, PA 15213
| | - Lloyd Harvey
- Dept. of Neurology, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, PA 15213
| | - C. Edward Dixon
- Dept. of Neurosurgery, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, PA 15213
| | - Dandan Sun
- Dept. of Neurology, Brain Trauma Research Center, University of Pittsburgh, Pittsburgh, PA 15213
| |
Collapse
|
44
|
Li S, Yang L, Selzer ME, Hu Y. Neuronal endoplasmic reticulum stress in axon injury and neurodegeneration. Ann Neurol 2013; 74:768-77. [PMID: 23955583 PMCID: PMC3963272 DOI: 10.1002/ana.24005] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 08/04/2013] [Accepted: 08/07/2013] [Indexed: 12/13/2022]
Abstract
Injuries to central nervous system axons result not only in Wallerian degeneration of the axon distal to the injury, but also in death or atrophy of the axotomized neurons, depending on injury location and neuron type. No method of permanently avoiding these changes has been found, despite extensive knowledge concerning mechanisms of secondary neuronal injury. The autonomous endoplasmic reticulum (ER) stress pathway in neurons has recently been implicated in retrograde neuronal degeneration. In addition to the emerging role of ER morphology in axon maintenance, we propose that ER stress is a common neuronal response to disturbances in axon integrity and a general mechanism for neurodegeneration. Thus, manipulation of the ER stress pathway could have important therapeutic implications for neuroprotection.
Collapse
Affiliation(s)
- Shaohua Li
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Temple University School of Medicine, Philadelphia, PA, USA
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Liu Yang
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Temple University School of Medicine, Philadelphia, PA, USA
| | - Michael E. Selzer
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Temple University School of Medicine, Philadelphia, PA, USA
- Department of Neurology, Temple University School of Medicine, Philadelphia, PA, USA
| | - Yang Hu
- Shriners Hospitals Pediatric Research Center (Center for Neural Repair and Rehabilitation), Temple University School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
45
|
Urra H, Dufey E, Lisbona F, Rojas-Rivera D, Hetz C. When ER stress reaches a dead end. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2013; 1833:3507-3517. [PMID: 23988738 DOI: 10.1016/j.bbamcr.2013.07.024] [Citation(s) in RCA: 325] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2013] [Revised: 07/25/2013] [Accepted: 07/30/2013] [Indexed: 02/06/2023]
Abstract
Endoplasmic reticulum (ER) stress is a common feature of several physiological and pathological conditions affecting the function of the secretory pathway. To restore ER homeostasis, an orchestrated signaling pathway is engaged that is known as the unfolded protein response (UPR). The UPR has a primary function in stress adaptation and cell survival; however, under irreversible ER stress a switch to pro-apoptotic signaling events induces apoptosis of damaged cells. The mechanisms that initiate ER stress-dependent apoptosis are not fully understood. Several pathways have been described where we highlight the participation of the BCL-2 family of proteins and ER calcium release. In addition, recent findings also suggest that microRNAs and oxidative stress are relevant players on the transition from adaptive to cell death programs. Here we provide a global and integrated overview of the signaling networks that may determine the elimination of a cell under chronic ER stress. This article is part of a Special Section entitled: Cell Death Pathways.
Collapse
Affiliation(s)
- Hery Urra
- Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Estefanie Dufey
- Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Fernanda Lisbona
- Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Diego Rojas-Rivera
- Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Claudio Hetz
- Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA 02115, USA; Neurounion Biomedical Foundation, Santiago, Chile.
| |
Collapse
|
46
|
Garweg JG, Tappeiner C, Halberstadt M. Pathophysiology of proliferative vitreoretinopathy in retinal detachment. Surv Ophthalmol 2013; 58:321-9. [PMID: 23642514 DOI: 10.1016/j.survophthal.2012.12.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2012] [Revised: 12/02/2012] [Accepted: 12/04/2012] [Indexed: 10/26/2022]
Abstract
Because proliferative vitreoretinopathy cannot be effectively treated, its prevention is indispensable for the success of surgery for retinal detachment. The elaboration of preventive and therapeutic strategies depends upon the identification of patients who are genetically predisposed to develop the disease, as well as upon an understanding of the biological process involved and the role of local factors, such as the status of the uveovascular barrier. Detachment of the retina or vitreous activates glia to release cytokines and ATP, which not only protect the neuroretina but also promote inflammation, retinal ischemia, cell proliferation, and tissue remodeling. The vitreal microenvironment favors cellular de-differentiation and proliferation of cells with nonspecific nutritional requirements. This may render a pharmacological inhibition of their growth difficult without causing damage to the pharmacologically vulnerable neuroretina. Moreover, reattachment of the retina relies upon the local induction of a controlled wound-healing response involving macrophages and proliferating glia. Hence, the functional outcome of proliferative vitreoretinopathy will be determined by the equilibrium established between protective and destructive repair mechanisms, which will be influenced by the location and the degree of damage to the photoreceptor cells that is induced by peri-retinal gliosis.
Collapse
Affiliation(s)
- Justus G Garweg
- Swiss Eye Institute and University of Bern, Bern, Switzerland.
| | | | | |
Collapse
|
47
|
Sano R, Hou YCC, Hedvat M, Correa RG, Shu CW, Krajewska M, Diaz PW, Tamble CM, Quarato G, Gottlieb RA, Yamaguchi M, Nizet V, Dahl R, Thomas DD, Tait SW, Green DR, Fisher PB, Matsuzawa SI, Reed JC. Endoplasmic reticulum protein BI-1 regulates Ca²⁺-mediated bioenergetics to promote autophagy. Genes Dev 2012; 26:1041-54. [PMID: 22588718 DOI: 10.1101/gad.184325.111] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Autophagy is a lysosomal degradation pathway that converts macromolecules into substrates for energy production during nutrient-scarce conditions such as those encountered in tumor microenvironments. Constitutive mitochondrial uptake of endoplasmic reticulum (ER) Ca²⁺ mediated by inositol triphosphate receptors (IP₃Rs) maintains cellular bioenergetics, thus suppressing autophagy. We show that the ER membrane protein Bax inhibitor-1 (BI-1) promotes autophagy in an IP₃R-dependent manner. By reducing steady-state levels of ER Ca²⁺ via IP₃Rs, BI-1 influences mitochondrial bioenergetics, reducing oxygen consumption, impacting cellular ATP levels, and stimulating autophagy. Furthermore, BI-1-deficient mice show reduced basal autophagy, and experimentally reducing BI-1 expression impairs tumor xenograft growth in vivo. BI-1's ability to promote autophagy could be dissociated from its known function as a modulator of IRE1 signaling in the context of ER stress. The results reveal BI-1 as a novel autophagy regulator that bridges Ca²⁺ signaling between ER and mitochondria, reducing cellular oxygen consumption and contributing to cellular resilience in the face of metabolic stress.
Collapse
Affiliation(s)
- Renata Sano
- Sanford-Burnham Medical Research Institute, La Jolla, California 92037, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Protein disulfide isomerase-associated 6 is an ATF6-inducible ER stress response protein that protects cardiac myocytes from ischemia/reperfusion-mediated cell death. J Mol Cell Cardiol 2012; 53:259-67. [PMID: 22609432 DOI: 10.1016/j.yjmcc.2012.05.005] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2012] [Revised: 05/07/2012] [Accepted: 05/08/2012] [Indexed: 12/30/2022]
Abstract
Proper folding of secreted and transmembrane proteins made in the rough endoplasmic reticulum (ER) requires oxygen for disulfide bond formation. Accordingly, ischemia can impair ER protein folding and initiate the ER stress response, which we previously showed is activated in the ischemic heart and in culture cardiac myocytes subjected to simulated ischemia. ER stress and ischemia activate the transcription factor, activating transcription factor 6 (ATF6), which induces numerous genes, many of which have not been identified, or examined in the heart. Using an ATF6 transgenic mouse model, we previously showed that ATF6 protected the heart from ischemic damage; however, the mechanism of this protection remains to be determined. In this study, we showed that, in the mouse heart, and in cultured cardiac myocytes, ATF6 induced the protein disulfide isomerase associated 6 (PDIA6) gene, which encodes an ER enzyme that catalyzes protein disulfide bond formation. Moreover, in cultured cardiac myocytes, ER stress-mediated PDIA6 promoter activation was ATF6-dependent, and required an ER stress response element (ERSE) and a nearby CCAAT box element. Electromobility shift assays and chromatin immunoprecipitation showed that ATF6 bound to the ERSE in the PDIA6 promoter, in vitro, and in the mouse heart, in vivo. Gain- and loss-of-function studies showed that PDIA6 protected cardiac myocytes against simulated ischemia/reperfusion-induced death in a manner that was dependent on the catalytic activity of PDIA6. Thus, by facilitating disulfide bond formation, and enhanced ER protein folding, PDIA6 may contribute to the protective effects of ATF6 in the ischemic mouse heart.
Collapse
|
49
|
Schoch KM, Madathil SK, Saatman KE. Genetic manipulation of cell death and neuroplasticity pathways in traumatic brain injury. Neurotherapeutics 2012; 9:323-37. [PMID: 22362424 PMCID: PMC3337028 DOI: 10.1007/s13311-012-0107-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Traumatic brain injury (TBI) initiates a complex cascade of secondary neurodegenerative mechanisms contributing to cell dysfunction and necrotic and apoptotic cell death. The injured brain responds by activating endogenous reparative processes to counter the neurodegeneration or remodel the brain to enhance functional recovery. A vast array of genetically altered mice provide a unique opportunity to target single genes or proteins to better understand their role in cell death and endogenous repair after TBI. Among the earliest targets for transgenic and knockout studies in TBI have been programmed cell death mediators, such as the Bcl-2 family of proteins, caspases, and caspase-independent pathways. In addition, the role of cell cycle regulatory elements in the posttraumatic cell death pathway has been explored in mouse models. As interest grows in neuroplasticity in TBI, the use of transgenic and knockout mice in studies focused on gliogenesis, neurogenesis, and the balance of growth-promoting and growth-inhibiting molecules has increased in recent years. With proper consideration of potential effects of constitutive gene alteration, traditional transgenic and knockout models can provide valuable insights into TBI pathobiology. Through increasing sophistication of conditional and cell-type specific genetic manipulations, TBI studies in genetically altered mice will be increasingly useful for identification and validation of novel therapeutic targets.
Collapse
Affiliation(s)
- Kathleen M. Schoch
- Spinal Cord and Brain Injury Research Center and Department of Physiology, University of Kentucky College of Medicine, B473 Biomedical and Biological Sciences Research Building (BBSRB), 741 South Limestone Street, Lexington, KY 40536 USA
| | - Sindhu K. Madathil
- Spinal Cord and Brain Injury Research Center and Department of Physiology, University of Kentucky College of Medicine, B473 Biomedical and Biological Sciences Research Building (BBSRB), 741 South Limestone Street, Lexington, KY 40536 USA
| | - Kathryn E. Saatman
- Spinal Cord and Brain Injury Research Center and Department of Physiology, University of Kentucky College of Medicine, B473 Biomedical and Biological Sciences Research Building (BBSRB), 741 South Limestone Street, Lexington, KY 40536 USA
| |
Collapse
|
50
|
Lee GH, Kim HR, Chae HJ. Bax inhibitor-1 regulates the expression of P450 2E1 through enhanced lysosome activity. Int J Biochem Cell Biol 2012; 44:600-11. [DOI: 10.1016/j.biocel.2011.12.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2011] [Revised: 12/20/2011] [Accepted: 12/22/2011] [Indexed: 10/14/2022]
|