1
|
Wang F, Han X, Mu Q, Chen H, Wu Y, Kang Y, Liu Y. Cerebrospinal fluid mesencephalic astrocyte-derived neurotrophic factor: A moderating effect on sleep time and cognitive function. J Psychiatr Res 2024; 176:33-39. [PMID: 38838432 DOI: 10.1016/j.jpsychires.2024.05.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 04/15/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Sleeping late has been associated with cognitive impairment, and insufficient sleep can affect the secretion of feeding-related cytokines. Feeding-related cytokines may contribute to cognitive deficits resulting from delayed bedtime. Glial cell line-derived neurotrophic factor (GDNF) and mesencephalic astrocyte-derived neurotrophic factor (MANF), which are feeding-related neurotrophic factors, have been associated with improved cognitive function and neuroprotective abilities. Enhanced expression of GDNF and MANF is linked to increased energy expenditure and hyperphagia, respectively. AIMS This study aimed to investigate the association between cerebrospinal fluid (CSF) GDNF, MANF, cognition, and sleep time and to explore the moderating effects of GDNF and MANF on cognitive impairment in individuals who sleep late. METHOD This cross-sectional study included participants (mean age 31.76 ± 10.22 years) who were categorized as ≤23 o'clock sleepers (n = 66) and >23 o'clock sleepers (n = 125) based on sleep time. Cognition was assessed using Montreal Cognitive Assessment (MoCA), and GDNF and MANF levels in CSF were measured. RESULTS MANF may play a moderating role in the relationship between sleep time and cognition (R2 = 0.06, β = 0.59, p = 0.031). Age showed a negative correlation with MoCA scores (R2 = 0.08, β = -0.18), while education exhibited a positive correlation (β = 0.17, both p < 0.05). Only ≤23 o'clock sleepers exhibited a negative correlation between MANF levels and BMI (r = -0.35, p = 0.005). CONCLUSIONS This study provides hitherto undocumented evidence of the potential protective effect of CSF MANF on cognitive impairment of late sleepers, which suggests that maintaining a regular sleep schedule may contribute to cognition and overall health, with MANF playing a role in this process.
Collapse
Affiliation(s)
- Fan Wang
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, China; Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830063, China.
| | - Xiaoli Han
- Clinical Nutrition Department, Friendship Hospital, Urumqi, 830049, China
| | - Qingshuang Mu
- Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830063, China
| | - Hongxu Chen
- Xinjiang Key Laboratory of Neurological Disorder Research, The Second Affiliated Hospital of Xinjiang Medical University, Urumqi, 830063, China
| | - Yan Wu
- Beijing Hui-Long-Guan Hospital, Peking University, Beijing, 100096, China
| | - Yimin Kang
- Medical Neurobiology Lab, Inner Mongolia Medical University, Huhhot, 010110, China
| | - Yanlong Liu
- School of Mental Health, Wenzhou Medical University, Wenzhou, 325035, China; Zhejiang Provincial Clinical Research Center for Mental Disorders, The Affiliated Wenzhou Kangning Hospital, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
2
|
1,25(OH) 2D 3 Alleviates Aβ(25-35)-Induced Tau Hyperphosphorylation, Excessive Reactive Oxygen Species, and Apoptosis Through Interplay with Glial Cell Line-Derived Neurotrophic Factor Signaling in SH-SY5Y Cells. Int J Mol Sci 2020; 21:ijms21124215. [PMID: 32545801 PMCID: PMC7352552 DOI: 10.3390/ijms21124215] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/09/2020] [Accepted: 06/11/2020] [Indexed: 01/14/2023] Open
Abstract
Amyloid beta (Aβ) accumulation in the brain is one of the major pathological features of Alzheimer's disease. The active form of vitamin D (1,25(OH)2D3), which acts via its nuclear hormone receptor, vitamin D receptor (VDR), has been implicated in the treatment of Aβ pathology, and is thus considered as a neuroprotective agent. However, its underlying molecular mechanisms of action are not yet fully understood. Here, we aim to investigate whether the molecular mechanisms of 1,25(OH)2D3 in ameliorating Aβ toxicity involve an interplay of glial cell line-derived neurotrophic factor (GDNF)-signaling in SH-SY5Y cells. Cells were treated with Aβ(25-35) as the source of toxicity, followed by the addition of 1,25(OH)2D3 with or without the GDNF inhibitor, heparinase III. The results show that 1,25(OH)2D3 modulated Aβ-induced reactive oxygen species, apoptosis, and tau protein hyperphosphorylation in SH-SY5Y cells. Additionally, 1,25(OH)2D3 restored the decreasing GDNF and the inhibited phosphorylation of the phosphatidylinositol 3 kinase (PI3K)/protein kinase B (Akt)/glycogen synthase kinase-3β (GSK-3β) protein expressions. In the presence of heparinase III, these damaging effects evoked by Aβ were not abolished by 1,25(OH)2D3. It appears 1,25(OH)2D3 is beneficial for the alleviation of Aβ neurotoxicity, and it might elicit its neuroprotection against Aβ neurotoxicity through an interplay with GDNF-signaling.
Collapse
|
3
|
Xie Q, Chen X, Meng ZM, Huang XL, Zhang Q, Zhou JQ, Zhang L, He FQ, Zou YP, Gan HT. Glial-derived neurotrophic factor regulates enteric mast cells and ameliorates dextran sulfate sodium-induced experimental colitis. Int Immunopharmacol 2020; 85:106638. [PMID: 32470881 DOI: 10.1016/j.intimp.2020.106638] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 05/11/2020] [Accepted: 05/22/2020] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Although interactions between enteric glial cells (EGCs) and enteric mast cells have been demonstrated to play an important role in the pathogenesis of inflammatory bowel disease (IBD), the exact mechanisms by which EGCs regulate enteric mast cells are still unknown. The aims of this study were to investigate whether glial-derived neurotrophic factor (GDNF), which has been confirmed to be produced mostly by EGCs, might regulate enteric mast cells and ameliorate dextran sulfate sodium (DSS)-induced experimental colitis. METHODS Recombinant adenoviral vectors encoding GDNF (Ad-GDNF) were administered intracolonically in experimental colitis induced by DSS. The disease activity index and histological score were measured. The expression of tumour necrosis factor-α (TNF-α), interleukin-6 and myeloperoxidase (MPO) activity were measured by ELISA assay. The expression of trypsin and β-hexosaminidase were evaluated. GDNF specific receptor (GFR-α1/RET) was detected. The calcium reflux was tested by microplate reader. The expression p-JNK was analyzed by western blot assay. RESULTS GDNF resulted in a significant inhibition of the activation of enteric mast cells by down-regulating JNK signal pathway, lessening intracellular calcium influx, and then reducing the degranulation as well as the expression of pro-inflammatory cytokines via combing with its receptor (GFR-α1/RET) in mast cells, and these inhibitory effects were abrogated by treatment with neutralizing antibody against GDNF. Moreover, the administration of GDNF led to an amelioration of experimental colitis. CONCLUSIONS GDNF are able to regulate enteric mast cells and ameliorate experimental colitis. GDNF might be an important mediator of the cross-talk between EGCs and enteric mast cells, and GDNF might be a useful therapeutic drug for IBD.
Collapse
Affiliation(s)
- Qin Xie
- Department of Geriatric Medicine and Gastroenterology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China; Chinese Academy of Sciences Sichuan Translational Medical Research Hospital, Chengdu 610072, China
| | - Xi Chen
- Department of Gastroenterology and the Center of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Zhang Min Meng
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiao Li Huang
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Qiao Zhang
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Jin Qiu Zhou
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Zhang
- Department of Geriatric Medicine and Gastroenterology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Fu Qian He
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Yu Pei Zou
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Hua Tian Gan
- Department of Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China; Department of Gastroenterology and the Center of Inflammatory Bowel Disease, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China.
| |
Collapse
|
4
|
Xu S, Di Z, He Y, Wang R, Ma Y, Sun R, Li J, Wang T, Shen Y, Fang S, Feng L, Shen Y. Mesencephalic astrocyte-derived neurotrophic factor (MANF) protects against Aβ toxicity via attenuating Aβ-induced endoplasmic reticulum stress. J Neuroinflammation 2019; 16:35. [PMID: 30760285 PMCID: PMC6373169 DOI: 10.1186/s12974-019-1429-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/03/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Extracellular accumulation of amyloid β-peptide (Aβ) is one of pathological hallmarks of Alzheimer's disease (AD) and contributes to the neuronal loss. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER) stress-inducible neurotrophic factor. Many groups, including ours, have proved that MANF rescues neuronal loss in several neurological disorders, such as Parkinson's disease and cerebral ischemia. However, whether MANF exerts its protective effect against Aβ neurotoxicity in AD remains unknown. METHODS In the present study, the characteristic expressions of MANF in Aβ1-42-treated neuronal cells as well as in the brains of APP/PS1 transgenic mice were analyzed by immunofluorescence staining, qPCR, and Western blot. The effects of MANF overexpression, MANF knockdown, or recombination human MANF protein (rhMANF) on neuron viability, apoptosis, and the expression of ER stress-related proteins following Aβ1-42 exposure were also investigated. RESULTS The results showed the increased expressions of MANF, as well as ER stress markers immunoglobulin-binding protein (BiP) and C/EBP homologous protein (CHOP), in the brains of the APP/PS1 transgenic mice and Aβ1-42-treated neuronal cells. MANF overexpression or rhMANF treatment partially protected against Aβ1-42-induced neuronal cell death, associated with marked decrease of cleaved caspase-3, whereas MANF knockdown with siRNA aggravated Aβ1-42 cytotoxicity including caspase-3 activation. Further study demonstrated that the expressions of BiP, ATF6, phosphorylated-IRE1, XBP1s, phosphorylated-eIF2α, ATF4, and CHOP were significantly downregulated by MANF overexpression or rhMANF treatment in neuronal cells following Aβ1-42 exposure, whereas knockdown of MANF has the opposite effect. CONCLUSIONS These findings demonstrate that MANF may exert neuroprotective effects against Aβ-induced neurotoxicity through attenuating ER stress, suggesting that an applicability of MANF as a therapeutic candidate for AD.
Collapse
Affiliation(s)
- Shengchun Xu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Zemin Di
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yufeng He
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Runjie Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yuyang Ma
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Rui Sun
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Jing Li
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Tao Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yujun Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Shengyun Fang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Center for Biomedical Engineering and Technology, University of Maryland, Baltimore, MD, USA
| | - Lijie Feng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China. .,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China. .,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China. .,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China. .,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
5
|
Panagaki T, Gengler S, Hölscher C. The Novel DA–CH3 Dual Incretin Restores Endoplasmic Reticulum Stress and Autophagy Impairments to Attenuate Alzheimer-Like Pathology and Cognitive Decrements in the APPSWE/PS1ΔE9 Mouse Model. J Alzheimers Dis 2018; 66:195-218. [DOI: 10.3233/jad-180584] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Theodora Panagaki
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| | - Simon Gengler
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| | - Christian Hölscher
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, UK
| |
Collapse
|
6
|
Sampaio TB, Savall AS, Gutierrez MEZ, Pinton S. Neurotrophic factors in Alzheimer's and Parkinson's diseases: implications for pathogenesis and therapy. Neural Regen Res 2017; 12:549-557. [PMID: 28553325 PMCID: PMC5436343 DOI: 10.4103/1673-5374.205084] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neurotrophic factors comprise essential secreted proteins that have several functions in neural and non-neural tissues, mediating the development, survival and maintenance of peripheral and central nervous system. Therefore, neurotrophic factor issue has been extensively investigated into the context of neurodegenerative diseases. Alzheimer's disease and Parkinson's disease show changes in the regulation of specific neurotrophic factors and their receptors, which appear to be critical for neuronal degeneration. Indeed, neurotrophic factors prevent cell death in degenerative processes and can enhance the growth and function of affected neurons in these disorders. Based on recent reports, this review discusses the main findings related to the neurotrophic factor support – mainly brain-derived neurotrophic factor and glial cell line-derived neurotrophic factor – in the survival, proliferation and maturation of affected neurons in Alzheimer's disease and Parkinson's disease as well as their putative application as new therapeutic approach for these diseases management.
Collapse
Affiliation(s)
- Tuane Bazanella Sampaio
- Departamento de Farmacologia, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, SC, Brazil.,Universidade Federal do Pampa - Campus Uruguaiana, Uruguaiana, RS, Brazil
| | - Anne Suely Savall
- Universidade Federal do Pampa - Campus Uruguaiana, Uruguaiana, RS, Brazil
| | | | - Simone Pinton
- Universidade Federal do Pampa - Campus Uruguaiana, Uruguaiana, RS, Brazil
| |
Collapse
|
7
|
Perri ER, Thomas CJ, Parakh S, Spencer DM, Atkin JD. The Unfolded Protein Response and the Role of Protein Disulfide Isomerase in Neurodegeneration. Front Cell Dev Biol 2016; 3:80. [PMID: 26779479 PMCID: PMC4705227 DOI: 10.3389/fcell.2015.00080] [Citation(s) in RCA: 98] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/03/2015] [Indexed: 12/11/2022] Open
Abstract
The maintenance and regulation of proteostasis is a critical function for post-mitotic neurons and its dysregulation is increasingly implicated in neurodegenerative diseases. Despite having different clinical manifestations, these disorders share similar pathology; an accumulation of misfolded proteins in neurons and subsequent disruption to cellular proteostasis. The endoplasmic reticulum (ER) is an important component of proteostasis, and when the accumulation of misfolded proteins occurs within the ER, this disturbs ER homeostasis, giving rise to ER stress. This triggers the unfolded protein response (UPR), distinct signaling pathways that whilst initially protective, are pro-apoptotic if ER stress is prolonged. ER stress is increasingly implicated in neurodegenerative diseases, and emerging evidence highlights the complexity of the UPR in these disorders, with both protective and detrimental components being described. Protein Disulfide Isomerase (PDI) is an ER chaperone induced during ER stress that is responsible for the formation of disulfide bonds in proteins. Whilst initially considered to be protective, recent studies have revealed unconventional roles for PDI in neurodegenerative diseases, distinct from its normal function in the UPR and the ER, although these mechanisms remain poorly defined. However, specific aspects of PDI function may offer the potential to be exploited therapeutically in the future. This review will focus on the evidence linking ER stress and the UPR to neurodegenerative diseases, with particular emphasis on the emerging functions ascribed to PDI in these conditions.
Collapse
Affiliation(s)
- Emma R Perri
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Melbourne, VIC, Australia
| | - Colleen J Thomas
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University Melbourne, VIC, Australia
| | - Sonam Parakh
- Department of Biomedical Sciences, Faculty of Medicine and Human Science, Macquarie University Sydney, NSW, Australia
| | - Damian M Spencer
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Melbourne, VIC, Australia
| | - Julie D Atkin
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe UniversityMelbourne, VIC, Australia; Department of Biomedical Sciences, Faculty of Medicine and Human Science, Macquarie UniversitySydney, NSW, Australia
| |
Collapse
|
8
|
Revilla S, Ursulet S, Álvarez-López MJ, Castro-Freire M, Perpiñá U, García-Mesa Y, Bortolozzi A, Giménez-Llort L, Kaliman P, Cristòfol R, Sarkis C, Sanfeliu C. Lenti-GDNF gene therapy protects against Alzheimer's disease-like neuropathology in 3xTg-AD mice and MC65 cells. CNS Neurosci Ther 2014; 20:961-72. [PMID: 25119316 DOI: 10.1111/cns.12312] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2014] [Revised: 07/17/2014] [Accepted: 07/18/2014] [Indexed: 02/01/2023] Open
Abstract
AIMS Glial cell-derived neurotrophic factor (GDNF) is emerging as a potent neurotrophic factor with therapeutic potential against a range of neurodegenerative conditions including Alzheimer's disease (AD). We assayed the effects of GDNF treatment in AD experimental models through gene-therapy procedures. METHODS Recombinant lentiviral vectors were used to overexpress GDNF gene in hippocampal astrocytes of 3xTg-AD mice in vivo, and also in the MC65 human neuroblastoma that conditionally overexpresses the 99-residue carboxyl-terminal (C99) fragment of the amyloid precursor protein. RESULTS After 6 months of overexpressing GDNF, 10-month-old 3xTg-AD mice showed preserved learning and memory, while their counterparts transduced with a green fluorescent protein vector showed cognitive loss. GDNF therapy did not significantly reduce amyloid and tau pathology, but rather, induced a potent upregulation of brain-derived neurotrophic factor that may act in concert with GDNF to protect neurons from atrophy and degeneration. MC65 cells overexpressing GDNF showed an abolishment of oxidative stress and cell death that was at least partially mediated by a reduced presence of intracellular C99 and derived amyloid β oligomers. CONCLUSIONS GDNF induced neuroprotection in the AD experimental models used. Lentiviral vectors engineered to overexpress GDNF showed to be safe and effective, both as a potential gene therapy and as a tool to uncover the mechanisms of GDNF neuroprotection, including cross talk between astrocytes and neurons in the injured brain.
Collapse
Affiliation(s)
- Susana Revilla
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC, Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Crosstalk between Endoplasmic Reticulum Stress and Protein Misfolding in Neurodegenerative Diseases. ACTA ACUST UNITED AC 2013. [DOI: 10.1155/2013/256404] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Under physiological conditions, the endoplasmic reticulum (ER) is a central subcellular compartment for protein quality control in the secretory pathway that prevents protein misfolding and aggregation. Instrumental in protein quality control in the ER is the unfolded protein response (UPR), which is activated upon ER stress to reestablish homeostasis through a sophisticated transcriptionally and translationally regulated signaling network. However, this response can lead to apoptosis if the stress cannot be alleviated. The presence of abnormal protein aggregates containing specific misfolded proteins is recognized as the basis of numerous human conformational disorders, including neurodegenerative diseases. Here, I will highlight the overwhelming evidence that the presence of specific aberrant proteins in Alzheimer’s disease (AD), Parkinson’s disease (PD), Huntington’s disease (HD), prion diseases, and Amyotrophic Lateral Sclerosis (ALS) is intimately associated with perturbations in the ER protein quality control machinery that become incompetent to restore protein homeostasis and shift adaptive programs toward the induction of apoptotic signaling to eliminate irreversibly damaged neurons. Increasing our understanding about the deadly crosstalk between ER dysfunction and protein misfolding in these neurodegenerative diseases may stimulate the development of novel therapeutic strategies able to support neuronal survival and ameliorate disease progression.
Collapse
|
10
|
Cornejo VH, Hetz C. The unfolded protein response in Alzheimer’s disease. Semin Immunopathol 2013; 35:277-92. [DOI: 10.1007/s00281-013-0373-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 03/13/2013] [Indexed: 01/05/2023]
|
11
|
Yang R, Chen L, Wang H, Xu B, Tomimoto H, Chen L. Anti-amnesic effect of neurosteroid PREGS in Aβ25-35-injected mice through σ1 receptor- and α7nAChR-mediated neuroprotection. Neuropharmacology 2012; 63:1042-50. [PMID: 22884465 DOI: 10.1016/j.neuropharm.2012.07.035] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 07/16/2012] [Accepted: 07/19/2012] [Indexed: 11/30/2022]
Abstract
A single intracerebroventricular injection of β-amyloid 25-35 peptide (Aβ(25-35)) (9 nmol/mouse) induces the spatial cognitive deterioration and approximately 50% loss of pyramidal cells in hippocampal CA1 region within 1 week. The present study focused on exploring the effects of neurosteroid pregnenolone sulfate (PREGS), in comparison with the selective agonists of sigma-1 receptor (σ(1)R) and α7 nicotinic acetylcholine receptor (α7nAChR), on the cognitive deficits and the death of pyramidal cells in Aβ(25-35)-mice. Herein, we reported that the administration of PREGS (1-100 mg/kg) for 7 days after Aβ(25-35)-injection could dose-dependently ameliorate the cognitive deficits and attenuate the apoptosis of pyramidal cells. Either the σ(1)R antagonist NE100 or the α7nAChR antagonist MLA could block the neuroprotection of PREGS in Aβ(25-35)-mice. Both the σ(1)R agonist PRE084 and the α7nAChR agonist DMXB could mimic the PREGS-neuroprotection against the Aβ(25-35)-neurotoxicity. The neuroprotection of PRE084 was attenuated by MLA, but the DMXB-action was insensitive to NE100. The neuroprotection of PREGS, PRE084 or DMXB was blocked by the phosphatidylinositol-3-kinase (PI3K) inhibitor LY294002, whereas only the effect of PREGS or PRE084 was sensitive to the MAPK/ERK kinase (MEK) inhibitor U0126. PREGS prevented Aβ(25-35)-inhibited Akt (Serine/threonine kinase) phosphorylation leading to increase in caspase-3 activity, which was σ(1)R- and α7nAChR-dependent. By contrast, PREGS-rescued reduction of extracellular signal-related kinase-2 (ERK2) phosphorylation in Aβ(25-35)-mice only required the activation of σ(1)R. Blockage of PREGS-neuroprotection by LY294002 significantly attenuated its anti-amnesic effect in Aβ(25-35)-mice. The findings indicate that the anti-amnesic effects of PREGS in Aβ(25-35)-mice depend on the σ(1)R- and α7nAChR-mediated neuroprotection.
Collapse
Affiliation(s)
- Rong Yang
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | | | | | | | | | | |
Collapse
|
12
|
Matus S, Glimcher LH, Hetz C. Protein folding stress in neurodegenerative diseases: a glimpse into the ER. Curr Opin Cell Biol 2011; 23:239-52. [PMID: 21288706 DOI: 10.1016/j.ceb.2011.01.003] [Citation(s) in RCA: 178] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 01/05/2011] [Accepted: 01/11/2011] [Indexed: 01/19/2023]
Abstract
Several neurodegenerative diseases share common neuropathology, primarily featuring the presence in the brain of abnormal protein inclusions containing specific misfolded proteins. Recent evidence indicates that alteration in organelle function is a common pathological feature of protein misfolding disorders, highlighting perturbations in the homeostasis of the endoplasmic reticulum (ER). Signs of ER stress have been detected in most experimental models of neurological disorders and more recently in brain samples from human patients with neurodegenerative disease. To cope with ER stress, cells activate an integrated signaling response termed the unfolded protein response (UPR), which aims to reestablish homeostasis in part through regulation of genes involved in protein folding, quality control and degradation pathways. Here we discuss the particular mechanisms currently proposed to be involved in the generation of protein folding stress in different neurodegenerative conditions and speculate about possible therapeutic interventions.
Collapse
Affiliation(s)
- Soledad Matus
- Center for Molecular Studies of Cell, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | | | | |
Collapse
|
13
|
Granholm AC, Zaman V, Godbee J, Smith M, Ramadan R, Umphlet C, Randall P, Bhat NR, Rohrer B, Middaugh LD, Boger HA. Prenatal LPS increases inflammation in the substantia nigra of Gdnf heterozygous mice. Brain Pathol 2010; 21:330-48. [PMID: 20969653 DOI: 10.1111/j.1750-3639.2010.00457.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Prenatal systemic inflammation has been implicated in neurological diseases, but optimal animal models have not been developed. We investigated whether a partial genetic deletion of glial cell line-derived neurotrophic factor (Gdnf(+/-)) increased vulnerability of dopamine (DA) neurons to prenatal lipopolysaccharide (LPS). LPS [0.01 mg/kg intraperitoneal (i.p.)] or saline was administered to wild-type (WT) or Gdnf(+/-) pregnant mice on gestational day 9.5. Male offspring were examined at 3 weeks, 3 and 12 months of age. There was a progressive degeneration of tyrosine hydroxylase (TH)-positive neurons in the substantia nigra (SN) with age in Gdnf(+/-) but not in WT mice, with no observed effects on locus coeruleus (LC) noradrenergic neurons or DA neurons of the ventral tegmental area. Inflammatory markers were elevated in SN of LPS treated offspring, with exacerbation in Gdnf(+/-) mice. Intracellular accumulation of α-synuclein (α-syn) immunoreactivity in DA neurons of SN was observed in all groups of Gdnf(+/-) and in WT mice with prenatal LPS, with altered distribution between pars reticulata (pr) and pars compacta (pc). The findings suggest that prenatal LPS leads to accelerated neuropathology in the SN with age, and that a partial loss of GDNF exacerbates these effects, providing a novel model for age-related neuropathology of the nigrostriatal DA system.
Collapse
Affiliation(s)
- Ann-Charlotte Granholm
- Department of Neurosciences, Medical University of South Carolina, Center on Aging, Charleston, SC 29425, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Xing B, Xin T, Zhao L, Hunter RL, Chen Y, Bing G. Glial cell line-derived neurotrophic factor protects midbrain dopaminergic neurons against lipopolysaccharide neurotoxicity. J Neuroimmunol 2010; 225:43-51. [PMID: 20471698 DOI: 10.1016/j.jneuroim.2010.04.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2009] [Revised: 04/18/2010] [Accepted: 04/19/2010] [Indexed: 02/01/2023]
Abstract
Aberrant microglia activation causes dopaminergic neuronal loss and nitric oxide produced by microglia plays a critical role in dopaminergic neuronal degeneration. However, no study has determined if GDNF protects dopaminergic neurons via inhibiting nitric oxide generation in Parkinson's disease animal model. We report that GDNF not only reduces lipopolysaccharide-induced degeneration of dopaminergic neurons, suppresses microglia activation and nitric oxide generation, but also reverses the inhibition of phosphoinositide 3-kinase (PI3K) in dopaminergic neurons and microglia. It suggests that the neuroprotective effect of GDNF on dopaminergic neurons may be related to its suppression of microglia activation-mediated nitric oxide via releasing the inhibition of PI3K in both neurons and microglia.
Collapse
Affiliation(s)
- Bin Xing
- Department of Anatomy & Neurobiology, University of Kentucky, Lexington, KY 40536, USA.
| | | | | | | | | | | |
Collapse
|
15
|
Kosuge Y, Sekikawa-Nishida K, Negi H, Ishige K, Ito Y. Characterization of chronic glutamate-mediated motor neuron toxicity in organotypic spinal cord culture prepared from ALS model mice. Neurosci Lett 2009; 454:165-9. [PMID: 19429077 DOI: 10.1016/j.neulet.2009.03.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 03/05/2009] [Accepted: 03/05/2009] [Indexed: 10/21/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neurodegenerative disease characterized by selective loss of motor neurons. Although organotypic spinal slice cultures (OSCs) exposed to inhibitors of glutamate uptake have been used as a model of ALS for screening of potentially therapeutic drugs, little development of such drugs has been achieved. In the present study we attempted to establish OSCs from G93A SOD1 transgenic mice (G93A) and to characterize the specific cell death pathway in motoneurons using glial cell line-derived neurotrophic factor (GDNF) in these mice. In the presence of GDNF, the number of surviving neurons in the OSCs was dramatically increased in both G93A and control mice. Exposure to threo-hydroxyaspartate (THA), a glutamate transport inhibitor, for 14 days induced loss of motoneurons in OSCs in G93A and control mice. In OSCs cultured with GDNF, THA-induced motoneuronal death was significantly inhibited in G93A mice, whereas that in control mice was not significantly affected. Moreover, the cleaved form of caspase-12 was increased after THA in the OSCs in G93A but not in control mice, and the activation of caspase-12 was attenuated by OSCs cultured with GDNF. These results suggest that the pathway responsible for motoneuronal death induced by THA in OSCs in G93A mice involves not only in excitotoxicity but also other mechanisms, and that the caspase-12-dependent ER stress pathway plays a role in spinal neuronal death in G93A mice. Moreover, OSCs prepared from the G93A mouse model of ALS may provide a suitable in vitro drug screening model for ALS.
Collapse
Affiliation(s)
- Yasuhiro Kosuge
- Research Unit of Pharmacology, Department of Clinical Pharmacy, College of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-shi, Chiba 274-8555, Japan
| | | | | | | | | |
Collapse
|
16
|
Osada N, Kosuge Y, Kihara T, Ishige K, Ito Y. Apolipoprotein E-deficient mice are more vulnerable to ER stress after transient forebrain ischemia. Neurochem Int 2009; 54:403-9. [PMID: 19428781 DOI: 10.1016/j.neuint.2009.01.010] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2008] [Revised: 01/15/2009] [Accepted: 01/19/2009] [Indexed: 10/21/2022]
Abstract
Apolipoprotein E-deficient (apoE(-/-)) mice have been shown to have increased vulnerability to neuronal damage induced by cerebral ischemia; however, the mechanism of this increased vulnerability remains unclear. In order to define the role of the apoE protein against ischemia-induced ER stress and cell death, experiments were performed to compare ER stress-associated chaperones and signal proteins in the hippocampus of apoE(-/-) mice to those of WT mice after being subjected to forebrain ischemia and reperfusion. Although neuronal loss in area CA1-CA3 of the hippocampus was observed 3 days after ischemia in both types of mice, the damage in apoE(-/-) mice was more severe. In apoE(-/-) mice, a more extensive increase in 78-kDa glucose-regulated protein (GRP78) was observed after the insult, whereas the level of GRP94 was not changed. The expression of both C/EBP homologous protein (CHOP) and caspase-12 was increased in the hippocampus in both WT and apoE(-/-) mice after ischemia. The increased levels of CHOP in apoE(-/-) mice were significantly higher than those in WT mice, whereas the levels of caspase-12 in the two were comparable. Furthermore, whereas the levels of c-Jun N-terminal kinase (JNK), p-JNK1 and p-JNK2 in WT mice were unchanged after ischemia, they were significantly increased in apoE(-/-) mice 24h and 48h after ischemia. These results suggest that increased vulnerability of the hippocampus to forebrain ischemia and reperfusion in apoE(-/-) mice is at least partly attributable to perturbed induction of an ER chaperone, GRP 94, and enhancement of the CHOP- and JNK-dependent apoptotic pathway in the hippocampus.
Collapse
Affiliation(s)
- Nobuhiro Osada
- Research Unit of Pharmacology, Department of Clinical Pharmacy, College of Pharmacy, Nihon University, 7-7-1 Narashinodai, Funabashi-shi, Chiba 274-8555, Japan
| | | | | | | | | |
Collapse
|
17
|
Drexler HCA. Synergistic apoptosis induction in leukemic cells by the phosphatase inhibitor salubrinal and proteasome inhibitors. PLoS One 2009; 4:e4161. [PMID: 19129918 PMCID: PMC2613525 DOI: 10.1371/journal.pone.0004161] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Accepted: 12/06/2008] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Cells adapt to endoplasmic reticulum (ER)-stress by arresting global protein synthesis while simultaneously activating specific transcription factors and their downstream targets. These processes are mediated in part by the phosphorylation-dependent inactivation of the translation initiation factor eIF2alpha. Following restoration of homeostasis protein synthesis is resumed when the serine/threonine-protein phosphatase PP1 dephosphorylates and reactivates eIF2alpha. Proteasome inhibitors, used to treat multiple myeloma patients evoke ER-stress and apoptosis by blocking the ER-associated degradation of misfolded proteins (ERAD), however, the role of eIF2alpha phosphorylation in leukemic cells under conditions of proteasome inhibitor-mediated ER stress is currently unclear. METHODOLOGY AND PRINCIPAL FINDINGS Bcr-Abl-positive and negative leukemic cell lines were used to investigate the functional implications of PP1-related phosphatase activities on eIF2alpha phosphorylation in proteasome inhibitor-mediated ER stress and apoptosis. Rather unexpectedly, salubrinal, a recently identified PP1 inhibitor capable to protect against ER stress in various model systems, strongly synergized with proteasome inhibitors to augment apoptotic death of different leukemic cell lines. Salubrinal treatment did not affect the phosphorlyation status of eIF2alpha. Furthermore, the proapoptotic effect of salubrinal occurred independently from the chemical nature of the proteasome inhibitor, was recapitulated by a second unrelated phosphatase inhibitor and was unaffected by overexpression of a dominant negative eIF2alpha S51A variant that can not be phosphorylated. Salubrinal further aggravated ER-stress and proteotoxicity inflicted by the proteasome inhibitors on the leukemic cells since characteristic ER stress responses, such as ATF4 and CHOP synthesis, XBP1 splicing, activation of MAP kinases and eventually apoptosis were efficiently abrogated by the translational inhibitor cycloheximide. CONCLUSIONS Although PP1 activity does not play a major role in regulating the ER stress response in leukemic cells, phosphatase signaling nevertheless significantly limits proteasome inhibitor-mediated ER-stress and apoptosis. Inclusion of specific phosphatase inhibitors might therefore represent an option to improve current proteasome inhibitor-based treatment modalities for hematological cancers.
Collapse
Affiliation(s)
- Hannes C A Drexler
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, Muenster, Germany.
| |
Collapse
|
18
|
Pertusa M, García-Matas S, Mammeri H, Adell A, Rodrigo T, Mallet J, Cristòfol R, Sarkis C, Sanfeliu C. Expression of GDNF transgene in astrocytes improves cognitive deficits in aged rats. Neurobiol Aging 2008; 29:1366-79. [PMID: 17399854 DOI: 10.1016/j.neurobiolaging.2007.02.026] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2006] [Revised: 02/14/2007] [Accepted: 02/20/2007] [Indexed: 01/25/2023]
Abstract
Glial cell line-derived neurotrophic factor (GDNF) was assayed for its neurotrophic effects against the neuronal atrophy that causes cognitive deficits in old age. Aged Fisher 344 rats with impairment in the Morris water maze received intrahippocampal injections at the dorsal CA1 area of either a lentiviral vector encoding human GDNF or the same vector encoding human green fluorescent protein as a control. Recombinant lentiviral vectors constructed with human cytomegalovirus promotor and pseudotyped with lyssavirus Mokola glycoprotein specifically transduced the astrocytes in vivo. Astrocyte-secreted GDNF enhanced neuron function as shown by local increases in synthesis of the neurotransmitters acetylcholine, dopamine and serotonin. This neurotrophic effect led to cognitive improvement of the rats as early as 2 weeks after gene transduction. Spatial learning and memory testing showed a significant gain in cognitive abilities due to GDNF exposure, whereas control-transduced rats kept their performance at the chance level. These results confirm the broad spectrum of the neurotrophic action of GDNF and open new gene therapy possibilities for reducing age-related neurodegeneration.
Collapse
Affiliation(s)
- M Pertusa
- Departament de Farmacologia i Toxicologia, Institut d'Investigacions Biomèdiques de Barcelona (IIBB), CSIC-IDIBAPS, Rosselló 161, 08036 Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
He S, Yaung J, Kim YH, Barron E, Ryan SJ, Hinton DR. Endoplasmic reticulum stress induced by oxidative stress in retinal pigment epithelial cells. Graefes Arch Clin Exp Ophthalmol 2008; 246:677-83. [DOI: 10.1007/s00417-008-0770-2] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2007] [Revised: 01/05/2008] [Accepted: 01/10/2008] [Indexed: 01/20/2023] Open
|
20
|
NF-κB activated by ER calcium release inhibits Aβ-mediated expression of CHOP protein: Enhancement by AD-linked mutant presenilin 1. Exp Neurol 2007; 208:169-76. [DOI: 10.1016/j.expneurol.2007.04.009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2006] [Revised: 04/19/2007] [Accepted: 04/24/2007] [Indexed: 02/03/2023]
|
21
|
Li X, Peng C, Li L, Ming M, Yang D, Le W. Glial cell-derived neurotrophic factor protects against proteasome inhibition-induced dopamine neuron degeneration by suppression of endoplasmic reticulum stress and caspase-3 activation. J Gerontol A Biol Sci Med Sci 2007; 62:943-50. [PMID: 17895431 DOI: 10.1093/gerona/62.9.943] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Evidence has shown that ubiquitin proteasome system (UPS) impairment plays an important role in the dopamine (DA) neurodegeneration in Parkinson's disease (PD). It has been reported that application of proteasomal inhibitor lactacystin in ventral mesencephalon (VM) cultures can cause DA neurodegeneration, although the underlying mechanisms are not clear. Herein, we used the lactacystin-induced DA cell degeneration model to study the neuroprotection of glial cell-derived neurotrophic factor (GDNF) in VM cultures. We measured the expression of endoplasmic reticulum stress (ERS)-related genes, and determined the caspase-3 activation, apoptotic cell death, as well as alpha-synuclein-positive inclusions in DA neurons. We found that GDNF treatment significantly suppressed the expression of ERS-related genes and inhibited the activation of caspase-3 and apoptotic cell death without affecting alpha-synuclein-positive inclusions in DA neurons. Our study suggests that the protection of GDNF against DA neurodegeneration in the UPS impairment model is associated with ERS and caspase-3 suppression.
Collapse
Affiliation(s)
- Xuping Li
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences/Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | |
Collapse
|
22
|
Wang X, Wang B, Fan Z, Shi X, Ke ZJ, Luo J. Thiamine deficiency induces endoplasmic reticulum stress in neurons. Neuroscience 2007; 144:1045-56. [PMID: 17137721 PMCID: PMC1819404 DOI: 10.1016/j.neuroscience.2006.10.008] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2006] [Revised: 10/04/2006] [Accepted: 10/05/2006] [Indexed: 10/23/2022]
Abstract
Thiamine (vitamin B1) deficiency (TD) causes region selective neuronal loss in the brain; it has been used to model neurodegeneration that accompanies mild impairment of oxidative metabolism. The mechanisms for TD-induced neurodegeneration remain incompletely elucidated. Inhibition of protein glycosylation, perturbation of calcium homeostasis and reduction of disulfide bonds provoke the accumulation of unfolded proteins in the endoplasmic reticulum (ER), and cause ER stress. Recently, ER stress has been implicated in a number of neurodegenerative models. We demonstrated here that TD up-regulated several markers of ER stress, such as glucose-regulated protein (GRP) 78, growth arrest and DNA-damage inducible protein or C/EBP-homologus protein (GADD153/Chop), phosphorylation of eIF2alpha and cleavage of caspase-12 in the cerebellum and the thalamus of mice. Furthermore, ultrastructural analysis by electron microscopic study revealed an abnormality in ER structure. To establish an in vitro model of TD in neurons, we treated cultured cerebellar granule neurons (CGNs) with amprolium, a potent inhibitor of thiamine transport. Exposure to amprolium caused apoptosis and the generation of reactive oxygen species in CGNs. Similar to the observation in vivo, TD up-regulated markers for ER stress. Treatment of a selective inhibitor of caspase-12 significantly alleviated amprolium-induced death of CGNs. Thus, ER stress may play a role in TD-induced brain damage.
Collapse
Affiliation(s)
- X Wang
- Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, PR China
| | | | | | | | | | | |
Collapse
|
23
|
Suzumura A, Takeuchi H, Zhang G, Kuno R, Mizuno T. Roles of Glia-Derived Cytokines on Neuronal Degeneration and Regeneration. Ann N Y Acad Sci 2006; 1088:219-29. [PMID: 17192568 DOI: 10.1196/annals.1366.012] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Accumulation of activated microglia and reactive astrocytes is observed around degenerating neurons in various inflammatory or degenerative disorders in the central nervous system. These reactive glial cells may play either neurotoxic or neuroprotective roles. In this study, we examined the effects of glia-derived cytokines on neuronal degeneration and regeneration. Neuron-rich cultures were stimulated with supernatant of microglia and astrocytes stimulated with LPS, or a various concentrations of recombinant cytokines. Neurotoxicity was evaluated by an MTS assay. Neuronal damage was also evaluated by a frequency of dendritic beading, which was found to be an early feature of neuronal damage toward cell death. Effects of the cytokines on production of neurotrophic factors by astrocytes were also examined by RT-PCR for the expression of mRNA. Supernatant of LPS-stimulated microglia induced neuronal cell death. However, all the recombinant cytokines examined did not induce cell death, while IFNgamma and TNFalpha induced dendrite beading, an early feature of neuronal damage. IL-1beta and TNFalpha enhanced the production of neurotrophic factors by astrocytes. These observations suggest that glial cell-derived cytokines may synergistically function in neuronal degeneration with other toxic factors produced by activated microglia, and that some of them may also function in regeneration by inducing neurotrophic factors.
Collapse
Affiliation(s)
- Akio Suzumura
- Department of Neuroimmunology, Research Institute of Environmental Medicine, Nagoya University, Furo-cho, Chikusa, Nagoya 464-8601, Japan.
| | | | | | | | | |
Collapse
|
24
|
Meunier J, Ieni J, Maurice T. The anti-amnesic and neuroprotective effects of donepezil against amyloid beta25-35 peptide-induced toxicity in mice involve an interaction with the sigma1 receptor. Br J Pharmacol 2006; 149:998-1012. [PMID: 17057756 PMCID: PMC2014636 DOI: 10.1038/sj.bjp.0706927] [Citation(s) in RCA: 235] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND AND PURPOSE The acetylcholinesterase inhibitor, donepezil, is also a high affinity sigma(1) receptor agonist. We examined the involvement of sigma(1) receptors in its anti-amnesic and neuroprotective properties against amyloid beta(25-35) peptide-induced toxicity in mice. EXPERIMENTAL APPROACH Mice were given an intracerebroventricular (i.c.v.) injection of Abeta(25-35) peptide (9 nmol) 7-9 days before being tested for spontaneous alternation and passive avoidance. Hippocampal lipid peroxidation was measured 7 days after Abeta(25-35) injection to evaluate oxidative stress. Donepezil, the sigma(1) agonist PRE-084 or the cholinesterase (ChE) inhibitors tacrine, rivastigmine and galantamine were administered either 20 min before behavioural sessions to check their anti-amnesic effects, or 20 min before Abeta(25-35) injection, or 24 h after Abeta(25-35) injection and then once daily before behavioural sessions, to check their pre- and post-i.c.v. neuroprotective activity, respectively. KEY RESULTS All the drugs tested were anti-amnesic, but only the effects of PRE-084 and donepezil were prevented by the sigma(1) antagonist BD1047. Only PRE-084 and donepezil showed neuroprotection when administered pre i.c.v.; they blocked lipid peroxidation and learning deficits, effects inhibited by BD1047. Post i.c.v., PRE-084 and donepezil showed complete neuroprotection whereas the other ChE inhibitors showed partial effects. BD1047 blocked these effects of PRE-084, attenuated those of donepezil, but did not affect the partial effects of the other ChE inhibitors. CONCLUSIONS AND IMPLICATIONS The potent anti-amnesic and neuroprotective effects of donepezil against Abeta(25-35)-induced toxicity involve both its cholinergic and sigma(1) agonistic properties. This dual action may explain its sustained activity compared to other ChE inhibitors.
Collapse
|
25
|
Yu MS, Suen KC, Kwok NS, So KF, Hugon J, Chang RCC. Beta-amyloid peptides induces neuronal apoptosis via a mechanism independent of unfolded protein responses. Apoptosis 2006; 11:687-700. [PMID: 16532272 DOI: 10.1007/s10495-006-5540-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Accumulation of beta-amyloid (Abeta) peptides in senile plaques is one of the pathological hallmarks in Alzheimer's disease (AD), which can trigger apoptosis. We have previously demonstrated that Abeta triggered calcium release from the ER. Depletion of ER Ca(2+) ions has been reported leading to unfolded protein responses (UPR). While hypothesis has been made about UPR and neurodegeneration in AD, little is known about the effects of extracellular accumulation of Abeta on UPR. We have shown previously that activation of PKR in Abeta-triggered apoptosis. Since UPR can trigger PKR, our study aims to elucidate whether extracellular accumulation of Abeta peptides induce UPR in cultured neurons. Our results showed that Abeta could not trigger UPR signalings including phosphorylation of PERK, alternative cleavage of xbp-1 mRNA and induction of transcription of xbp-1 and Gadd153. Taken together, our results suggest that extracellular accumulation of Abeta peptides induce apoptosis via a mechanism independent of UPR.
Collapse
Affiliation(s)
- Man-Shan Yu
- Laboratory of Neurodegenerative Diseases, Department of Anatomy, Research Centre of Heart, Brain, Hormone and Healthy Aging, Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong SAR
| | | | | | | | | | | |
Collapse
|
26
|
Kyuhou SI, Kato N, Gemba H. Emergence of endoplasmic reticulum stress and activated microglia in Purkinje cell degeneration mice. Neurosci Lett 2005; 396:91-6. [PMID: 16356646 DOI: 10.1016/j.neulet.2005.11.023] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2005] [Revised: 11/03/2005] [Accepted: 11/07/2005] [Indexed: 10/25/2022]
Abstract
In the current studies, we characterized the molecular and cellular mechanism of cell death in Purkinje cell degeneration (pcd) mice using real-time quantitative PCR, immunohistochemistry, and Western blotting. It appears that endoplasmic reticulum (ER) stress is involved in this degeneration of Purkinje cells because ER stress-related substrates, such as CHOP and caspase 12, were strongly activated in Purkinje cells of pcd mice during the third postnatal (P) week. A significant increase in the expression of the ER-specific chaperone BiP suggested that unfolded protein responses were induced. We also found that Purkinje cells underwent apoptosis via the activation of caspase 3 and subsequent fragmentation of DNA. In addition to the activation of apoptosis in Purkinje cells, many activated microglial cells are found to be present in the molecular layer of the cerebellar cortex. In the later phase of degeneration, there was conspicuous expression of inducible nitric oxide synthase (iNOS), and some Purkinje cells were strongly labeled with an antibody to nitrotyrosine, suggesting that Purkinje cells in pcd mice are damaged by nitric oxide released from microglial cells. Administration of minocycline, which may inhibit iNOS expression, delayed the death of Purkinje cells in pcd mice and mildly improved their motor abilities. These findings suggest that ER stress participates in the degeneration of Purkinje cells and that activation of microglia accelerates Purkinje cell death in pcd mice.
Collapse
Affiliation(s)
- Shin-ichi Kyuhou
- Department Physiology (II), Kansai Medical University, 10-15 Fumizono-cho, Moriguchi 570 8506, Japan.
| | | | | |
Collapse
|
27
|
Caraci F, Chisari M, Frasca G, Canonico PL, Battaglia A, Calafiore M, Battaglia G, Bosco P, Nicoletti F, Copani A, Sortino MA. Nicergoline, a drug used for age-dependent cognitive impairment, protects cultured neurons against beta-amyloid toxicity. Brain Res 2005; 1047:30-7. [PMID: 15882840 DOI: 10.1016/j.brainres.2005.04.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2004] [Revised: 03/29/2005] [Accepted: 04/01/2005] [Indexed: 11/25/2022]
Abstract
Nicergoline, a drug used for the treatment of Alzheimer's disease and other types of dementia, was tested for its ability to protect neurons against beta-amyloid toxicity. Pure cultures of rat cortical neurons were challenged with a toxic fragment of beta-amyloid peptide (betaAP(25-35)) and toxicity was assessed after 24 h. Micromolar concentrations of nicergoline or its metabolite, MDL, attenuated betaAP(25-35)-induced neuronal death, whereas MMDL (another metabolite of nicergoline), the alpha1-adrenergic receptor antagonist, prazosin, or the serotonin 5HT-2 receptor antagonist, methysergide, were inactive. Nicergoline increased the basal levels of Bcl-2 and reduced the increase in Bax levels induced by beta-amyloid, indicating that the drug inhibits the execution of an apoptotic program in cortical neurons. In mixed cultures of rat cortical cells containing both neurons and astrocytes, nicergoline and MDL were more efficacious than in pure neuronal cultures in reducing beta-amyloid neurotoxicity. Experiments carried out in pure cultures of astrocytes showed that a component of neuroprotection was mediated by a mechanism of glial-neuronal interaction. The conditioned medium of cultured astrocytes treated with nicergoline or MDL for 72-96 h (collected 24 h after drug withdrawal) was neuroprotective when transferred to pure neuronal cultures challenged with beta-amyloid. In cultured astrocytes, nicergoline increased the intracellular levels of transforming-growth factor-beta and glial-derived neurotrophic factor, two trophic factors that are known to protect neurons against beta-amyloid toxicity. These results raise the possibility that nicergoline reduces neurodegeneration in the Alzheimer's brain.
Collapse
Affiliation(s)
- Filippo Caraci
- Department of Pharmaceutical Sciences, University of Catania, Viale A. Doria, 6, 95125, Catania, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Gebicke-Haerter PJ. Microarrays and expression profiling in microglia research and in inflammatory brain disorders. J Neurosci Res 2005; 81:327-41. [PMID: 15948185 DOI: 10.1002/jnr.20479] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Expression profiling by using microarrays is a powerful tool for investigating transcriptional changes in a variety of diseases. In this survey, microarray data selected from the literature from in vivo and in vitro studies are scrutinized to find differentially expressed genes in common within specific inflammatory conditions in brain or microglial cell cultures, if there are at least two independent investigations available. Viral encephalitis, multiple sclerosis, epileptic seizures, ischemic lesions, and traumatic brain injury are the disorders covered. Moreover, by taking into account expression data obtained from cultured microglia, two examples are presented of how one can deal (or should not deal) with lists of candidate genes showing up in these kinds of studies without sophisticated software programs. Finally, some general remarks are made about pivotal issues when beginning to use microarray technology.
Collapse
|