1
|
Sun J, Lee K, Kutseikin S, Guerrero A, Rius B, Madhavan A, Buasakdi C, Cheong KN, Chatterjee P, Rosen DA, Yoon L, Ardejani MS, Mendoza A, Rosarda JD, Saez E, Kelly JW, Wiseman RL. Identification of a Selective Pharmacologic IRE1/XBP1s Activator with Enhanced Tissue Exposure. ACS Chem Biol 2025; 20:993-1003. [PMID: 40231944 DOI: 10.1021/acschembio.4c00867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Activation of the IRE1/XBP1s signaling arm of the unfolded protein response (UPR) has emerged as a promising strategy to mitigate etiologically diverse diseases. Despite this promise, few compounds are available to selectively activate IRE1/XBP1s signaling to probe the biologic and therapeutic implications of this pathway in human disease. Recently, we identified the compound IXA4 as a highly selective activator of protective IRE1/XBP1s signaling. While IXA4 has proven useful for increasing IRE1/XBP1s signaling in cultured cells and mouse liver, the utility of this compound is restricted by its limited activity in other tissues. To broaden our ability to pharmacologically interrogate the impact of IRE1/XBP1s signaling in vivo, we sought to identify IRE1/XBP1s activators with greater tissue activity than IXA4. We reanalyzed 'hits' from the high throughput screen used to identify IXA4, selecting compounds from structural classes not previously pursued. We then performed global RNAseq to confirm that these compounds showed transcriptome-wide selectivity for IRE1/XBP1s activation. Functional profiling revealed compound IXA62 as a selective IRE1/XBP1s activator that reduced Aβ secretion from CHO7PA2 cells and enhanced glucose-stimulated insulin secretion from rat insulinoma cells, mimicking the effects of IXA4 in these assays. IXA62 robustly and selectively activated IRE1/XBP1s signaling in the liver of mice dosed compound intraperitoneally or orally. In treated mice, IXA62 showed broader tissue activity, relative to IXA4, inducing expression of IRE1/XBP1s target genes in additional tissues such as kidney and lung. Collectively, our results designate IXA62 as a selective IRE1/XBP1s signaling activating compound with enhanced tissue activity, which increases our ability to pharmacologically probe the biologic significance and potential therapeutic utility of enhancing adaptive IRE1/XBP1s signaling in vivo.
Collapse
Affiliation(s)
- Jie Sun
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Kyunga Lee
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
| | - Sergei Kutseikin
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Adrian Guerrero
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
| | - Bibiana Rius
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Aparajita Madhavan
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Chavin Buasakdi
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Ka-Neng Cheong
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
- Department of Immunology and Microbial Science, Scripps Research, La Jolla, California 92037, United States
| | - Priyadarshini Chatterjee
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Dorian A Rosen
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Leonard Yoon
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
| | - Maziar S Ardejani
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
| | - Alejandra Mendoza
- Department of Immunology and Microbial Science, Scripps Research, La Jolla, California 92037, United States
| | - Jessica D Rosarda
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Enrique Saez
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| | - Jeffery W Kelly
- Department of Chemistry, Scripps Research, La Jolla, California 92037, United States
- The Skaggs Institute for Chemical Biology, Scripps Research, La Jolla, California 92037, United States
| | - R Luke Wiseman
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, California 92037, United States
| |
Collapse
|
2
|
Valenzuela V, Becerra D, Astorga JI, Fuentealba M, Diaz G, Bargsted L, Chacón C, Martinez A, Gozalvo R, Jackson K, Morales V, Heras ML, Tamburini G, Petrucelli L, Sardi SP, Plate L, Hetz C. Artificial enforcement of the unfolded protein response reduces disease features in multiple preclinical models of ALS/FTD. Mol Ther 2025; 33:1226-1245. [PMID: 39799393 PMCID: PMC11897772 DOI: 10.1016/j.ymthe.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 06/05/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025] Open
Abstract
Amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) are part of a spectrum of diseases that share several causative genes, resulting in a combinatory of motor and cognitive symptoms and abnormal protein aggregation. Multiple unbiased studies have revealed that proteostasis impairment at the level of the endoplasmic reticulum (ER) is a transversal pathogenic feature of ALS/FTD. The transcription factor XBP1s is a master regulator of the unfolded protein response (UPR), the main adaptive pathway to cope with ER stress. Here, we provide evidence of suboptimal activation of the UPR in ALS/FTD models under experimental ER stress. To artificially engage the UPR, we intracerebroventricularly administrated adeno-associated viruses (AAVs) to express the active form of XBP1 (XBP1s) in the nervous system of ALS/FTD models. XBP1s expression improved motor performance and extended lifespan of mutant SOD1 mice, associated with reduced protein aggregation. AAV-XBP1s administration also attenuated disease progression in models of TDP-43 and C9orf72 pathogenesis. Proteomic profiling of spinal cord tissue revealed that XBP1s overexpression improved proteostasis and modulated the expression of a cluster of synaptic and cell morphology proteins. Our results suggest that strategies to improve ER proteostasis may serve as a pan-therapeutic strategy to treat ALS/FTD.
Collapse
Affiliation(s)
- Vicente Valenzuela
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; Biomedical Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Daniela Becerra
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; Biomedical Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - José I Astorga
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; Biomedical Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Matías Fuentealba
- FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, USA
| | - Guillermo Diaz
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; Biomedical Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Leslie Bargsted
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Carlos Chacón
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; Biomedical Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Alexis Martinez
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; Biomedical Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Romina Gozalvo
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; Biomedical Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | | | - Vania Morales
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; Biomedical Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Macarena Las Heras
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; Biomedical Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Giovanni Tamburini
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; Biomedical Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Leonard Petrucelli
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Mayo Graduate School, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Lars Plate
- Department of Chemistry and Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Claudio Hetz
- Program of Cellular and Molecular Biology, Biomedical Sciences Institute (ICBM), Universidad de Chile, Santiago, Chile; Biomedical Neuroscience, Faculty of Medicine, Universidad de Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, USA.
| |
Collapse
|
3
|
Zhao X, Hu X, Wang W, Lu S. Macrophages dying from ferroptosis promote microglia-mediated inflammatory responses during spinal cord injury. Int Immunopharmacol 2024; 143:113281. [PMID: 39357207 DOI: 10.1016/j.intimp.2024.113281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/15/2024] [Accepted: 09/26/2024] [Indexed: 10/04/2024]
Abstract
The neurological deficits following traumatic spinal cord injury are associated with severe patient disability and economic consequences. Currently, an increasing number of studies are focusing on the importance of ferroptosis during acute organ injuries. However, the spatial and temporal distribution patterns of ferroptosis during SCI and the details of its role are largely unknown. In this study, in vivo experiments revealed that microglia are in close proximity to macrophages, the major cell type that undergoes ferroptosis following SCI. Furthermore, we found that ferroptotic macrophages aggravate SCI by inducing the proinflammatory properties of microglia. In vitro studies further revealed ferroptotic macrophages increased the expression of IL-1β, IL-6, and IL-23 in microglia. Mechanistically, due to the activation of the NF-κB signaling pathway, the expression of IL-1β and IL-6 was increased. In addition, we established that increased levels of oxidative phosphorylation cause mitochondrial reactive oxygen species generation and unfolded protein response activation and trigger an inflammatory response marked by an increase in IL-23 production. Our findings identified that targeting ferroptosis and IL-23 could be an effective strategy for promoting neurological recovery after SCI.
Collapse
Affiliation(s)
- Xuan Zhao
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Xinli Hu
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Diseases, Beijing, China
| | - Wei Wang
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Diseases, Beijing, China.
| | - Shibao Lu
- Department of Orthopedics, Xuanwu Hospital, Capital Medical University, Beijing, China; National Clinical Research Center for Geriatric Diseases, Beijing, China.
| |
Collapse
|
4
|
Chiang W, Urban JM, Yanchik-Slade F, Stout A, Hammond JM, Nilsson BL, Gelbard HA, Krauss TD. Hybrid Amyloid Quantum Dot Nano-Bio Assemblies to Probe Neuroinflammatory Damage. ACS Chem Neurosci 2024; 15:3124-3135. [PMID: 39146244 PMCID: PMC11378299 DOI: 10.1021/acschemneuro.4c00183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/29/2024] [Accepted: 08/07/2024] [Indexed: 08/17/2024] Open
Abstract
Various oligomeric species of amyloid-beta have been proposed to play different immunogenic roles in the cellular pathology of Alzheimer's Disease. The dynamic interconversion between various amyloid oligomers and fibrillar assemblies makes it difficult to elucidate the role each potential aggregation state may play in driving neuroinflammatory and neurodegenerative pathology. The ability to identify the amyloid species that are key and essential drivers of these pathological hallmarks of Alzheimer's Disease is of fundamental importance for also understanding downstream events including tauopathies that mediate neuroinflammation with neurologic deficits. Here, we report the design and construction of a quantum dot mimetic for larger spherical oligomeric amyloid species as an "endogenously" fluorescent proxy for this cytotoxic assembly of amyloid to investigate its role in inducing inflammatory and stress response states in neuronal and glial cell types. The design parameters and construction protocol developed here may be adapted for developing quantum dot nano-bio assemblies for other biological systems of interest, particularly neurodegenerative diseases involving other protein aggregates.
Collapse
Affiliation(s)
- Wesley Chiang
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
- Department
of Biochemistry and Biophysics, University
of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jennifer M. Urban
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
| | - Francine Yanchik-Slade
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
| | - Angela Stout
- Center
for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Jennetta M. Hammond
- Center
for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Bradley L. Nilsson
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
| | - Harris A. Gelbard
- Center
for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, New York 14642, United States
- Departments
of Pediatrics, Neuroscience, and Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York 14642, United States
| | - Todd D. Krauss
- Department
of Chemistry, University of Rochester, Rochester, New York 14627-0216, United
States
- The
Institute of Optics, University of Rochester
Medical Center, Rochester, New York 14627-0216, United States
| |
Collapse
|
5
|
Liu Y, Cui F, Xu A, Wang B, Ma Y, Zhang Q, Sun Q, Zheng Y, Xue Y, Sun Y, Bian L. Interaction Between the PERK/ATF4 Branch of the Endoplasmic Reticulum Stress and Mitochondrial One-Carbon Metabolism Regulates Neuronal Survival After Intracerebral Hemorrhage. Int J Biol Sci 2024; 20:4277-4296. [PMID: 39247810 PMCID: PMC11379068 DOI: 10.7150/ijbs.93787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 07/21/2024] [Indexed: 09/10/2024] Open
Abstract
Recent investigations have revealed that oxidative stress can lead to neuronal damage and disrupt mitochondrial and endoplasmic reticulum functions after intracerebral hemorrhage (ICH). However, there is limited evidence elucidating their role in maintaining neuronal homeostasis. Metabolomics analysis, RNA sequencing, and CUT&Tag-seq were performed to investigate the mechanism underlying the interaction between the PERK/ATF4 branch of the endoplasmic reticulum stress (ERS) and mitochondrial one-carbon (1C) metabolism during neuronal resistance to oxidative stress. The association between mitochondrial 1C metabolism and the PERK/ATF4 branch of the ERS after ICH was investigated using transcription factor motif analysis and co-immunoprecipitation. The findings revealed interactions between the GRP78/PERK/ATF4 and mitochondrial 1C metabolism, which are important in preserving neuronal homeostasis after ICH. ATF4 is an upstream transcription factor that directly regulates the expression of 1C metabolism genes. Additionally, the GRP78/PERK/ATF4 forms a negative regulatory loop with MTHFD2 because of the interaction between GRP78 and MTHFD2. This study presents evidence of disrupted 1C metabolism and the occurrence of ERS in neurons post-ICH. Supplementing exogenous NADPH or interfering with the PERK/ATF4 could reduce symptoms related to neuronal injuries, suggesting new therapeutic prospects for ICH.
Collapse
Affiliation(s)
- Yikui Liu
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fengzhen Cui
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
- School of Public Health, Guangdong Medical University, Dongguan, China
| | - Aoqian Xu
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Baofeng Wang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuxiao Ma
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qixiang Zhang
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingfang Sun
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongtao Zheng
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuxiao Xue
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhao Sun
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liuguan Bian
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Arcos J, Grunenwald F, Sepulveda D, Jerez C, Urbina V, Huerta T, Troncoso-Escudero P, Tirado D, Perez A, Diaz-Espinoza R, Nova E, Kubitscheck U, Rodriguez-Gatica JE, Hetz C, Toledo J, Ahumada P, Rojas-Rivera D, Martín-Montañez E, Garcia-Fernandez M, Vidal RL. IGF2 prevents dopaminergic neuronal loss and decreases intracellular alpha-synuclein accumulation in Parkinson's disease models. Cell Death Discov 2023; 9:438. [PMID: 38042807 PMCID: PMC10693583 DOI: 10.1038/s41420-023-01734-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/05/2023] [Accepted: 11/21/2023] [Indexed: 12/04/2023] Open
Abstract
Parkinson's disease (PD) is the second most common late-onset neurodegenerative disease and the predominant cause of movement problems. PD is characterized by motor control impairment by extensive loss of dopaminergic neurons in the substantia nigra pars compacta (SNpc). This selective dopaminergic neuronal loss is in part triggered by intracellular protein inclusions called Lewy bodies, which are composed mainly of misfolded alpha-synuclein (α-syn) protein. We previously reported insulin-like growth factor 2 (IGF2) as a key protein downregulated in PD patients. Here we demonstrated that IGF2 treatment or IGF2 overexpression reduced the α-syn aggregates and their toxicity by IGF2 receptor (IGF2R) activation in cellular PD models. Also, we observed IGF2 and its interaction with IGF2R enhance the α-syn secretion. To determine the possible IGF2 neuroprotective effect in vivo we used a gene therapy approach in an idiopathic PD model based on α-syn preformed fibrils intracerebral injection. IGF2 gene therapy revealed a significantly preventing of motor impairment in idiopathic PD model. Moreover, IGF2 expression prevents dopaminergic neuronal loss in the SN together with a decrease in α-syn accumulation (phospho-α-syn levels) in the striatum and SN brain region. Furthermore, the IGF2 neuroprotective effect was associated with the prevention of synaptic spines loss in dopaminergic neurons in vivo. The possible mechanism of IGF2 in cell survival effect could be associated with the decrease of the intracellular accumulation of α-syn and the improvement of dopaminergic synaptic function. Our results identify to IGF2 as a relevant factor for the prevention of α-syn toxicity in both in vitro and preclinical PD models.
Collapse
Affiliation(s)
- Javiera Arcos
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Felipe Grunenwald
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Denisse Sepulveda
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Carolina Jerez
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Valentina Urbina
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Tomas Huerta
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Paulina Troncoso-Escudero
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Molecular Diagnostic and Biomarkers Laboratory, Department of Pathology, Faculty of Medicine Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Daniel Tirado
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Escuela de Tecnología Médica, Universidad Mayor, Santiago, Chile
| | - Angela Perez
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
- Escuela de Tecnología Médica, Universidad Mayor, Santiago, Chile
| | - Rodrigo Diaz-Espinoza
- Departamento de Biología, Facultad de Química y Biología, Universidad de Santiago de Chile, Santiago, Chile
| | - Esteban Nova
- Departamento de Química, Facultad de Ciencias Naturales, Matemáticas y Medio Ambiente, Universidad Tecnológica Metropolitana, Santiago, Chile
| | - Ulrich Kubitscheck
- Clausius Institute of Physical and Theoretical Chemistry, University of Bonn, Bonn, Germany
| | | | - Claudio Hetz
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Jorge Toledo
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile
| | - Pablo Ahumada
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile
| | - Diego Rojas-Rivera
- Escuela de Tecnología Médica, Universidad Mayor, Santiago, Chile
- Escuela de Biotecnología, Universidad Mayor, Santiago, Chile
- Center for Biomedicine, Universidad Mayor, Santiago, Chile
| | - Elisa Martín-Montañez
- Department of Pharmacology, Faculty of Medicine, Biomedical Research Institute of Malaga, University of Malaga, Malaga, Spain
| | - María Garcia-Fernandez
- Department of Human Physiology, Faculty of Medicine, Biomedical Research Institute of Malaga, University of Malaga, Malaga, Spain
| | - René L Vidal
- Center for Integrative Biology, Universidad Mayor, Santiago, Chile.
- Biomedical Neuroscience Institute, University of Chile, Santiago, Chile.
- Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.
- Escuela de Tecnología Médica, Universidad Mayor, Santiago, Chile.
- Escuela de Biotecnología, Universidad Mayor, Santiago, Chile.
| |
Collapse
|
7
|
Melnikova A, Ishii H, Tamatani T, Hattori T, Takarada-Iemata M, Hori O. Neuroprotective role of calreticulin after spinal cord injury in mice. Neurosci Res 2023; 195:29-36. [PMID: 37295503 DOI: 10.1016/j.neures.2023.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/09/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023]
Abstract
Accumulating evidence suggests that endoplasmic reticulum (ER) stress and unfolded protein response (UPR) are involved in the pathology of spinal cord injury (SCI). To determine the role of the UPR-target molecule in the pathophysiology of SCI, we analyzed the expression and the possible function of calreticulin (CRT), a molecular chaperone in the ER with high Ca2+ binding capacity, in a mouse SCI model. Spinal cord contusion was induced in T9 by using the Infinite Horizon impactor. Quantitative real-time polymerase chain reaction confirmed increase of Calr mRNA after SCI. Immunohistochemistry revealed that CRT expression was observed mainly in neurons in the control (sham operated) condition, while it was strongly observed in microglia/macrophages after SCI. Comparative analysis between wild-type (WT) and Calr+/- mice revealed that the recovery of hindlimb locomotion was reduced in Calr+/- mice, based on the evaluation using the Basso Mouse Scale and inclined-plane test. Immunohistochemistry also revealed more accumulation of immune cells in Calr+/- mice than in WT mice, at the epicenter 3 days and at the caudal region 7 days after SCI. Consistently, the number of damaged neuron was higher in Calr+/- mice at the caudal region 7 days after SCI. These results suggest a regulatory role of CRT in the neuroinflammation and neurodegeneration after SCI.
Collapse
Affiliation(s)
- Anastasiia Melnikova
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan; Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia
| | - Hiroshi Ishii
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Takashi Tamatani
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Mika Takarada-Iemata
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.
| |
Collapse
|
8
|
Chiang W, Urban JM, Yanchik-Slade F, Stout A, Nilsson BL, Gelbard HA, Krauss TD. Hybrid Amyloid Quantum Dot Nanoassemblies to Probe Neuroinflammatory Damage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.30.555592. [PMID: 37693630 PMCID: PMC10491264 DOI: 10.1101/2023.08.30.555592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
Various oligomeric species of amyloid-beta have been proposed to play different immunogenic roles in the cellular pathology of Alzheimer's Disease. However, investigating the role of a homogenous single oligomeric species has been difficult due to highly dynamic oligomerization and fibril formation kinetics that convert between many species. Here we report the design and construction of a quantum dot mimetic for larger spherical oligomeric amyloid species as an "endogenously" fluorescent proxy for this cytotoxic species to investigate its role in inducing inflammatory and stress response states in neuronal and glial cell types.
Collapse
Affiliation(s)
- Wesley Chiang
- Department of Biochemistry and Biophysics, University of Rochester Medical Center, Rochester, NY, 14642
| | - Jennifer M. Urban
- Department of Chemistry, Rochester, New York 14627-0216, United States
| | | | - Angela Stout
- Center for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, NY, 14642
| | | | - Harris A. Gelbard
- Center for Neurotherapeutics Discovery and Department of Neurology, University of Rochester Medical Center, Rochester, NY, 14642
- Departments of Pediatrics, Neuroscience, and Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, 14642
| | - Todd D. Krauss
- Department of Chemistry, Rochester, New York 14627-0216, United States
- The Institute of Optics, Rochester, New York 14627-0216, United States
| |
Collapse
|
9
|
Duran-Aniotz C, Poblete N, Rivera-Krstulovic C, Ardiles ÁO, Díaz-Hung ML, Tamburini G, Sabusap CMP, Gerakis Y, Cabral-Miranda F, Diaz J, Fuentealba M, Arriagada D, Muñoz E, Espinoza S, Martinez G, Quiroz G, Sardi P, Medinas DB, Contreras D, Piña R, Lourenco MV, Ribeiro FC, Ferreira ST, Rozas C, Morales B, Plate L, Gonzalez-Billault C, Palacios AG, Hetz C. The unfolded protein response transcription factor XBP1s ameliorates Alzheimer's disease by improving synaptic function and proteostasis. Mol Ther 2023; 31:2240-2256. [PMID: 37016577 PMCID: PMC10362463 DOI: 10.1016/j.ymthe.2023.03.028] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 02/03/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023] Open
Abstract
Alteration in the buffering capacity of the proteostasis network is an emerging feature of Alzheimer's disease (AD), highlighting the occurrence of endoplasmic reticulum (ER) stress. The unfolded protein response (UPR) is the main adaptive pathway to cope with protein folding stress at the ER. Inositol-requiring enzyme-1 (IRE1) operates as a central ER stress sensor, enabling the establishment of adaptive and repair programs through the control of the expression of the transcription factor X-box binding protein 1 (XBP1). To artificially enforce the adaptive capacity of the UPR in the AD brain, we developed strategies to express the active form of XBP1 in the brain. Overexpression of XBP1 in the nervous system using transgenic mice reduced the load of amyloid deposits and preserved synaptic and cognitive function. Moreover, local delivery of XBP1 into the hippocampus of an 5xFAD mice using adeno-associated vectors improved different AD features. XBP1 expression corrected a large proportion of the proteomic alterations observed in the AD model, restoring the levels of several synaptic proteins and factors involved in actin cytoskeleton regulation and axonal growth. Our results illustrate the therapeutic potential of targeting UPR-dependent gene expression programs as a strategy to ameliorate AD features and sustain synaptic function.
Collapse
Affiliation(s)
- Claudia Duran-Aniotz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Latin American Institute for Brain Health (BrainLat), Universidad Adolfo Ibáñez, Santiago, Chile; Center for Social and Cognitive Neuroscience (CSCN), School of Psychology, Universidad Adolfo Ibanez, Santiago, Chile.
| | - Natalia Poblete
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Catalina Rivera-Krstulovic
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Álvaro O Ardiles
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Mei Li Díaz-Hung
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Giovanni Tamburini
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Carleen Mae P Sabusap
- Department of Chemistry and Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Yannis Gerakis
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Felipe Cabral-Miranda
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Javier Diaz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Matias Fuentealba
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Diego Arriagada
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Ernesto Muñoz
- FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Department of Biology, Faculty of Sciences and Department of Neurosciences, Faculty of Medicina, Universidad de Chile, Santiago, Chile
| | - Sandra Espinoza
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Gabriela Martinez
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Gabriel Quiroz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Pablo Sardi
- Rare and Neurological Diseases Therapeutic Area, Sanofi, Framingham, MA, USA
| | - Danilo B Medinas
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Darwin Contreras
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Ricardo Piña
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Mychael V Lourenco
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Felipe C Ribeiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; D'Or Institute for Research and Education, Rio de Janeiro, Brazil
| | - Carlos Rozas
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Bernardo Morales
- Laboratory of Neuroscience, Department of Biology, Faculty of Chemistry and Biology, University of Santiago de Chile, Santiago, Chile
| | - Lars Plate
- Department of Chemistry and Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Christian Gonzalez-Billault
- FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Department of Biology, Faculty of Sciences and Department of Neurosciences, Faculty of Medicina, Universidad de Chile, Santiago, Chile
| | - Adrian G Palacios
- Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute (BNI), Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health, and Metabolism (GERO), Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA 94945, USA.
| |
Collapse
|
10
|
Gao Y, Wei GZ, Forston MD, Rood B, Hodges ER, Burke D, Andres K, Morehouse J, Armstrong C, Glover C, Slomnicki LP, Ding J, Chariker JH, Rouchka EC, Saraswat Ohri S, Whittemore SR, Hetman M. Opposite modulation of functional recovery following contusive spinal cord injury in mice with oligodendrocyte-selective deletions of Atf4 and Chop/Ddit3. Sci Rep 2023; 13:9193. [PMID: 37280306 PMCID: PMC10244317 DOI: 10.1038/s41598-023-36258-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 05/31/2023] [Indexed: 06/08/2023] Open
Abstract
The integrated stress response (ISR)-activated transcription factors ATF4 and CHOP/DDIT3 may regulate oligodendrocyte (OL) survival, tissue damage and functional impairment/recovery in white matter pathologies, including traumatic spinal cord injury (SCI). Accordingly, in OLs of OL-specific RiboTag mice, Atf4, Chop/Ddit3 and their downstream target gene transcripts were acutely upregulated at 2, but not 10, days post-contusive T9 SCI coinciding with maximal loss of spinal cord tissue. Unexpectedly, another, OL-specific upregulation of Atf4/Chop followed at 42 days post-injury. However, wild type versus OL-specific Atf4-/- or Chop-/- mice showed similar white matter sparing and OL loss at the injury epicenter, as well as unaffected hindlimb function recovery as determined by the Basso mouse scale. In contrast, the horizontal ladder test revealed persistent worsening or improvement of fine locomotor control in OL-Atf4-/- or OL-Chop-/- mice, respectively. Moreover, chronically, OL-Atf-/- mice showed decreased walking speed during plantar stepping despite greater compensatory forelimb usage. Therefore, ATF4 supports, while CHOP antagonizes, fine locomotor control during post-SCI recovery. No correlation between those effects and white matter sparing together with chronic activation of the OL ISR suggest that in OLs, ATF4 and CHOP regulate function of spinal cord circuitries that mediate fine locomotor control during post-SCI recovery.
Collapse
Affiliation(s)
- Yonglin Gao
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - George Z Wei
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA
- MD/PhD Program, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Michael D Forston
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Benjamin Rood
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Emily R Hodges
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Darlene Burke
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Kariena Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Johnny Morehouse
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Christine Armstrong
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Charles Glover
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
| | - Lukasz P Slomnicki
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Jixiang Ding
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, 40292, USA
| | - Julia H Chariker
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, University of Louisville, Louisville, KY, 40292, USA
| | - Eric C Rouchka
- Kentucky Biomedical Research Infrastructure Network Bioinformatics Core, University of Louisville, Louisville, KY, 40292, USA
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40292, USA
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, 511 S. Floyd St., MDR616, Louisville, KY, 40202, USA.
- Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, 40292, USA.
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.
- Department of Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, KY, 40292, USA.
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, 40292, USA.
| |
Collapse
|
11
|
Saraswat Ohri S, Andres KR, Howard RM, Brown BL, Forston MD, Hetman M, Whittemore SR. Acute Pharmacological Inhibition of Protein Kinase R-Like Endoplasmic Reticulum Kinase Signaling After Spinal Cord Injury Spares Oligodendrocytes and Improves Locomotor Recovery. J Neurotrauma 2023; 40:1007-1019. [PMID: 36503284 PMCID: PMC10162120 DOI: 10.1089/neu.2022.0177] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK) is a major signal transducer of the endoplasmic reticulum stress response (ERSR) pathway. Outcomes of PERK activation range from abrogating ER stress to induction of cell death, dependent on its level, duration, and cellular context. Current data demonstrate that after mouse spinal cord injury (SCI), acute inhibition of PERK (0-72 h) with the small molecule inhibitor GSK2656157 reduced ERSR while improving white matter sparing and hindlimb locomotion recovery. GSK2656157-treated mice showed increased numbers of oligodendrocytes at the injury epicenter. Moreover, GSK2656157 protected cultured primary mouse oligodendrocyte precursor cells from ER stress-induced cytotoxicity. These findings suggest that in the context of SCI, excessive acute activation of PERK contributes to functionally relevant white matter damage. Pharmacological inhibition of PERK is a potential strategy to protect central nervous system (CNS) white matter following acute injuries, including SCI.
Collapse
Affiliation(s)
- Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Interdisciplinary Program in Translational Neuroscience, and Department of University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Kariena R. Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Russell M. Howard
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Brandon L. Brown
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Interdisciplinary Program in Translational Neuroscience, and Department of University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Anatomical Sciences and Neurobiology, and University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Michael D. Forston
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Anatomical Sciences and Neurobiology, and University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Interdisciplinary Program in Translational Neuroscience, and Department of University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Anatomical Sciences and Neurobiology, and University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Pharmacology and Toxicology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| | - Scott R. Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Interdisciplinary Program in Translational Neuroscience, and Department of University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Neurological Surgery, University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Anatomical Sciences and Neurobiology, and University of Louisville, School of Medicine, Louisville, Kentucky, USA
- Pharmacology and Toxicology, University of Louisville, School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
12
|
Fan Q, Takarada-Iemata M, Okitani N, Tamatani T, Ishii H, Hattori T, Kiryu-Seo S, Kiyama H, Hori O. Brain injury triggers cell-type-specific and time-dependent endoplasmic reticulum stress responses. Glia 2023; 71:667-681. [PMID: 36412235 DOI: 10.1002/glia.24303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Revised: 10/29/2022] [Accepted: 11/03/2022] [Indexed: 11/23/2022]
Abstract
The unfolded protein response (UPR) is a signal transduction network that responds to endoplasmic reticulum (ER) stress by coordinating protein homeostasis to maintain cell viability. The UPR can also trigger cell death when adaptive responses fail to improve protein homeostasis. Despite accumulating evidence suggesting that the UPR plays a role in neurodegenerative diseases and brain insults, our understanding of how ER stress is induced under neuropathological conditions is limited. Here, we investigated the cell- and time-specific patterns of the ER stress response after brain injury using ER stress-activated indicator (ERAI) mice, which enable monitoring of the UPR in vivo via increased fluorescence of a spliced XBP-1 protein fused with the green fluorescent protein (GFP) variant Venus. Following cortical stab injury of ERAI mice, the GFP signal and number of GFP+ cells increased in the ipsilateral cortex throughout the observation period (6 h to 7 days post-injury), confirming the induction of the UPR. GFP signals were observed in injured neurons early (from 6 h) after brain injury. However, non-neuronal cells, mainly endothelial cells followed by astrocytes, accounted for the majority of GFP+ cells after brain injury. Similar results were obtained in a mouse model of focal cerebral ischemia. These findings suggest that activation of the UPR in both neuronal and non-neuronal cells, especially endothelial cells and astrocytes, may play an important role in and could be a potential therapeutic target for acute brain injuries.
Collapse
Affiliation(s)
- Qiyan Fan
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Mika Takarada-Iemata
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Nahoko Okitani
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Takashi Tamatani
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Hiroshi Ishii
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Tsuyoshi Hattori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Sumiko Kiryu-Seo
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroshi Kiyama
- Department of Functional Anatomy and Neuroscience, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Osamu Hori
- Department of Neuroanatomy, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
13
|
Vestuto V, Di Sarno V, Musella S, Di Dona G, Moltedo O, Gomez-Monterrey IM, Bertamino A, Ostacolo C, Campiglia P, Ciaglia T. New Frontiers on ER Stress Modulation: Are TRP Channels the Leading Actors? Int J Mol Sci 2022; 24:185. [PMID: 36613628 PMCID: PMC9820239 DOI: 10.3390/ijms24010185] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/15/2022] [Accepted: 12/20/2022] [Indexed: 12/24/2022] Open
Abstract
The endoplasmic reticulum (ER) is a dynamic structure, playing multiple roles including calcium storage, protein synthesis and lipid metabolism. During cellular stress, variations in ER homeostasis and its functioning occur. This condition is referred as ER stress and generates a cascade of signaling events termed unfolded protein response (UPR), activated as adaptative response to mitigate the ER stress condition. In this regard, calcium levels play a pivotal role in ER homeostasis and therefore in cell fate regulation since calcium signaling is implicated in a plethora of physiological processes, but also in disease conditions such as neurodegeneration, cancer and metabolic disorders. A large body of emerging evidence highlighted the functional role of TRP channels and their ability to promote cell survival or death depending on endoplasmic reticulum stress resolution, making them an attractive target. Thus, in this review we focused on the TRP channels' correlation to UPR-mediated ER stress in disease pathogenesis, providing an overview of their implication in the activation of this cellular response.
Collapse
Affiliation(s)
- Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Veronica Di Sarno
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Simona Musella
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Giorgio Di Dona
- Pineta Grande Hospital, Via Domiziana, km 30/00, 81030 Castel Volturno, CE, Italy
| | - Ornella Moltedo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | | | - Alessia Bertamino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| | - Carmine Ostacolo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
- Department of Pharmacy, University of Naples Federico II, Via D. Montesano 49, 80131 Napoli, NA, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
- European Biomedical Research Institute of Salerno, Via S. De Renzi 50, 84125 Salerno, SA, Italy
| | - Tania Ciaglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084 Fisciano, SA, Italy
| |
Collapse
|
14
|
Chen L, Bi M, Zhang Z, Du X, Chen X, Jiao Q, Jiang H. The functions of IRE1α in neurodegenerative diseases: Beyond ER stress. Ageing Res Rev 2022; 82:101774. [PMID: 36332756 DOI: 10.1016/j.arr.2022.101774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 10/19/2022] [Accepted: 10/29/2022] [Indexed: 11/05/2022]
Abstract
Inositol-requiring enzyme 1 α (IRE1α) is a type I transmembrane protein that resides in the endoplasmic reticulum (ER). IRE1α, which is the primary sensor of ER stress, has been proven to maintain intracellular protein homeostasis by activating X-box binding protein 1 (XBP1). Further studies have revealed novel physiological functions of the IRE1α, such as its roles in mRNA and protein degradation, inflammation, immunity, cell proliferation and cell death. Therefore, the function of IRE1α is not limited to its role in ER stress; IRE1α is also important for regulating other processes related to cellular physiology. Furthermore, IRE1α plays a key role in neurodegenerative diseases that are caused by the phosphorylation of Tau protein, the accumulation of α-synuclein (α-syn) and the toxic effects of mutant Huntingtin (mHtt). Therefore, targeting IRE1α is a valuable approach for treating neurodegenerative diseases and regulating cell functions. This review discusses the role of IRE1α in different cellular processes, and emphasizes the importance of IRE1α in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ling Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Mingxia Bi
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Zhen Zhang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xixun Du
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Xi Chen
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China
| | - Qian Jiao
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China.
| | - Hong Jiang
- Department of Physiology, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, School of Basic Medicine, Qingdao University, Qingdao, China; University of Health and Rehabilitation Sciences, Qingdao, China.
| |
Collapse
|
15
|
The Proteostasis Network: A Global Therapeutic Target for Neuroprotection after Spinal Cord Injury. Cells 2022; 11:cells11213339. [PMID: 36359735 PMCID: PMC9658791 DOI: 10.3390/cells11213339] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/14/2022] [Accepted: 10/20/2022] [Indexed: 01/18/2023] Open
Abstract
Proteostasis (protein homeostasis) is critical for cellular as well as organismal survival. It is strictly regulated by multiple conserved pathways including the ubiquitin-proteasome system, autophagy, the heat shock response, the integrated stress response, and the unfolded protein response. These overlapping proteostasis maintenance modules respond to various forms of cellular stress as well as organismal injury. While proteostasis restoration and ultimately organism survival is the main evolutionary driver of such a regulation, unresolved disruption of proteostasis may engage pro-apoptotic mediators of those pathways to eliminate defective cells. In this review, we discuss proteostasis contributions to the pathogenesis of traumatic spinal cord injury (SCI). Most published reports focused on the role of proteostasis networks in acute/sub-acute tissue damage post-SCI. Those reports reveal a complex picture with cell type- and/or proteostasis mediator-specific effects on loss of neurons and/or glia that often translate into the corresponding modulation of functional recovery. Effects of proteostasis networks on such phenomena as neuro-repair, post-injury plasticity, as well as systemic manifestations of SCI including dysregulation of the immune system, metabolism or cardiovascular function are currently understudied. However, as potential interventions that target the proteostasis networks are expected to impact many cell types across multiple organ systems that are compromised after SCI, such therapies could produce beneficial effects across the wide spectrum of highly variable human SCI.
Collapse
|
16
|
Kukharsky MS, Everett MW, Lytkina OA, Raspopova MA, Kovrazhkina EA, Ovchinnikov RK, Antohin AI, Moskovtsev AA. Protein Homeostasis Dysregulation in Pathogenesis of Neurodegenerative Diseases. Mol Biol 2022. [DOI: 10.1134/s0026893322060115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
17
|
Restoration of ER proteostasis attenuates remote apoptotic cell death after spinal cord injury by reducing autophagosome overload. Cell Death Dis 2022; 13:381. [PMID: 35444186 PMCID: PMC9021197 DOI: 10.1038/s41419-022-04830-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 03/30/2022] [Accepted: 04/04/2022] [Indexed: 02/07/2023]
Abstract
The pathogenic mechanisms that underlie the progression of remote degeneration after spinal cord injury (SCI) are not fully understood. In this study, we examined the relationship between endoplasmic reticulum (ER) stress and macroautophagy, hereafter autophagy, and its contribution to the secondary damage and outcomes that are associated with remote degeneration after SCI. Using a rat model of spinal cord hemisection at the cervical level, we measured ER stress and autophagy markers in the axotomized neurons of the red nucleus (RN). In SCI animals, mRNA and protein levels of markers of ER stress, such as GRP78, CHOP, and GADD34, increased 1 day after the injury, peaking on Day 5. Notably, in SCI animals, the increase of ER stress markers correlated with a blockade in autophagic flux, as evidenced by the increase in microtubule-associated protein 2 light chain 3 (LC3-II) and p62/SQSTM1 (p62) and the decline in LAMP1 and LAMP2 levels. After injury, treatment with guanabenz protected neurons from UPR failure and increased lysosomes biogenesis, unblocking autophagic flux. These effects correlated with greater activation of TFEB and improved neuronal survival and functional recovery—effects that persisted after suspension of the treatment. Collectively, our results demonstrate that in remote secondary damage, impairments in autophagic flux are intertwined with ER stress, an association that contributes to the apoptotic cell death and functional damage that are observed after SCI.
Collapse
|
18
|
Li H, Wen W, Luo J. Targeting Endoplasmic Reticulum Stress as an Effective Treatment for Alcoholic Pancreatitis. Biomedicines 2022; 10:biomedicines10010108. [PMID: 35052788 PMCID: PMC8773075 DOI: 10.3390/biomedicines10010108] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/28/2021] [Accepted: 12/30/2021] [Indexed: 02/04/2023] Open
Abstract
Pancreatitis and alcoholic pancreatitis are serious health concerns with an urgent need for effective treatment strategies. Alcohol is a known etiological factor for pancreatitis, including acute pancreatitis (AP) and chronic pancreatitis (CP). Excessive alcohol consumption induces many pathological stress responses; of particular note is endoplasmic reticulum (ER) stress and adaptive unfolded protein response (UPR). ER stress results from the accumulation of unfolded/misfolded protein in the ER and is implicated in the pathogenesis of alcoholic pancreatitis. Here, we summarize the possible mechanisms by which ER stress contributes to alcoholic pancreatitis. We also discuss potential approaches targeting ER stress and UPR in developing novel therapeutic strategies for the disease.
Collapse
Affiliation(s)
- Hui Li
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (H.L.); (W.W.)
| | - Wen Wen
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (H.L.); (W.W.)
| | - Jia Luo
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (H.L.); (W.W.)
- Iowa City VA Health Care System, Iowa City, IA 52246, USA
- Correspondence: ; Tel.: +1-319-335-2256
| |
Collapse
|
19
|
Moraga P, Aravena R, Urra H, Hetz C. Assays to Study IRE1 Activation and Signaling. Methods Mol Biol 2022; 2378:141-168. [PMID: 34985699 DOI: 10.1007/978-1-0716-1732-8_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The endoplasmic reticulum (ER) stress sensor IRE1 is a a major player of the unfolded protein response (UPR), the main pathway driving adaptation processes to restore proteostasis. In addition, overactivation of IRE1 signaling contributes to a variety of pathologies including diabetes, neurodegenerative diseases, and cancer. Under ER stress, IRE1 auto-transphosphorylates and oligomerizes, triggering the activation of its endoribonuclease domain located in the cytosolic region. Active IRE1 catalyzes the splicing of the mRNA encoding for the XBP1 transcription factor, in addition to degrade several RNAs through a process known as regulated IRE1-dependent decay of mRNA (RIDD). Besides its role as an UPR transducer, several posttranslational modifications and protein-protein interactions can regulate IRE1 activity and modulate its signaling in the absence of stress. Thus, investigating the function of IRE1 in physiology and disease requires the use of complementary approaches. Here, we provide detailed protocols to perform four different assays to study IRE1 activation and signaling: (i) Phos-tag gels to evaluate the phosphorylation status of IRE1, (ii) microscopy using TREX-IRE1-GFP cells to measure IRE1 oligomerization, (iii) conventional RT-PCR to assess XBP1 mRNA processing, and (iv) quantitative PCR to determine the levels of canonical UPR target genes and the degradation of several mRNAs that are target of RIDD. We propose to use these experimental strategies as "gold standards" to study IRE1 signaling.
Collapse
Affiliation(s)
- Paloma Moraga
- Faculty of Medicine, Biomedical Neuroscience Institute (BNI), University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile
| | - Raul Aravena
- Faculty of Medicine, Biomedical Neuroscience Institute (BNI), University of Chile, Santiago, Chile
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile
| | - Hery Urra
- Faculty of Medicine, Biomedical Neuroscience Institute (BNI), University of Chile, Santiago, Chile.
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile.
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile.
| | - Claudio Hetz
- Faculty of Medicine, Biomedical Neuroscience Institute (BNI), University of Chile, Santiago, Chile.
- Center for Geroscience, Brain Health and Metabolism (GERO), Santiago, Chile.
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences (ICBM), University of Chile, Santiago, Chile.
- The Buck Institute for Research in Aging, Novato, CA, USA.
| |
Collapse
|
20
|
Begenisic T, Pavese C, Aiachini B, Nardone A, Rossi D. Dynamics of biomarkers across the stages of traumatic spinal cord injury - implications for neural plasticity and repair. Restor Neurol Neurosci 2021; 39:339-366. [PMID: 34657853 DOI: 10.3233/rnn-211169] [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] [Indexed: 12/31/2022]
Abstract
BACKGROUND Traumatic spinal cord injury (SCI) is a complex medical condition causing significant physical disability and psychological distress. While the adult spinal cord is characterized by poor regenerative potential, some recovery of neurological function is still possible through activation of neural plasticity mechanisms. We still have limited knowledge about the activation of these mechanisms in the different stages after human SCI. OBJECTIVE In this review, we discuss the potential role of biomarkers of SCI as indicators of the plasticity mechanisms at work during the different phases of SCI. METHODS An extensive review of literature related to SCI pathophysiology, neural plasticity and humoral biomarkers was conducted by consulting the PubMed database. Research and review articles from SCI animal models and SCI clinical trials published in English until January 2021 were reviewed. The selection of candidates for humoral biomarkers of plasticity after SCI was based on the following criteria: 1) strong evidence supporting involvement in neural plasticity (mandatory); 2) evidence supporting altered expression after SCI (optional). RESULTS Based on selected findings, we identified two main groups of potential humoral biomarkers of neural plasticity after SCI: 1) neurotrophic factors including: Brain derived neurotrophic factor (BDNF), Nerve growth factor (NGF), Neurotrofin-3 (NT-3), and Insulin-like growth factor 1 (IGF-1); 2) other factors including: Tumor necrosis factor-alpha (TNF-α), Matrix Metalloproteinases (MMPs), and MicroRNAs (miRNAs). Plasticity changes associated with these biomarkers often can be both adaptive (promoting functional improvement) and maladaptive. This dual role seems to be influenced by their concentrations and time-window during SCI. CONCLUSIONS Further studies of dynamics of biomarkers across the stages of SCI are necessary to elucidate the way in which they reflect the remodeling of neural pathways. A better knowledge about the mechanisms underlying plasticity could guide the selection of more appropriate therapeutic strategies to enhance positive spinal network reorganization.
Collapse
Affiliation(s)
- Tatjana Begenisic
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Chiara Pavese
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.,Neurorehabilitation and Spinal Units, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| | - Beatrice Aiachini
- Neurorehabilitation and Spinal Units, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| | - Antonio Nardone
- Department of Clinical-Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy.,Neurorehabilitation and Spinal Units, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| | - Daniela Rossi
- Laboratory for Research on Neurodegenerative Disorders, ICS Maugeri SPA SB, Institute of Pavia, IRCCS, Pavia, Italy
| |
Collapse
|
21
|
Chang L, Liu X, Chen J, Liu H, Wang G, Wang G, Liao X, Shen X. Attenuation of Activated eIF2α Signaling by ISRIB Treatment After Spinal Cord Injury Improves Locomotor Function. J Mol Neurosci 2021; 72:585-597. [PMID: 34647267 PMCID: PMC8921087 DOI: 10.1007/s12031-021-01920-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 09/22/2021] [Indexed: 11/30/2022]
Abstract
Following spinal cord injury (SCI), multiple signaling cascades are activated instantaneously in the injured segments of the spinal cord to create a complex and pathogenic microenvironment, making it difficult to treat SCI. Nevertheless, the significance of the integrated stress response (ISR) to the series of physiological and pathological changes that occur after SCI remains unclear. Through western blotting (WB), we determined that the autophosphorylation of stress receptors (GCN2, PERK, PKR, and HRI) was enhanced after SCI, leading to increased phosphorylation of eIF2α at Ser51. Strikingly, we found that eIF2α was highly phosphorylated at 1 day post injury (dpi) and that this hypophosphorylation was maintained thereafter in the spinal cord, especially in neurons, which suggests that intervening with eIF2α phosphorylation may be a treatment strategy for SCI. Therefore, we employed the small molecule ISRIB, which inhibits eIF2α phosphorylation when the ISR is activated at moderate or low levels but not when the ISR is highly activated. Daily intraperitoneal injection of ISRIB significantly inhibited ISR signaling after SCI, reduced the cytosolic localization of RNA-binding proteins, and decreased neuronal apoptosis. Histological and functional experiments further demonstrated that treatment with ISRIB after SCI effectively curbed morphological deterioration and promoted the recovery of locomotor function. In summary, the ISR plays an important role in SCI, and ISRIB is a promising drug for the treatment of SCI.
Collapse
Affiliation(s)
- Lei Chang
- Department of Spine Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University), No.61, West Jiefang Road, Changsha, 410005, China
| | - Xiangyang Liu
- Department of Spine Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University), No.61, West Jiefang Road, Changsha, 410005, China
| | - Jing Chen
- Department of Endocrinology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Hongzhe Liu
- Department of Spine Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University), No.61, West Jiefang Road, Changsha, 410005, China
| | - Guoping Wang
- Department of Spine Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University), No.61, West Jiefang Road, Changsha, 410005, China
| | - Guohua Wang
- Department of Spine Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University), No.61, West Jiefang Road, Changsha, 410005, China
| | - Xiaoyun Liao
- Department of Anesthesiology, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University), Changsha, China
| | - Xiongjie Shen
- Department of Spine Surgery, Hunan Provincial People's Hospital, The First Affiliated Hospital of Hunan Normal University), No.61, West Jiefang Road, Changsha, 410005, China.
| |
Collapse
|
22
|
Ren Z, Hu Y, Guo D, Guan Z, Chen L, He J, Yu W. Increased miR‑187‑3p expression after cerebral ischemia/reperfusion induces apoptosis via initiation of endoplasmic reticulum stress. Neurosci Lett 2021; 759:135947. [PMID: 34015413 DOI: 10.1016/j.neulet.2021.135947] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 04/13/2021] [Accepted: 05/05/2021] [Indexed: 12/16/2022]
Abstract
Ischemia/reperfusion (I/R) injury induces activation of the endoplasmic reticulum stress (ERS) pathway, accompanied by an increase in apoptosis. Multiple microRNAs (miRNAs/miRs) are dysregulated during I/R and contribute to I/R-induced injury. miRNAs act as suppressors of gene expression and negatively regulate gene expression by targeting the protein-coding sequence (CDS) of specific target mRNAs. Seipin is an endoplasmic reticulum protein that has recently been associated with ERS. We previously reported that seipin is the target gene of miR‑187‑3p. Therefore, we explored the involvement of miR-187-3p in I/R-induced ERS via the regulation of seipin. A rat MCAO/R model was established by 1 h of occlusion and 24 h reperfusion. Neurological deficits and infarction area were examined. PC12 cells were exposed to oxygen‑glucose deprivation/reoxygenation (OGD/R) to model I/R. Expression levels of miR-187-3p and proteins related to ERS and apoptosis were measured using RT-PCR, western blotting, immunofluorescence, and immunohistochemistry, respectively. TUNEL staining was used to assay apoptosis. MCAO/R-induced morphological changes were analyzed with Nissl staining and Hematoxylin-eosin staining. I/R-induced ERS was closely associated with an increase in miR-1873p and a decrease in seipin expression. miR-187-3p agomir further activated the ERS pathway and promoted apoptosis but decreased seipin expression levels; these effects were reversed by miR-187-3p antagomir. Moreover, seipin knockdown aggravated ERS in PC12 cells after OGD/R, and this change was rescued by seipin overexpression. miR-187-3p antagomir did not suppress ERS and apoptosis in seipin knockdown PC12 cells after OGD/R. Our findings demonstrate that the inhibition of miR‑187‑3p attenuated I/R‑induced cerebral injury by regulating seipin-mediated ERS.
Collapse
Affiliation(s)
- Zhenkui Ren
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, China; Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, 550004, China; Laboratory Department of People's Hospital of Southwest Guizhou Autonomous Prefecture, Xingyi, Guizhou, 562400, China
| | - Yumei Hu
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, China; Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Dongfen Guo
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, China; Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Zhizhong Guan
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, China; Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, 550004, China; Department of Pathology, Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou, 550004, China
| | - Ling Chen
- Laboratory of Reproductive Medicine, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Jun He
- Department of Laboratory Medicine, The Second People's Hospital of Guizhou Province, Guiyang, 550002, China; Department of Immunology, Guizhou Medical University, Guiyang, 550004, China.
| | - Wenfeng Yu
- Key Laboratory of Endemic and Ethnic Diseases, Ministry of Education, School of Basic Medical Science, Guizhou Medical University, China; Key Laboratory of Medical Molecular Biology, Guizhou Medical University, Guiyang, Guizhou, 550004, China.
| |
Collapse
|
23
|
Vidal RL, Sepulveda D, Troncoso-Escudero P, Garcia-Huerta P, Gonzalez C, Plate L, Jerez C, Canovas J, Rivera CA, Castillo V, Cisternas M, Leal S, Martinez A, Grandjean J, Sonia D, Lashuel HA, Martin AJM, Latapiat V, Matus S, Sardi SP, Wiseman RL, Hetz C. Enforced dimerization between XBP1s and ATF6f enhances the protective effects of the UPR in models of neurodegeneration. Mol Ther 2021; 29:1862-1882. [PMID: 33545358 PMCID: PMC8116614 DOI: 10.1016/j.ymthe.2021.01.033] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 11/14/2020] [Accepted: 01/21/2021] [Indexed: 02/07/2023] Open
Abstract
Alteration to endoplasmic reticulum (ER) proteostasis is observed in a variety of neurodegenerative diseases associated with abnormal protein aggregation. Activation of the unfolded protein response (UPR) enables an adaptive reaction to recover ER proteostasis and cell function. The UPR is initiated by specialized stress sensors that engage gene expression programs through the concerted action of the transcription factors ATF4, ATF6f, and XBP1s. Although UPR signaling is generally studied as unique linear signaling branches, correlative evidence suggests that ATF6f and XBP1s may physically interact to regulate a subset of UPR target genes. In this study, we designed an ATF6f/XBP1s fusion protein termed UPRplus that behaves as a heterodimer in terms of its selective transcriptional activity. Cell-based studies demonstrated that UPRplus has a stronger effect in reducing the abnormal aggregation of mutant huntingtin and α-synuclein when compared to XBP1s or ATF6 alone. We developed a gene transfer approach to deliver UPRplus into the brain using adeno-associated viruses (AAVs) and demonstrated potent neuroprotection in vivo in preclinical models of Parkinson's disease and Huntington's disease. These results support the concept in which directing UPR-mediated gene expression toward specific adaptive programs may serve as a possible strategy to optimize the beneficial effects of the pathway in different disease conditions.
Collapse
Affiliation(s)
- René L Vidal
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.
| | - Denisse Sepulveda
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Paulina Troncoso-Escudero
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Paula Garcia-Huerta
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Constanza Gonzalez
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Lars Plate
- Department of Chemistry, Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA; Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Carolina Jerez
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - José Canovas
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Claudia A Rivera
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Valentina Castillo
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Marisol Cisternas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Sirley Leal
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Alexis Martinez
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Julia Grandjean
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Donzelli Sonia
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, 1015 Lausanne, Switzerland
| | - Alberto J M Martin
- Laboratorio de Biología de Redes, Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Veronica Latapiat
- Laboratorio de Biología de Redes, Centro de Genómica y Bioinformática, Facultad de Ciencias, Universidad Mayor, Santiago, Chile
| | - Soledad Matus
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Fundacion Ciencia Vida, Santiago 7780272, Chile; Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia 7510157, Santiago, Chile
| | - S Pablo Sardi
- Rare and Neurological Diseases Therapeutic Area, Sanofi, 49 New York Avenue, Framingham, MA, USA
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA 94945, USA.
| |
Collapse
|
24
|
Wei GZ, Saraswat Ohri S, Khattar NK, Listerman AW, Doyle CH, Andres KR, Karuppagounder SS, Ratan RR, Whittemore SR, Hetman M. Hypoxia-inducible factor prolyl hydroxylase domain (PHD) inhibition after contusive spinal cord injury does not improve locomotor recovery. PLoS One 2021; 16:e0249591. [PMID: 33819286 PMCID: PMC8021188 DOI: 10.1371/journal.pone.0249591] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 03/19/2021] [Indexed: 12/12/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is a devastating neurological condition that involves both primary and secondary tissue loss. Various cytotoxic events including hypoxia, hemorrhage and blood lysis, bioenergetic failure, oxidative stress, endoplasmic reticulum (ER) stress, and neuroinflammation contribute to secondary injury. The HIF prolyl hydroxylase domain (PHD/EGLN) family of proteins are iron-dependent, oxygen-sensing enzymes that regulate the stability of hypoxia inducible factor-1α (HIF-1α) and also mediate oxidative stress caused by free iron liberated from the lysis of blood. PHD inhibition improves outcome after experimental intracerebral hemorrhage (ICH) by reducing activating transcription factor 4 (ATF4)-driven neuronal death. As the ATF4-CHOP (CCAAT-enhancer-binding protein homologous protein) pathway plays a role in the pathogenesis of contusive SCI, we examined the effects of PHD inhibition in a mouse model of moderate T9 contusive SCI in which white matter damage is the primary driver of locomotor dysfunction. Pharmacological inhibition of PHDs using adaptaquin (AQ) moderately lowers acute induction of Atf4 and Chop mRNAs and prevents the acute decline of oligodendrocyte (OL) lineage mRNAs, but does not improve long-term recovery of hindlimb locomotion or increase chronic white matter sparing. Conditional genetic ablation of all three PHD isoenzymes in OLs did not affect Atf4, Chop or OL mRNAs expression levels, locomotor recovery, and white matter sparing after SCI. Hence, PHDs may not be suitable targets to improve outcomes in traumatic CNS pathologies that involve acute white matter injury.
Collapse
Affiliation(s)
- George Z Wei
- University of Louisville School of Medicine, Louisville, Kentucky, United States of America.,Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.,Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Nicolas K Khattar
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.,Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Adam W Listerman
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Catherine H Doyle
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Saravanan S Karuppagounder
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Neurological Institute, White Plains, NY, United States of America.,Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY, United States of America
| | - Rajiv R Ratan
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Neurological Institute, White Plains, NY, United States of America.,Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY, United States of America
| | - Scott R Whittemore
- Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.,Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Michal Hetman
- University of Louisville School of Medicine, Louisville, Kentucky, United States of America.,Department of Pharmacology & Toxicology, University of Louisville School of Medicine, Louisville, KY, United States of America.,Kentucky Spinal Cord Injury Research Center, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Neurological Surgery, University of Louisville School of Medicine, Louisville, KY, United States of America.,Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY, United States of America
| |
Collapse
|
25
|
Hetz C. Adapting the proteostasis capacity to sustain brain healthspan. Cell 2021; 184:1545-1560. [PMID: 33691137 DOI: 10.1016/j.cell.2021.02.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 01/20/2021] [Accepted: 02/01/2021] [Indexed: 12/13/2022]
Abstract
Sustaining neuronal proteostasis during the course of our life is a central aspect required for brain function. The dynamic nature of synaptic composition and abundance is a requisite to drive cognitive and motor processes involving a tight control of many aspects of protein biosynthesis and degradation. Through the concerted action of specialized stress sensors, the proteostasis network monitors and limits the accumulation of damaged, misfolded, or aggregated proteins. These stress pathways signal to the cytosol and nucleus to reprogram gene expression, enabling adaptive programs to recover cell function. During aging, the activity of the proteostasis network declines, which may increase the risk of accumulating abnormal protein aggregates, a hallmark of most neurodegenerative diseases. Here, I discuss emerging concepts illustrating the functional significance of adaptive signaling pathways to normal brain physiology and their contribution to age-related disorders. Pharmacological and gene therapy strategies to intervene and boost proteostasis are expected to extend brain healthspan and ameliorate disease states.
Collapse
Affiliation(s)
- Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, USA.
| |
Collapse
|
26
|
Jin LY, Li J, Wang KF, Xia WW, Zhu ZQ, Wang CR, Li XF, Liu HY. Blood-Spinal Cord Barrier in Spinal Cord Injury: A Review. J Neurotrauma 2021; 38:1203-1224. [PMID: 33292072 DOI: 10.1089/neu.2020.7413] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The blood-spinal cord barrier (BSCB), a physical barrier between the blood and spinal cord parenchyma, prevents the toxins, blood cells, and pathogens from entering the spinal cord and maintains a tightly controlled chemical balance in the spinal environment, which is necessary for proper neural function. A BSCB disruption, however, plays an important role in primary and secondary injury processes related to spinal cord injury (SCI). After SCI, the structure of the BSCB is broken down, which leads directly to leakage of blood components. At the same time, the permeability of the BSCB is also increased. Repairing the disruption of the BSCB could alleviate the SCI pathology. We review the morphology and pathology of the BSCB and progression of therapeutic methods targeting BSCB in SCI.
Collapse
Affiliation(s)
- Lin-Yu Jin
- Department of Spinal Surgery, Peking University People's Hospital, Peking University, Beijing, P.R. China
| | - Jie Li
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, P.R. China
| | - Kai-Feng Wang
- Department of Spinal Surgery, Peking University People's Hospital, Peking University, Beijing, P.R. China
| | - Wei-Wei Xia
- Department of Spinal Surgery, Peking University People's Hospital, Peking University, Beijing, P.R. China
| | - Zhen-Qi Zhu
- Department of Spinal Surgery, Peking University People's Hospital, Peking University, Beijing, P.R. China
| | - Chun-Ru Wang
- Beijing National Laboratory for Molecular Sciences, Key Laboratory of Molecular Nanostructure and Nanotechnology, CAS Research/Education Center for Excellence in Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing, P.R. China
| | - Xin-Feng Li
- Department of Spinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P.R. China
| | - Hai-Ying Liu
- Department of Spinal Surgery, Peking University People's Hospital, Peking University, Beijing, P.R. China
| |
Collapse
|
27
|
Zarini-Gakiye E, Vaezi G, Parivar K, Sanadgol N. Age and Dose-Dependent Effects of Alpha-Lipoic Acid on Human Microtubule- Associated Protein Tau-Induced Endoplasmic Reticulum Unfolded Protein Response: Implications for Alzheimer's Disease. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 20:451-464. [PMID: 33573583 DOI: 10.2174/1871527320666210126114442] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Revised: 10/21/2020] [Accepted: 11/23/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND In human tauopathies, pathological aggregation of misfolded/unfolded proteins, particularly microtubule-associated protein tau (MAPT, tau) is considered to be an essential mechanism that triggers the induction of endoplasmic reticulum (ER) stress. OBJECTIVE Here, we assessed the molecular effects of natural antioxidant alpha-lipoic acid (ALA) in human tauR406W (hTau)-induced ER unfolded protein response (ERUPR) in fruit flies. METHODS In order to reduce hTau neurotoxicity during brain development, we used a transgenic model of tauopathy where the maximum toxicity was observed in adult flies. Then, the effects of ALA (0.001, 0.005, and 0.025% w/w of diet) in htau-induced ERUPR and behavioral dysfunctions in the ages 20 and 30 days were evaluated in Drosophila melanogaster. RESULTS Data from expression (mRNA and protein) patterns of htau, analysis of eyes external morphology as well as larvae olfactory memory were confirmed by our tauopathy model. Moreover, the expression of ERUPR-related proteins involving Activating Transcription Factor 6 (ATF6), inositol regulating enzyme 1 (IRE1), and protein kinase RNA-like ER kinase (PERK) wase upregulated and locomotor function decreased in both ages of the model flies. Remarkably, the lower dose of ALA modified ERUPR and supported the reduction of behavioral deficits in youngest adults through the enhancement of GRP87/Bip, reduction of ATF6, downregulation of PERK-ATF4 pathway, and activation of the IRE1-XBP1 pathway. On the other hand, only a higher dose of ALA affected the ERUPR via moderation of PERK-ATF4 signaling in the oldest adults. As ALA also exerts higher protective effects on the locomotor function of younger adults when htauR406Wis expressed in all neurons (htau-elav) and mushroom body neurons (htau-ok), we proposed that ALA has age-dependent effects in this model. CONCLUSION Taken together, based on our results, we conclude that aging potentially influences the ALA effective dose and mechanism of action on tau-induced ERUPR. Further molecular studies will warrant possible therapeutic applications of ALA in age-related tauopathies.
Collapse
Affiliation(s)
- Elahe Zarini-Gakiye
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Gholamhassan Vaezi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Kazem Parivar
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Nima Sanadgol
- Department of Biology, Faculty of Sciences, University of Zabol, Zabol, Iran
| |
Collapse
|
28
|
Axonal Organelles as Molecular Platforms for Axon Growth and Regeneration after Injury. Int J Mol Sci 2021; 22:ijms22041798. [PMID: 33670312 PMCID: PMC7918155 DOI: 10.3390/ijms22041798] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 02/06/2023] Open
Abstract
Investigating the molecular mechanisms governing developmental axon growth has been a useful approach for identifying new strategies for boosting axon regeneration after injury, with the goal of treating debilitating conditions such as spinal cord injury and vision loss. The picture emerging is that various axonal organelles are important centers for organizing the molecular mechanisms and machinery required for growth cone development and axon extension, and these have recently been targeted to stimulate robust regeneration in the injured adult central nervous system (CNS). This review summarizes recent literature highlighting a central role for organelles such as recycling endosomes, the endoplasmic reticulum, mitochondria, lysosomes, autophagosomes and the proteasome in developmental axon growth, and describes how these organelles can be targeted to promote axon regeneration after injury to the adult CNS. This review also examines the connections between these organelles in developing and regenerating axons, and finally discusses the molecular mechanisms within the axon that are required for successful axon growth.
Collapse
|
29
|
Ohri SS, Howard RM, Liu Y, Andres KR, Hetman M, Whittemore SR. Oligodendrocyte-specific deletion of Xbp1 exacerbates the endoplasmic reticulum stress response and restricts locomotor recovery after thoracic spinal cord injury. Glia 2021; 69:424-435. [PMID: 32926479 PMCID: PMC8931742 DOI: 10.1002/glia.23907] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 08/21/2020] [Accepted: 08/23/2020] [Indexed: 12/31/2022]
Abstract
The endoplasmic reticulum stress response (ERSR) is activated in various neurodegenerative diseases and/or after CNS traumatic injuries. The ERSR is comprised of three major arms, PERK, IRE-1, and activating transcription factor-6, with the latter two contributing to the unfolded protein response (UPR). PERK activity overlaps with the integrated stress response (ISR) kinases, PKR, HRI, and GCN2 which all signal through, eukaryotic initiation factor 2α, ATF4, and CHOP. All initially attempt to restore endoplasmic reticulum (ER) homeostasis, but if ER stress is unresolved, ATF4/CHOP-mediated cell death is initiated. Here, we investigate the contribution of the inositol-requiring protein-1α-X-box binding protein-1 (XBP1)-mediated UPR signaling pathway to the pathogenesis of spinal cord injury (SCI). We demonstrate that deletion of Xbp1 caused an exacerbated ATF4/CHOP signaling in cultured mouse oligodendrocyte (OL) progenitor cells and enhanced their sensitivity to ER stress. Similar effects were also observed with the Xbp1 pathway inhibitor toyocamycin. Furthermore, OL lineage-specific loss of Xbp1 resulted in enhanced ISR in mice that underwent moderate contusive SCI at the T9 level. Consistently, post-injury recovery of hindlimb locomotion and white matter sparing were reduced in OL Xbp1-deficient mice, which correlated with chronically decreased relative density of OPCs and OLs at the injury epicenter at 6 weeks post-SCI. We conclude that the IRE1-XBP1-mediated UPR signaling pathway contributes to restoration of ER homeostasis in OLs and is necessary for enhanced white matter sparing and functional recovery post-SCI.
Collapse
Affiliation(s)
- Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
- Departments of Neurological Surgery, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
| | - Russell M. Howard
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
- Departments of Neurological Surgery, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
| | - Yu Liu
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
- Departments of Neurological Surgery, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
| | - Kariena R. Andres
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
- Departments of Neurological Surgery, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
- Departments of Neurological Surgery, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
- Departments of Pharmacology & Toxicology, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
- Departments of Anatomical Sciences & Neurobiology, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
| | - Scott R. Whittemore
- Kentucky Spinal Cord Injury Research Center, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
- Departments of Neurological Surgery, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
- Departments of Pharmacology & Toxicology, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
- Departments of Anatomical Sciences & Neurobiology, University of Louisville, School of Medicine, 511 S. Floyd St., MDR 616, Louisville, KY 40202, USA
| |
Collapse
|
30
|
Potential of extracellular vesicles in the Parkinson's disease - Pathological mediators and biomarkers. Neurochem Int 2021; 144:104974. [PMID: 33485881 DOI: 10.1016/j.neuint.2021.104974] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 01/08/2023]
Abstract
Parkinson's disease (PD) is an age-related neurodegenerative disorder characterized by the progressive deterioration of motor function. Histopathologically, it is widely accepted that the progressive death of selected dopaminergic neuronal populations and the accumulation of hallmark Lewy bodies (LBs) composed of α-synuclein (α-syn) might be the two vital pathogenesis. Extracellular vesicles (EVs) are cell-derived membranous vesicles that are liberated from virtually all cell types including neurons, and harbor a variety of proteins, DNA, mRNA, and lipids. The roles of these vesicles include cell-cell signaling, removal of unwanted proteins, and transfer of pathogens (including misfolded proteins) between cells. In PD, EVs not only enhance the spread of α-syn at distant sites and reduce their clearance but also mediate other PD pathogenesis such as the activation of microglia and the dysfunction of autophagy and lysosomal degradation systems. Recently, clinical evidence for the diagnostic performance of EV-associated biomarkers, particularly exosome biomarkers, has merged. In this regard, we reviewed the recent understanding of the biological roles of EVs as important tools for biomarker discovery and pathological regulators of PD, and discuss the main concerns and challenges for the application of EV biomarkers in the clinical setting.
Collapse
|
31
|
Chu HS, Peterson C, Jun A, Foster J. Targeting the integrated stress response in ophthalmology. Curr Eye Res 2021; 46:1075-1088. [PMID: 33474991 DOI: 10.1080/02713683.2020.1867748] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Purpose: To summarize the Integrated Stress Response (ISR) in the context of ophthalmology, with special interest on the cornea and anterior segment. Results: The ISR is a powerful and conserved signaling pathway that allows for cells to respond to a diverse array of both intracellular and extracellular stressors. The pathway is classically responsible for coordination of the cellular response to amino acid starvation, ultraviolet light, heme dysregulation, viral infection, and unfolded protein. Under normal circumstances, it is considered pro-survival and a necessary mechanism through which protein translation is controlled. However, in cases of severe or prolonged stress the pathway can promote apoptosis, and loss of normal cellular phenotype. The activation of this pathway culminates in the global inhibition of cap-dependent protein translation and the canonical expression of the activating transcription factor 4 (ATF4). Conclusion:The eye is uniquely exposed to ISR responsive stressors due to its environmental exposure and relative isolation from the circulatory system which are necessary for its function. We will discuss how this pathway is critical for the proper function of the tissue, its role in development, as well as how targeting of the pathway could alleviate key aspects of diverse ophthalmic diseases.
Collapse
Affiliation(s)
- Hsiao-Sang Chu
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA.,Department of Ophthalmology, National Taiwan University Hospital, College of Medicine, National Taiwan University, Taipei City, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei City, Taiwan
| | - Cornelia Peterson
- Department of Molecular & Comparative Pathobiology, Johns Hopkins University, Baltimore, MD, USA
| | - Albert Jun
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| | - James Foster
- Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
32
|
Layalle S, They L, Ourghani S, Raoul C, Soustelle L. Amyotrophic Lateral Sclerosis Genes in Drosophila melanogaster. Int J Mol Sci 2021; 22:ijms22020904. [PMID: 33477509 PMCID: PMC7831090 DOI: 10.3390/ijms22020904] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating adult-onset neurodegenerative disease characterized by the progressive degeneration of upper and lower motoneurons. Most ALS cases are sporadic but approximately 10% of ALS cases are due to inherited mutations in identified genes. ALS-causing mutations were identified in over 30 genes with superoxide dismutase-1 (SOD1), chromosome 9 open reading frame 72 (C9orf72), fused in sarcoma (FUS), and TAR DNA-binding protein (TARDBP, encoding TDP-43) being the most frequent. In the last few decades, Drosophila melanogaster emerged as a versatile model for studying neurodegenerative diseases, including ALS. In this review, we describe the different Drosophila ALS models that have been successfully used to decipher the cellular and molecular pathways associated with SOD1, C9orf72, FUS, and TDP-43. The study of the known fruit fly orthologs of these ALS-related genes yielded significant insights into cellular mechanisms and physiological functions. Moreover, genetic screening in tissue-specific gain-of-function mutants that mimic ALS-associated phenotypes identified disease-modifying genes. Here, we propose a comprehensive review on the Drosophila research focused on four ALS-linked genes that has revealed novel pathogenic mechanisms and identified potential therapeutic targets for future therapy.
Collapse
Affiliation(s)
- Sophie Layalle
- The Neuroscience Institute of Montpellier, INSERM, University of Montpellier, 34091 Montpellier, France; (S.L.); (L.T.); (S.O.)
| | - Laetitia They
- The Neuroscience Institute of Montpellier, INSERM, University of Montpellier, 34091 Montpellier, France; (S.L.); (L.T.); (S.O.)
| | - Sarah Ourghani
- The Neuroscience Institute of Montpellier, INSERM, University of Montpellier, 34091 Montpellier, France; (S.L.); (L.T.); (S.O.)
| | - Cédric Raoul
- The Neuroscience Institute of Montpellier, INSERM, University of Montpellier, 34091 Montpellier, France; (S.L.); (L.T.); (S.O.)
- Laboratory of Neurobiology, Kazan Federal University, 420008 Kazan, Russia
- Correspondence: (C.R.); (L.S.)
| | - Laurent Soustelle
- The Neuroscience Institute of Montpellier, INSERM, University of Montpellier, 34091 Montpellier, France; (S.L.); (L.T.); (S.O.)
- Correspondence: (C.R.); (L.S.)
| |
Collapse
|
33
|
Medinas DB, Hazari Y, Hetz C. Disruption of Endoplasmic Reticulum Proteostasis in Age-Related Nervous System Disorders. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2021; 59:239-278. [PMID: 34050870 DOI: 10.1007/978-3-030-67696-4_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Endoplasmic reticulum (ER) stress is a prominent cellular alteration of diseases impacting the nervous system that are associated to the accumulation of misfolded and aggregated protein species during aging. The unfolded protein response (UPR) is the main pathway mediating adaptation to ER stress, but it can also trigger deleterious cascades of inflammation and cell death leading to cell dysfunction and neurodegeneration. Genetic and pharmacological studies in experimental models shed light into molecular pathways possibly contributing to ER stress and the UPR activation in human neuropathies. Most of experimental models are, however, based on the overexpression of mutant proteins causing familial forms of these diseases or the administration of neurotoxins that induce pathology in young animals. Whether the mechanisms uncovered in these models are relevant for the etiology of the vast majority of age-related sporadic forms of neurodegenerative diseases is an open question. Here, we provide a systematic analysis of the current evidence linking ER stress to human pathology and the main mechanisms elucidated in experimental models. Furthermore, we highlight the recent association of metabolic syndrome to increased risk to undergo neurodegeneration, where ER stress arises as a common denominator in the pathogenic crosstalk between peripheral organs and the nervous system.
Collapse
Affiliation(s)
- Danilo B Medinas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile. .,Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, University of Chile, Santiago, Chile. .,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.
| | - Younis Hazari
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile. .,Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, University of Chile, Santiago, Chile. .,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile. .,Buck Institute for Research on Aging, Novato, CA, USA.
| |
Collapse
|
34
|
Xu W, Wang C, Hua J. X-box binding protein 1 (XBP1) function in diseases. Cell Biol Int 2020; 45:731-739. [PMID: 33325615 DOI: 10.1002/cbin.11533] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/12/2020] [Accepted: 12/13/2020] [Indexed: 12/15/2022]
Abstract
The accumulation of unfolded or misfolded proteins in the endoplasmic reticulum (ER) causes endoplasmic reticulum stress (ERS), which is characteristic of cells with high levels of secretory activity and is involved in a variety of diseases. In response to ERS, cells initiate an adaptive process named the unfolding protein response (UPR) to maintain intracellular homeostasis and survival. However, long term and unresolved ERS can also induce apoptosis. As the most conserved signaling branch of UPR, the IRE1-XBP1 pathway plays an important role in both physiological and pathological states, and its activity has a profound impact on disease progression and prognosis. Here, the latest research progress of IRE1-XBP1 pathway in cancer, metabolic diseases, and other diseases was briefly introduced, and the relationship between several diseases and this pathway was analyzed. Besides, the new understanding and prospect of IRE1-XBP1 pathway regulating male reproduction were reviewed.
Collapse
Affiliation(s)
- Wenjing Xu
- Shaanxi Centre of Stem Cells Engineering & Technology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| | - Congrong Wang
- Translational Medical Center for Stem Cell Therapy, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jinlian Hua
- Shaanxi Centre of Stem Cells Engineering & Technology, College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi, China
| |
Collapse
|
35
|
Sahib S, Sharma A, Menon PK, Muresanu DF, Castellani RJ, Nozari A, Lafuente JV, Bryukhovetskiy I, Tian ZR, Patnaik R, Buzoianu AD, Wiklund L, Sharma HS. Cerebrolysin enhances spinal cord conduction and reduces blood-spinal cord barrier breakdown, edema formation, immediate early gene expression and cord pathology after injury. PROGRESS IN BRAIN RESEARCH 2020; 258:397-438. [PMID: 33223040 DOI: 10.1016/bs.pbr.2020.09.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Spinal cord evoked potentials (SCEP) are good indicators of spinal cord function in health and disease. Disturbances in SCEP amplitudes and latencies during spinal cord monitoring predict spinal cord pathology following trauma. Treatment with neuroprotective agents preserves SCEP and reduces cord pathology after injury. The possibility that cerebrolysin, a balanced composition of neurotrophic factors improves spinal cord conduction, attenuates blood-spinal cord barrier (BSCB) disruption, edema formation, and cord pathology was examined in spinal cord injury (SCI). SCEP is recorded from epidural space over rat spinal cord T9 and T12 segments after peripheral nerves stimulation. SCEP consists of a small positive peak (MPP), followed by a prominent negative peak (MNP) that is stable before SCI. A longitudinal incision (2mm deep and 5mm long) into the right dorsal horn (T10 and T11 segments) resulted in an immediate long-lasting depression of the rostral MNP with an increase in the latencies. Pretreatment with either cerebrolysin (CBL 5mL/kg, i.v. 30min before) alone or TiO2 nanowired delivery of cerebrolysin (NWCBL 2.5mL/kg, i.v.) prevented the loss of MNP amplitude and even enhanced further from the pre-injury level after SCI without affecting latencies. At 5h, SCI induced edema, BSCB breakdown, and cell injuries were significantly reduced by CBL and NWCBL pretreatment. Interestingly this effect on SCEP and cord pathology was still prominent when the NWCBL was delivered 2min after SCI. Moreover, expressions of c-fos and c-jun genes that are prominent at 5h in untreated SCI are also considerably reduced by CBL and NWCBL treatment. These results are the first to show that CBL and NWCBL enhanced SCEP activity and thwarted the development of cord pathology after SCI. Furthermore, NWCBL in low doses has superior neuroprotective effects on SCEP and cord pathology, not reported earlier. The functional significance and future clinical potential of CBL and NWCBL in SCI are discussed.
Collapse
Affiliation(s)
- Seaab Sahib
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Aruna Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| | - Preeti K Menon
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden; Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Dafin F Muresanu
- Department of Clinical Neurosciences, University of Medicine & Pharmacy, Cluj-Napoca, Romania; "RoNeuro" Institute for Neurological Research and Diagnostic, Cluj-Napoca, Romania
| | - Rudy J Castellani
- Department of Pathology, University of Maryland, Baltimore, MD, United States
| | - Ala Nozari
- Anesthesiology & Intensive Care, Massachusetts General Hospital, Boston, MA, United States
| | - José Vicente Lafuente
- LaNCE, Department of Neuroscience, University of the Basque Country (UPV/EHU), Leioa, Bizkaia, Spain
| | - Igor Bryukhovetskiy
- Department of Fundamental Medicine, School of Biomedicine, Far Eastern Federal University, Vladivostok, Russia; Laboratory of Pharmacology, National Scientific Center of Marine Biology, Far East Branch of the Russian Academy of Sciences, Vladivostok, Russia
| | - Z Ryan Tian
- Department of Chemistry & Biochemistry, University of Arkansas, Fayetteville, AR, United States
| | - Ranjana Patnaik
- Department of Biomaterials, School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University, Varanasi, India
| | - Anca D Buzoianu
- Department of Clinical Pharmacology and Toxicology, "Iuliu Hatieganu" University of Medicine and Pharmacy, Cluj-Napoca, Romania
| | - Lars Wiklund
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden
| | - Hari Shanker Sharma
- International Experimental Central Nervous System Injury & Repair (IECNSIR), Department of Surgical Sciences, Anesthesiology & Intensive Care Medicine, Uppsala University Hospital, Uppsala University, Uppsala, Sweden.
| |
Collapse
|
36
|
Saraswat Ohri S, Burke DA, Andres KR, Hetman M, Whittemore SR. Acute Neural and Proteostasis Messenger Ribonucleic Acid Levels Predict Chronic Locomotor Recovery after Contusive Spinal Cord Injury. J Neurotrauma 2020; 38:365-372. [PMID: 33076743 DOI: 10.1089/neu.2020.7258] [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] [Indexed: 12/12/2022] Open
Abstract
One of the difficulties in identifying novel therapeutic strategies to manage central nervous system (CNS) trauma is the need for behavioral assays to assess chronic functional recovery. In vitro assays and/or acute behavioral assessments cannot accurately predict long-term functional outcome. Using data from 13 independent T9 moderate contusive spinal cord injury (SCI) studies, we asked whether the ratio of acute (24-72 h post-injury) changes in the levels of neuron-, oligodendrocyte-, astrocyte-specific and/or endoplasmic reticulum stress response (ERSR) messenger ribonucleic acids (mRNAs) could predict the extent of chronic functional recovery. Increased levels of neuron, oligodendrocyte, and astrocyte mRNAs all correlated with enhanced Basso Mouse Scale (BMS) scores. Reduced levels of the ERSR mRNAs Atf4 and Chop correlate with improved chronic locomotor function. Neither neural or ERSR mRNAs were predictive for chronic recovery across all behavioral changes. The ratio of oligodendrocyte/ERSR mRNAs, however, did predict "improved," "no change," or "worse" functional recovery. Neuronal/ERSR mRNA ratios predicted functional improvement, but could not distinguish between worse or no change outcomes. Astrocyte/ERSR mRNA ratios were not predictive. This approach can be used to confirm biological action of injected drugs in vivo and to optimize dose and therapeutic window. It may prove useful in cervical and lumbar SCI and in other traumatic CNS injuries such as traumatic brain injury and stroke, where prevention of neuronal loss is paramount to functional recovery. Although the current analysis was directed toward ERSR whose activity was targeted in all but one study, acute mRNA markers for other pathophysiological cascades may be as predictive of chronic recovery when those cascades are targeted for neuroprotection.
Collapse
Affiliation(s)
- Sujata Saraswat Ohri
- Kentucky Spinal Cord Injury Research Center and Departments of University of Louisville School of Medicine, Louisville, Kentucky, USA.,Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Darlene A Burke
- Kentucky Spinal Cord Injury Research Center and Departments of University of Louisville School of Medicine, Louisville, Kentucky, USA.,Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Kariena R Andres
- Kentucky Spinal Cord Injury Research Center and Departments of University of Louisville School of Medicine, Louisville, Kentucky, USA.,Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Michal Hetman
- Kentucky Spinal Cord Injury Research Center and Departments of University of Louisville School of Medicine, Louisville, Kentucky, USA.,Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA.,Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA.,Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| | - Scott R Whittemore
- Kentucky Spinal Cord Injury Research Center and Departments of University of Louisville School of Medicine, Louisville, Kentucky, USA.,Neurological Surgery, University of Louisville School of Medicine, Louisville, Kentucky, USA.,Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, Kentucky, USA.,Pharmacology and Toxicology, University of Louisville School of Medicine, Louisville, Kentucky, USA
| |
Collapse
|
37
|
Grandjean JMD, Madhavan A, Cech L, Seguinot BO, Paxman RJ, Smith E, Scampavia L, Powers ET, Cooley CB, Plate L, Spicer TP, Kelly JW, Wiseman RL. Pharmacologic IRE1/XBP1s activation confers targeted ER proteostasis reprogramming. Nat Chem Biol 2020; 16:1052-1061. [PMID: 32690944 PMCID: PMC7502540 DOI: 10.1038/s41589-020-0584-z] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 06/05/2020] [Indexed: 12/14/2022]
Abstract
Activation of the IRE1/XBP1s signaling arm of the unfolded protein response (UPR) is a promising strategy to correct defects in endoplasmic reticulum (ER) proteostasis implicated in diverse diseases. However, no pharmacologic activators of this pathway identified to date are suitable for ER proteostasis remodeling through selective activation of IRE1/XBP1s signaling. Here, we use high-throughput screening to identify non-toxic compounds that induce ER proteostasis remodeling through IRE1/XBP1s activation. We employ transcriptional profiling to stringently confirm that our prioritized compounds selectively activate IRE1/XBP1s signaling without activating other cellular stress-responsive signaling pathways. Furthermore, we demonstrate that our compounds improve ER proteostasis of destabilized variants of amyloid precursor protein (APP) through an IRE1-dependent mechanism and reduce APP-associated mitochondrial toxicity in cellular models. These results establish highly selective IRE1/XBP1s activating compounds that can be widely employed to define the functional importance of IRE1/XBP1s activity for ER proteostasis regulation in the context of health and disease.
Collapse
Affiliation(s)
- Julia M D Grandjean
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Aparajita Madhavan
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Lauren Cech
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Bryan O Seguinot
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Ryan J Paxman
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | - Emery Smith
- Scripps Research Molecular Screening Center, The Scripps Research Institute, Jupiter, FL, USA
| | - Louis Scampavia
- Scripps Research Molecular Screening Center, The Scripps Research Institute, Jupiter, FL, USA
| | - Evan T Powers
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
| | | | - Lars Plate
- Departments of Chemistry and Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Timothy P Spicer
- Scripps Research Molecular Screening Center, The Scripps Research Institute, Jupiter, FL, USA
| | - Jeffery W Kelly
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA, USA
- The Skaggs Institute for Chemical Biology, The Scripps Research Institute, La Jolla, CA, USA
| | - R Luke Wiseman
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA.
| |
Collapse
|
38
|
Li Y, Zhang J, Zhou K, Xie L, Xiang G, Fang M, Han W, Wang X, Xiao J. Elevating sestrin2 attenuates endoplasmic reticulum stress and improves functional recovery through autophagy activation after spinal cord injury. Cell Biol Toxicol 2020; 37:401-419. [PMID: 32740777 DOI: 10.1007/s10565-020-09550-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Accepted: 07/27/2020] [Indexed: 12/15/2022]
Abstract
Spinal cord injury (SCI) is a devastating neurological trauma that causes losses of motor and sensory function. Sestrin2, also known as hypoxia inducible gene 95, is emerging as a critical determinant of cell homeostasis in response to cellular stress. However, the role of sestrin2 in the neuronal response to endoplasmic reticulum (ER) stress and the potential mechanism remain undefined. In this study, we investigated the effects of sestrin2 on ER stress and delineated an underlying molecular mechanism after SCI. Here, we found that elevated sestrin2 is a protective process in neurons against chemical ER stress induced by tunicamycin (TM) or traumatic invasion, while treatment with PERK inhibitor or knockdown of ATF4 reduces sestrin2 expression upon ER stress. In addition, we demonstrated that overexpression of sestrin2 limits ER stress, promoting neuronal survival and improving functional recovery after SCI, which is associated with activation of autophagy and restoration of autophagic flux mediated by sestrin2. Moreover, we also found that sestrin2 activates autophagy dependent on the AMPK-mTOR signaling pathway. Consistently, inhibition of AMPK abrogates the effect of sestrin2 on the activation of autophagy, and blockage of autophagic flux abolishes the effect of sestrin2 on limiting ER stress and neural death. Together, our data reveal that upregulation of sestrin2 is an important resistance mechanism of neurons to ER stress and the potential role of sestrin2 as a therapeutic target for SCI. Graphical abstract.
Collapse
Affiliation(s)
- Yao Li
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.,Research Units of Clinical Translation of Cell Growth Factors and Diseases Research of Chinese Academy of Medical Science, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Jing Zhang
- Research Units of Clinical Translation of Cell Growth Factors and Diseases Research of Chinese Academy of Medical Science, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.,Research Units of Clinical Translation of Cell Growth Factors and Diseases Research of Chinese Academy of Medical Science, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Ling Xie
- Research Units of Clinical Translation of Cell Growth Factors and Diseases Research of Chinese Academy of Medical Science, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Guangheng Xiang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.,Research Units of Clinical Translation of Cell Growth Factors and Diseases Research of Chinese Academy of Medical Science, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Mingqiao Fang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.,Research Units of Clinical Translation of Cell Growth Factors and Diseases Research of Chinese Academy of Medical Science, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.,The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Wen Han
- Research Units of Clinical Translation of Cell Growth Factors and Diseases Research of Chinese Academy of Medical Science, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| | - Jian Xiao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China. .,Research Units of Clinical Translation of Cell Growth Factors and Diseases Research of Chinese Academy of Medical Science, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China.
| |
Collapse
|
39
|
Hetz C, Zhang K, Kaufman RJ. Mechanisms, regulation and functions of the unfolded protein response. Nat Rev Mol Cell Biol 2020; 21:421-438. [PMID: 32457508 DOI: 10.1038/s41580-020-0250-z] [Citation(s) in RCA: 1546] [Impact Index Per Article: 309.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/14/2020] [Indexed: 12/21/2022]
Abstract
Cellular stress induced by the abnormal accumulation of unfolded or misfolded proteins at the endoplasmic reticulum (ER) is emerging as a possible driver of human diseases, including cancer, diabetes, obesity and neurodegeneration. ER proteostasis surveillance is mediated by the unfolded protein response (UPR), a signal transduction pathway that senses the fidelity of protein folding in the ER lumen. The UPR transmits information about protein folding status to the nucleus and cytosol to adjust the protein folding capacity of the cell or, in the event of chronic damage, induce apoptotic cell death. Recent advances in the understanding of the regulation of UPR signalling and its implications in the pathophysiology of disease might open new therapeutic avenues.
Collapse
Affiliation(s)
- Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile. .,FONDAP Center for Geroscience Brain Health and Metabolism (GERO), Santiago, Chile. .,Program of Cellular and Molecular Biology, Institute of Biomedical Science, University of Chile, Santiago, Chile. .,Buck Institute for Research on Aging, Novato, CA, USA.
| | - Kezhong Zhang
- Center for Molecular Medicine and Genetics, Wayne State University School of Medicine, Detroit, MI, USA. .,Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA.
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
40
|
Montibeller L, Tan LY, Kim JK, Paul P, de Belleroche J. Tissue-selective regulation of protein homeostasis and unfolded protein response signalling in sporadic ALS. J Cell Mol Med 2020; 24:6055-6069. [PMID: 32324341 PMCID: PMC7294118 DOI: 10.1111/jcmm.15170] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/21/2020] [Accepted: 02/24/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a disorder that affects motor neurons in motor cortex and spinal cord, and the degeneration of both neuronal populations is a critical feature of the disease. Abnormalities in protein homeostasis (proteostasis) are well established in ALS. However, they have been investigated mostly in spinal cord but less so in motor cortex. Herein, we monitored the unfolded protein (UPR) and heat shock response (HSR), two major proteostasis regulatory pathways, in human post‐mortem tissue derived from the motor cortex of sporadic ALS (SALS) and compared them to those occurring in spinal cord. Although the UPR was activated in both tissues, specific expression of select UPR target genes, such as PDIs, was observed in motor cortex of SALS cases strongly correlating with oligodendrocyte markers. Moreover, we found that endoplasmic reticulum‐associated degradation (ERAD) and HSR genes, which were activated predominately in spinal cord, correlated with the expression of neuronal markers. Our results indicate that proteostasis is strongly and selectively activated in SALS motor cortex and spinal cord where subsets of these genes are associated with specific cell type. This study expands our understanding of convergent molecular mechanisms occurring in motor cortex and spinal cord and highlights cell type–specific contributions.
Collapse
Affiliation(s)
- Luigi Montibeller
- Neurogenetics Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Li Yi Tan
- Neurogenetics Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Joo Kyung Kim
- Neurogenetics Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Praveen Paul
- Neurogenetics Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| | - Jacqueline de Belleroche
- Neurogenetics Group, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, UK
| |
Collapse
|
41
|
Gorbatyuk MS, Starr CR, Gorbatyuk OS. Endoplasmic reticulum stress: New insights into the pathogenesis and treatment of retinal degenerative diseases. Prog Retin Eye Res 2020; 79:100860. [PMID: 32272207 DOI: 10.1016/j.preteyeres.2020.100860] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2019] [Revised: 03/08/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022]
Abstract
Physiological equilibrium in the retina depends on coordinated work between rod and cone photoreceptors and can be compromised by the expression of mutant proteins leading to inherited retinal degeneration (IRD). IRD is a diverse group of retinal dystrophies with multifaceted molecular mechanisms that are not fully understood. In this review, we focus on the contribution of chronically activated unfolded protein response (UPR) to inherited retinal pathogenesis, placing special emphasis on studies employing genetically modified animal models. As constitutively active UPR in degenerating retinas may activate pro-apoptotic programs associated with oxidative stress, pro-inflammatory signaling, dysfunctional autophagy, free cytosolic Ca2+ overload, and altered protein synthesis rate in the retina, we focus on the regulatory mechanisms of translational attenuation and approaches to overcoming translational attenuation in degenerating retinas. We also discuss current research on the role of the UPR mediator PERK and its downstream targets in degenerating retinas and highlight the therapeutic benefits of reprogramming PERK signaling in preclinical animal models of IRD. Finally, we describe pharmacological approaches targeting UPR in ocular diseases and consider their potential applications to IRD.
Collapse
Affiliation(s)
- Marina S Gorbatyuk
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA.
| | - Christopher R Starr
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA
| | - Oleg S Gorbatyuk
- The University of Alabama at Birmingham, Department of Optometry and Vision Science, School of Optometry, USA
| |
Collapse
|
42
|
Chi L, Jiao D, Nan G, Yuan H, Shen J, Gao Y. miR-9-5p attenuates ischemic stroke through targeting ERMP1-mediated endoplasmic reticulum stress. Acta Histochem 2019; 121:151438. [PMID: 31500865 DOI: 10.1016/j.acthis.2019.08.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/07/2019] [Accepted: 08/30/2019] [Indexed: 12/20/2022]
Abstract
Ischemic stroke (IS) is a cerebrovascular disease with serious neurological function impairment, which may activate endoplasmic reticulum (ER) stress. However, the underlying regulatory mechanism of ER stress under IS remains unclear. miR-9-5p is enriched in the brain tissues and plays a role in the pathological process of IS. Therefore, the purpose of this study is to explore the effect of miR-9 on ER stress and underlying mechanism in IS. Here, a middle cerebral artery occlusion (MCAO) rat model was utilized to examine the alteration of brain pathology, and the expressions of miR-9 and ER stress-related proteins. Then SH-SY5Y cells with oxygen-glucose deprivation (OGD) were performed to further evaluate the functional role of miR-9 and preliminary mechanism. The results showed that miR-9 levels were decreased in the ischemic region of rats after MCAO. MCAO significantly increased the brain infract volume, reduced Nissl bodies and cell apoptosis, and increased ER stress-related proteins (ERMP1, GRP78, p-PERK, p-eIF2α and CHOP). Furthermore, overexpression of miR-9 by miR-9 mimics increased cell viability, inhibited LDH activity and cell apoptosis, and inactivated ER stress in OGD-neurons. Luciferase activity results showed that miR-9 negatively regulated ERMP1 expression by directly targeting ERMP1 3' UTR. Subsequently, we found that ERMP1 overexpression reversed the inhibition of miR-9 on GRP78-PERK-CHOP pathway in OGD neurons. In summary, our results suggest that the attenuation of miR-9 on ischemic injury may be involved in targeting ERMP1-mediated ER stress, which provides an available target for IS treatment.
Collapse
|
43
|
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: 2.7] [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
|
44
|
Mori H, Saito YU, Iwahashi S, Ikemoto T, Imura S, Morine Y, Shimada M. Impact of Bevacizumab on Liver Damage After Massive Hepatectomy in Rats. In Vivo 2019; 33:1469-1476. [PMID: 31471394 DOI: 10.21873/invivo.11626] [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: 06/10/2019] [Revised: 07/08/2019] [Accepted: 07/12/2019] [Indexed: 11/10/2022]
Abstract
BACKGROUND The aim of this study was to evaluate the impact of pretreatment with bevacizumab on liver damage in a rat model of massive hepatectomy (Hx) model, as a surrogate model of massive Hx for liver metastasis from colorectal cancer. MATERIALS AND METHODS Male Wister rats (n=24) were separated into the following two groups: 90% Hx and 90% Hx plus bevacizumab group. Bevacizumab (5 mg/kg) was injected intraperitoneally 7 days before Hx. Samples of blood and remnant liver tissue were obtained 24 hours after hepatectomy and the following parameters were evaluated: Biochemical analysis; liver regeneration rate; survival rate; and real-time polymerase chain reaction for interleukin-1 beta (Il1b), tumor necrosis factor alpha (Tnfa), matrix metalloproteinase (Mmp) 2 and Mmp9 mRNA. In addition, samples of whole liver tissue were obtained immediately before Hx and real-time polymerase chain reaction was performed for X-box binding protein 1 (Xbp1), activating transcription factor 6 (Atf6), C/EBP homologous protein (Chop), glucose-regulated protein 78 (Grp78) and heat-shock protein 70 (Hsp70), as markers of endoplasmic reticulum stress response. RESULTS The levels of transaminases 24 hours after Hx were significantly reduced in the group pretreated with bevacizumab compared to that not pretreated (p<0.05). The liver regeneration rate at 24 hours after Hx was significantly increased in the group pretreated with bevacizumab compared with the group which underwent Hx alone (p<0.05). The survival rate for the group pretreated with bevacizumab tended to be higher than that of the Hx-only group, 72 hours after Hx (p=0.09). The expressions of Il1b, Mmp2 and Mmp9 mRNA 24 hours after Hx in the group pretreated with bevacizumab tended to be lower than that of rats which underwent Hx alone (p=0.11, 0.09 and 0.15, respectively). The expression of Xbp1, Chop, Grp78 and Hsp70 mRNA immediately before Hx in the group pretreated with bevacizumab were significantly higher than the 90% Hx group (p<0.05). CONCLUSION Bevacizumab pretreatment had protective effects on liver injury after massive hepatectomy in rats, apparently via the induction of the endoplasmic reticulum stress response, i.e. the so-called unfolded protein response.
Collapse
Affiliation(s)
- Hiroki Mori
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Y U Saito
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Shuichi Iwahashi
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Tetsuya Ikemoto
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Satoru Imura
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Yuji Morine
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| | - Mitsuo Shimada
- Department of Surgery, The University of Tokushima, Tokushima, Japan
| |
Collapse
|
45
|
Zhu Y, Zhang L, Fu R, Gao L, Feng G, Du C, Wang Z, Yan X. The change tendency of endoplasmic reticulum stress associated proteins in rats with spinal cord injury. Am J Transl Res 2019; 11:1938-1947. [PMID: 31105809 PMCID: PMC6511803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 03/17/2019] [Indexed: 06/09/2023]
Abstract
To investigate endoplasmic reticulum (ER) stress reactions in spinal cord injury rats by evaluating the expression of the glucose-regulated protein 78 (GRP78), C/EBP homologous transcription factor protein (CHOP), X-box binding protein 1 (XBP1), Eif-2α and Bad. SCI models were established using adult female mice. After SCI, the expression of endoplasmic reticulum stress-induced apoptosis proteins were examined in the mice at specific time points using immunohistochemistry and western blot. The results of immunohistochemistry showed that in spinal cord gray matter, Chop, Grp78, XBP1, Eif-2α and Bad were specifically detected in the cytoplasm of the cell. Compare with the SCI group, there was little expression in normal group and sham group. The expression of ER stress-induced apoptosis proteins were significantly increased after spinal cord injury, and the absolute expression was higher than normal group (P < 0.05). Western-Blot results showed that compare with the SCI group, there were little expression of ER stress-induced apoptosis proteins in normal group and sham group. The expression of ER stress-induced apoptosis proteins were significantly increased after spinal cord injury, and the absolute expression was higher than normal group (P < 0.05). These results suggest that some ER stress-induced apoptosis proteins, such as Chop, Grp78, XBP1, Eif-2α and Bad, were activated after spinal cord injury, but the precise regulatory mechanisms remain unclear. In the future, understanding of the precise mechanism of ER stress-mediated apoptosis in SCI may lead to the development of novel treatment strategies.
Collapse
Affiliation(s)
- Yonglin Zhu
- Shandong University27 Shanda Nanlu, Jinan 250100, Shandong, China
- Department of Bone and Joint, Yantai Affiliated Hospital of Binzhou Medical UniversityYantai 264001, Shandong, China
| | - Luping Zhang
- Institute of Human Anatomy and Histology and Embryology, Otology and Neuroscience Center, Binzhou Medical University346 Guanhai Road, Yantai 264003, Shandong, China
| | - Rongzhan Fu
- Shandong Provincial Qianfoshan HospitalJinan 250014, Shandong, China
| | - Limin Gao
- Institute of Human Anatomy and Histology and Embryology, Otology and Neuroscience Center, Binzhou Medical University346 Guanhai Road, Yantai 264003, Shandong, China
| | - Guoying Feng
- Institute of Human Anatomy and Histology and Embryology, Otology and Neuroscience Center, Binzhou Medical University346 Guanhai Road, Yantai 264003, Shandong, China
| | - Chao Du
- Institute of Human Anatomy and Histology and Embryology, Otology and Neuroscience Center, Binzhou Medical University346 Guanhai Road, Yantai 264003, Shandong, China
| | - Zhaojie Wang
- Department of Bone and Joint, Yantai Affiliated Hospital of Binzhou Medical UniversityYantai 264001, Shandong, China
| | - Xinfeng Yan
- Shandong University27 Shanda Nanlu, Jinan 250100, Shandong, China
- Shandong Provincial Qianfoshan HospitalJinan 250014, Shandong, China
| |
Collapse
|
46
|
Farley MM, Watkins TA. Intrinsic Neuronal Stress Response Pathways in Injury and Disease. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2019; 13:93-116. [PMID: 29414247 DOI: 10.1146/annurev-pathol-012414-040354] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
From injury to disease to aging, neurons, like all cells, may face various insults that can impact their function and survival. Although the consequences are substantially dictated by the type, context, and severity of insult, distressed neurons are far from passive. Activation of cellular stress responses aids in the preservation or restoration of nervous system function. However, stress responses themselves can further advance neuropathology and contribute significantly to neuronal dysfunction and neurodegeneration. Here we explore the recent advances in defining the cellular stress responses within neurodegenerative diseases and neuronal injury, and we emphasize axonal injury as a well-characterized model of neuronal insult. We highlight key findings and unanswered questions about neuronal stress response pathways, from the initial detection of cellular insults through the underlying mechanisms of the responses to their ultimate impact on the fates of distressed neurons.
Collapse
Affiliation(s)
- Madeline M Farley
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas 77030;
| | - Trent A Watkins
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas 77030;
| |
Collapse
|
47
|
Chen Z, Guo H, Lu Z, Sun K, Jin Q. Hyperglycemia aggravates spinal cord injury through endoplasmic reticulum stress mediated neuronal apoptosis, gliosis and activation. Biomed Pharmacother 2019; 112:108672. [PMID: 30784940 DOI: 10.1016/j.biopha.2019.108672] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 01/29/2019] [Accepted: 02/05/2019] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Hyperglycemia has been shown to influence prognostic outcome of spinal cord injury (SCI). However, the corresponding mechanism is not very clear. AIM This study is expected to explore the role of endoplasmic reticulum (ER) stress in hyperglycemia aggravated SCI. METHODS Hyperglycemia was established in rats by intraperitoneal (i.p.) injection of streptozotocin. SCI was performed at the T10 of spinal cord through weight dropping. ER stress was suppressed by oral gavage of 4-PBA. ER stress, histological change of the injured spinal cords, neuronal apoptosis, demyelination, glial proliferation, inflammatory factor production, blood-spinal cord barrier (BSCB) permeability, TJ (Occludin, Claudin5) and AJ (β-catenin, P120) protein degradation, and locomotor recovery were determined using western blotting, immunohistochemistry, HE staining, Evan's Blue assay, BBB scores and inclined plane test, respectively. In vitro, rat spinal cord neurons cells (RSCNCs) and cerebral microvascular endothelial cells (RCMECs) were stimulated with high glucose (HG) and/or thapsigargin (TG). The effects of HG and/or TG on RSCNCs apoptosis, and AJ and TJ expression by RCMECs were evaluated with flow cytometry and western blotting, respectively. RESULTS Hyperglycemic rats exhibited enhanced ER stress, increased neuronal apoptosis, aggravated demyelination, increased glial proliferation and inflammatory factors secretion, more serious BSCB disruption and disturbed locomotor recovery. ER stress inhibition alleviated hyperglycemia induced adverse effect on neuronal apoptosis and BSCB permeability, whereas showed little influence on glial activation and inflammation. CONCLUSION ER stress was aggravated in hyperglycemic rats after SCI, and subsequently promoted neuronal apoptosis and BSCB disruption in rats.
Collapse
Affiliation(s)
- Zhirong Chen
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Haohui Guo
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Zhidong Lu
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Kening Sun
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Qunhua Jin
- Department of Orthopedics, General Hospital of Ningxia Medical University, Yinchuan, 750004, Ningxia, China.
| |
Collapse
|
48
|
Muneer A, Shamsher Khan RM. Endoplasmic Reticulum Stress: Implications for Neuropsychiatric Disorders. Chonnam Med J 2019; 55:8-19. [PMID: 30740335 PMCID: PMC6351318 DOI: 10.4068/cmj.2019.55.1.8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 10/11/2018] [Accepted: 11/09/2018] [Indexed: 11/06/2022] Open
Abstract
The Endoplasmic reticulum (ER), an indispensable sub-cellular component of the eukaryotic cell carries out essential functions, is critical to the survival of the organism. The chaperone proteins and the folding enzymes which are multi-domain ER effectors carry out 3-dimensional conformation of nascent polypeptides and check misfolded protein aggregation, easing the exit of functional proteins from the ER. Diverse conditions, for instance redox imbalance, alterations in ionic calcium levels, and inflammatory signaling can perturb the functioning of the ER, leading to a build-up of unfolded or misfolded proteins in the lumen. This results in ER stress, and aiming to reinstate protein homeostasis, a well conserved reaction called the unfolded protein response (UPR) is elicited. Equally, in protracted cellular stress or inadequate compensatory reaction, UPR pathway leads to cell loss. Dysfunctional ER mechanisms are responsible for neuronal degeneration in numerous human diseases, for instance Alzheimer's, Parkinson's and Huntington's diseases. In addition, mounting proof indicates that ER stress is incriminated in psychiatric diseases like major depressive disorder, bipolar disorder, and schizophrenia. Accumulating evidence suggests that pharmacological agents regulating the working of ER may have a role in diminishing advancing neuronal dysfunction in neuropsychiatric disorders. Here, new findings are examined which link the foremost mechanisms connecting ER stress and cell homeostasis. Furthermore, a supposed new pathogenic model of major neuropsychiatry disorders is provided, with ER stress proposed as the pivotal step in disease development.
Collapse
Affiliation(s)
- Ather Muneer
- Islamic International Medical College, Riphah International University, Rawalpindi, Pakistan
| | | |
Collapse
|
49
|
Unlu I, Lu Y, Wang X. The cyclic phosphodiesterase CNP and RNA cyclase RtcA fine-tune noncanonical XBP1 splicing during ER stress. J Biol Chem 2018; 293:19365-19376. [PMID: 30355738 PMCID: PMC6302167 DOI: 10.1074/jbc.ra118.004872] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/12/2018] [Indexed: 12/21/2022] Open
Abstract
The activity of X box-binding protein 1 (XBP1), a master transcriptional regulator of endoplasmic reticulum (ER) homeostasis and the unfolded protein response (UPR), is controlled by a two-step noncanonical splicing reaction in the cytoplasm. The first step of nuclease cleavage by inositol-requiring enzyme 1 (IRE1), a protein kinase/endoribonuclease, is conserved in all eukaryotic cells. The second step of RNA ligation differs biochemically among species. In yeast, tRNA ligase 1 (Trl1) and tRNA 2'-phosphotransferase 1 (Tpt1) act through a 5'-PO4/3'-OH pathway. In metazoans, RNA 2',3'-cyclic phosphate and 5'-OH ligase (RtcB) ligate XBP1 exons via a 3'-PO4/5'-OH reaction. Although RtcB has been identified as the primary RNA ligase, evidence suggests that yeast-like ligase components may also operate in mammals. In this study, using mouse and human cell lines along with in vitro splicing assays, we investigated whether these components contribute to XBP1 splicing during ER stress. We found that the mammalian 2'-phosphotransferase Trpt1 does not contribute to XBP1 splicing even in the absence of RtcB. Instead, we found that 2',3'-cyclic nucleotide phosphodiesterase (CNP) suppresses RtcB-mediated XBP1 splicing by hydrolyzing 2',3'-cyclic phosphate into 2'-phosphate on the cleaved exon termini. By contrast, RNA 3'-terminal cyclase (RtcA), which converts 2'-phosphate back to 2',3'-cyclic phosphate, facilitated XBP1 splicing by increasing the number of compatible RNA termini for RtcB. Taken together, our results provide evidence that CNP and RtcA fine-tune XBP1 output during ER stress.
Collapse
Affiliation(s)
- Irem Unlu
- From the Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208
| | - Yanyan Lu
- From the Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208
| | - Xiaozhong Wang
- From the Department of Molecular Biosciences, Northwestern University, Evanston, Illinois 60208
| |
Collapse
|
50
|
Jin Y, Sumsuzzman DM, Choi J, Kang H, Lee SR, Hong Y. Molecular and Functional Interaction of the Myokine Irisin with Physical Exercise and Alzheimer's Disease. Molecules 2018; 23:molecules23123229. [PMID: 30544500 PMCID: PMC6321132 DOI: 10.3390/molecules23123229] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 12/03/2018] [Accepted: 12/05/2018] [Indexed: 12/19/2022] Open
Abstract
Irisin, a skeletal muscle-secreted myokine, produced in response to physical exercise, has protective functions in both the central and the peripheral nervous systems, including the regulation of brain-derived neurotrophic factors. In particular, irisin is capable of protecting hippocampus. Since this area is the region of the brain that is most susceptible to Alzheimer's disease (AD), such beneficial effect may inhibit or delay the emergence of neurodegenerative diseases, including AD. Also, the factors engaged in irisin formation appear to suppress Aβ aggregation, which is the pathological hallmark of AD. This review is based on the hypothesis that irisin produced by physical exercise helps to control AD progression. Herein, we describe the physiology of irisin and its potential role in delaying or preventing AD progression in human.
Collapse
Affiliation(s)
- Yunho Jin
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae 50834, Korea.
- Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Korea.
- Ubiquitous Healthcare & Anti-Aging Research Center (u-HARC), Inje University, Gimhae 50834, Korea.
| | - Dewan Md Sumsuzzman
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae 50834, Korea.
- Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Korea.
- Ubiquitous Healthcare & Anti-Aging Research Center (u-HARC), Inje University, Gimhae 50834, Korea.
| | - Jeonghyun Choi
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae 50834, Korea.
- Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Korea.
- Ubiquitous Healthcare & Anti-Aging Research Center (u-HARC), Inje University, Gimhae 50834, Korea.
| | - Hyunbon Kang
- Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Korea.
- Ubiquitous Healthcare & Anti-Aging Research Center (u-HARC), Inje University, Gimhae 50834, Korea.
- Department of Physical Therapy, Graduate School of Inje University, Gimhae 50834, Korea.
| | - Sang-Rae Lee
- National Primate Research Center (NPRC), Korea Research Institute of Bioscience and Biotechnology (KRIBB), Ochang 50834, Korea.
| | - Yonggeun Hong
- Department of Rehabilitation Science, Graduate School of Inje University, Gimhae 50834, Korea.
- Biohealth Products Research Center (BPRC), Inje University, Gimhae 50834, Korea.
- Ubiquitous Healthcare & Anti-Aging Research Center (u-HARC), Inje University, Gimhae 50834, Korea.
- Department of Physical Therapy, Graduate School of Inje University, Gimhae 50834, Korea.
- Department of Physical Therapy, College of Healthcare Medical Science & Engineering, Gimhae 50834, Korea.
| |
Collapse
|