1
|
Hahn KR, Kwon HJ, Kim DW, Hwang IK, Yoon YS. Therapeutic Options of Crystallin Mu and Protein Disulfide Isomerase A3 for Cuprizone-Induced Demyelination in Mouse Hippocampus. Neurochem Res 2024; 49:3078-3093. [PMID: 39164609 PMCID: PMC11449959 DOI: 10.1007/s11064-024-04227-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 08/06/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024]
Abstract
This study investigates the changes in hippocampal proteomic profiles during demyelination and remyelination using the cuprizone model. Employing two-dimensional gel electrophoresis and liquid chromatography-tandem mass spectrometry for protein profiling, we observed significant alterations in the expression of ketimine reductase mu-crystallin (CRYM) and protein disulfide isomerase A3 precursor (PDIA3) following exposure to and subsequent withdrawal from cuprizone. Immunohistochemical staining validated these protein expression patterns in the hippocampus, revealing that both PDIA3 and CRYM were downregulated in the hippocampal CA1 region during demyelination and upregulated during remyelination. Additionally, we explored the potential protective effects of CRYM and PDIA3 against cuprizone-induced demyelination by synthesizing cell-permeable Tat peptide-fusion proteins (Tat-CRYM and Tat-PDIA3) to facilitate their crossing through the blood-brain barrier. Our results indicated that administering Tat-CRYM and Tat-PDIA3 mitigated the reduction in proliferating cell and differentiated neuroblast counts compared to the group receiving cuprizone alone. Notably, Tat-PDIA3 demonstrated significant effects in enhancing myelin basic protein expression alongside phosphorylation of CREB in the hippocampus, suggesting its potential therapeutic role in the prevention or treatment of demyelination, and by extension, in conditions such as multiple sclerosis.
Collapse
Affiliation(s)
- Kyu Ri Hahn
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Hyun Jung Kwon
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
- Department of Biomedical Sciences, and Research Institute for Bioscience and Biotechnology, Hallym University, Chuncheon, 24252, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Gangneung-Wonju National University, Gangneung, 25457, South Korea
| | - In Koo Hwang
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell Biology, College of Veterinary Medicine, and Research Institute for Veterinary Science, Seoul National University, Seoul, 08826, South Korea.
| |
Collapse
|
2
|
Zhu YY, Zhang Q, Jia YC, Hou MJ, Zhu BT. Protein disulfide isomerase plays a crucial role in mediating chemically-induced, glutathione depletion-associated hepatocyte injury in vitro and in vivo. Cell Commun Signal 2024; 22:431. [PMID: 39243059 PMCID: PMC11378433 DOI: 10.1186/s12964-024-01798-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 08/18/2024] [Indexed: 09/09/2024] Open
Abstract
Recently we have shown that protein disulfide isomerase (PDI or PDIA1) is involved in mediating chemically-induced, glutathione (GSH) depletion-associated ferroptotic cell death through NOS activation (dimerization) and NO accumulation. The present study aims to determine the role of PDI in mediating chemically-induced hepatocyte injury in vitro and in vivo and whether PDI inhibitors can effectively protect against chemically-induced hepatocyte injury. We show that during the development of erastin-induced ferroptotic cell death, accumulation of cellular NO, ROS and lipid-ROS follows a sequential order, i.e., cellular NO accumulation first, followed by accumulation of cellular ROS, and lastly cellular lipid-ROS. Cellular NO, ROS and lipid-ROS each play a crucial role in mediating erastin-induced ferroptosis in cultured hepatocytes. In addition, it is shown that PDI is an important upstream mediator of erastin-induced ferroptosis through PDI-mediated conversion of NOS monomer to its dimer, which then leads to accumulation of cellular NO, ROS and lipid-ROS, and ultimately ferroptotic cell death. Genetic manipulation of PDI expression or pharmacological inhibition of PDI function each can effectively abrogate erastin-induced ferroptosis. Lastly, evidence is presented to show that PDI is also involved in mediating acetaminophen-induced liver injury in vivo using both wild-type C57BL/6J mice and hepatocyte-specific PDI conditional knockout (PDIfl/fl Alb-cre) mice. Together, our work demonstrates that PDI is an important upstream mediator of chemically-induced, GSH depletion-associated hepatocyte ferroptosis, and inhibition of PDI can effectively prevent this injury.
Collapse
Affiliation(s)
- Yan-Yin Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Blvd., Longgang District, Shenzhen, 518172, Guangdong, China
| | - Qi Zhang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Blvd., Longgang District, Shenzhen, 518172, Guangdong, China
| | - Yi-Chen Jia
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Blvd., Longgang District, Shenzhen, 518172, Guangdong, China
| | - Ming-Jie Hou
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Blvd., Longgang District, Shenzhen, 518172, Guangdong, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, 2001 Longxiang Blvd., Longgang District, Shenzhen, 518172, Guangdong, China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
3
|
Pirone A, Ciregia F, Lazzarini G, Miragliotta V, Ronci M, Zuccarini M, Zallocco L, Beghelli D, Mazzoni MR, Lucacchini A, Giusti L. Proteomic Profiling Reveals Specific Molecular Hallmarks of the Pig Claustrum. Mol Neurobiol 2023; 60:4336-4358. [PMID: 37095366 PMCID: PMC10293365 DOI: 10.1007/s12035-023-03347-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 04/13/2023] [Indexed: 04/26/2023]
Abstract
The present study, employing a comparative proteomic approach, analyzes the protein profile of pig claustrum (CLA), putamen (PU), and insula (IN). Pig brain is an interesting model whose key translational features are its similarities with cortical and subcortical structures of human brain. A greater difference in protein spot expression was observed in CLA vs PU as compared to CLA vs IN. The deregulated proteins identified in CLA resulted to be deeply implicated in neurodegenerative (i.e., sirtuin 2, protein disulfide-isomerase 3, transketolase) and psychiatric (i.e., copine 3 and myelin basic protein) disorders in humans. Metascape analysis of differentially expressed proteins in CLA vs PU comparison suggested activation of the α-synuclein pathway and L1 recycling pathway corroborating the involvement of these anatomical structures in neurodegenerative diseases. The expression of calcium/calmodulin-dependent protein kinase and dihydropyrimidinase like 2, which are linked to these pathways, was validated using western blot analysis. Moreover, the protein data set of CLA vs PU comparison was analyzed by Ingenuity Pathways Analysis to obtain a prediction of most significant canonical pathways, upstream regulators, human diseases, and biological functions. Interestingly, inhibition of presenilin 1 (PSEN1) upstream regulator and activation of endocannabinoid neuronal synapse pathway were observed. In conclusion, this is the first study presenting an extensive proteomic analysis of pig CLA in comparison with adjacent areas, IN and PUT. These results reinforce the common origin of CLA and IN and suggest an interesting involvement of CLA in endocannabinoid circuitry, neurodegenerative, and psychiatric disorders in humans.
Collapse
Affiliation(s)
- Andrea Pirone
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy.
| | - Federica Ciregia
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Giulia Lazzarini
- Department of Veterinary Sciences, University of Pisa, Pisa, Italy
| | | | - Maurizio Ronci
- Department of Medical, Oral and Biotechnological Sciences, University G. D'Annunzio of Chieti-Pescara, Chieti, Italy
- Interuniversitary Consortium for Engineering and Medicine, COIIM, Campobasso, Italy
| | - Mariachiara Zuccarini
- Department of Medical, Oral and Biotechnological Sciences, University G. D'Annunzio of Chieti-Pescara, Chieti, Italy
| | - Lorenzo Zallocco
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Daniela Beghelli
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | | | - Antonio Lucacchini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Laura Giusti
- School of Pharmacy, University of Camerino, Camerino, Italy
| |
Collapse
|
4
|
Kartik S, Pal R, Chaudhary MJ, Nath R, Kumar M, Binwal M, Bawankule DU. Neuroprotective role of chloroquine via modulation of autophagy and neuroinflammation in MPTP-induced Parkinson's disease. Inflammopharmacology 2023; 31:927-941. [PMID: 36715843 DOI: 10.1007/s10787-023-01141-z] [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: 07/14/2022] [Accepted: 01/19/2023] [Indexed: 01/31/2023]
Abstract
Parkinson's disease (PD) is a neuro-motor ailment that strikes adults in their older life and results in both motor and non-motor impairments. In neuronal and glial cells, PD has recently been linked to a dysregulated autophagic system and cerebral inflammation. Chloroquine (CQ), an anti-malarial drug, has been demonstrated to suppress autophagy in a variety of diseases, including cerebral ischemia, Alzheimer's disease (AD), and Traumatic brain injury (TBI), while its involvement in PD is still unclear. BALB/c mice were randomly allocated to one of four groups: 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP), CQ treatment with or without MPTP, or control. The CQ treatment group received CQ (intraperitoneally, 8 mg/kg body weight) after 1 h of MPTP induction on day 1, and it lasted for 7 days. CQ therapy preserves dopamine levels stable, inhibits tyrosine hydroxylase (TH) positive dopaminergic cell death, and lowers oxidative stress. CQ reduces the behavioural, motor, and cognitive deficits caused by MPTP after injury. Furthermore, CQ therapy slowed aberrant neuronal autophagy (microtubule-associated protein-1 light chain 3B; LC3B & Beclin1) and lowered expression levels of the inflammatory cytokines interleukin 1 (IL-1β) and tumour necrosis factor (TNF-α) in the mice brain. In addition, CQ's antioxidant and anti-inflammatory effects were also tested in MPTP-mediated cell death in PC12 cells, demonstrating that CQ has a neurorestorative impact by successfully rescuing MPTP-induced ROS generation and cell loss. Our findings show that CQ's can help to prevent dopaminergic degeneration and improve neurological function after MPTP intoxication by lowering the harmful effects of neuronal autophagy and cerebral inflammation.
Collapse
Affiliation(s)
- Shipra Kartik
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, UP, 226003, India
| | - Rishi Pal
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, UP, 226003, India.
| | - Manju J Chaudhary
- Department of Physiology, Government Medical College, Tirwa Road, Kannauj, UP, India
| | - Rajendra Nath
- Department of Pharmacology and Therapeutics, King George's Medical University, Lucknow, UP, 226003, India
| | - Madhu Kumar
- Department of Pathology, King George's Medical University, Lucknow, UP, 226003, India
| | - Monika Binwal
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, UP, 226015, India
| | - D U Bawankule
- Bioprospection and Product Development Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, UP, 226015, India
| |
Collapse
|
5
|
Sulimai NH, Brown J, Lominadze D. Fibrinogen, Fibrinogen-like 1 and Fibrinogen-like 2 Proteins, and Their Effects. Biomedicines 2022; 10:1712. [PMID: 35885017 PMCID: PMC9313381 DOI: 10.3390/biomedicines10071712] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/11/2022] [Accepted: 07/13/2022] [Indexed: 12/05/2022] Open
Abstract
Fibrinogen (Fg) and its derivatives play a considerable role in many diseases. For example, increased levels of Fg have been found in many inflammatory diseases, such as Alzheimer's disease, multiple sclerosis, traumatic brain injury, rheumatoid arthritis, systemic lupus erythematosus, and cancer. Although associations of Fg, Fg chains, and its derivatives with various diseases have been established, their specific effects and the mechanisms of actions involved are still unclear. The present review is the first attempt to discuss the role of Fg, Fg chains, its derivatives, and other members of Fg family proteins, such as Fg-like protein 1 and 2, in inflammatory diseases and their effects in immunomodulation.
Collapse
Affiliation(s)
- Nurul H. Sulimai
- Departments of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (N.H.S.); (J.B.)
| | - Jason Brown
- Departments of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (N.H.S.); (J.B.)
| | - David Lominadze
- Departments of Surgery, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA; (N.H.S.); (J.B.)
- Departments of Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL 33612, USA
| |
Collapse
|
6
|
Parakh S, Perri ER, Vidal M, Sultana J, Shadfar S, Mehta P, Konopka A, Thomas CJ, Spencer DM, Atkin JD. Protein disulphide isomerase (PDI) is protective against amyotrophic lateral sclerosis (ALS)-related mutant Fused in Sarcoma (FUS) in in vitro models. Sci Rep 2021; 11:17557. [PMID: 34475430 PMCID: PMC8413276 DOI: 10.1038/s41598-021-96181-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 07/26/2021] [Indexed: 12/04/2022] Open
Abstract
Mutations in Fused in Sarcoma (FUS) are present in familial and sporadic cases of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). FUS is localised in the nucleus where it has important functions in DNA repair. However, in ALS/FTD, mutant FUS mislocalises from the nucleus to the cytoplasm where it forms inclusions, a key pathological hallmark of neurodegeneration. Mutant FUS also inhibits protein import into the nucleus, resulting in defects in nucleocytoplasmic transport. Fragmentation of the neuronal Golgi apparatus, induction of endoplasmic reticulum (ER) stress, and inhibition of ER-Golgi trafficking are also associated with mutant FUS misfolding in ALS. Protein disulphide isomerase (PDI) is an ER chaperone previously shown to be protective against misfolding associated with mutant superoxide dismutase 1 (SOD1) and TAR DNA-binding protein-43 (TDP-43) in cellular and zebrafish models. However, a protective role against mutant FUS in ALS has not been previously described. In this study, we demonstrate that PDI is protective against mutant FUS. In neuronal cell line and primary cultures, PDI restores defects in nuclear import, prevents the formation of mutant FUS inclusions, inhibits Golgi fragmentation, ER stress, ER-Golgi transport defects, and apoptosis. These findings imply that PDI is a new therapeutic target in FUS-associated ALS.
Collapse
Affiliation(s)
- S Parakh
- Macquarie Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - E R Perri
- Macquarie Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - M Vidal
- Macquarie Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - J Sultana
- Macquarie Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - S Shadfar
- Macquarie Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - P Mehta
- Macquarie Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - A Konopka
- Macquarie Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| | - C J Thomas
- Department of Physiology, Anatomy and Microbiology, La Trobe University, Melbourne, VIC, 3086, Australia
| | - D M Spencer
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia
| | - J D Atkin
- Macquarie Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, 2109, Australia. .,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, 3086, Australia.
| |
Collapse
|
7
|
Momenzadeh S, Zamani S, Dehghan F, Barreiro C, Jami MS. Comparative proteome analyses highlight several exercise-like responses of mouse sciatic nerve after IP injection of irisin. Eur J Neurosci 2021; 53:3263-3278. [PMID: 33759230 DOI: 10.1111/ejn.15202] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 02/19/2021] [Accepted: 03/12/2021] [Indexed: 12/16/2022]
Abstract
Many beneficial effects of exercise on the nervous system are mediated by hormone (growth factor)/receptor signaling. Considering the accumulating evidence on the similarity of some beneficial effects, irisin can be a proposed effector of exercise; however, the mechanism underlying these effects remains largely unknown. More evidence on the mechanism of action might reveal its potential as a treatment strategy to substitute exercise recovery protocols for nerve injuries in physically disabled patients. To evaluate the underlying mechanism of irisin involvement in nerve adaptation and exerting beneficial effects, we studied the proteome profile alteration of mouse sciatic nerve after irisin administration. We also compared it with two 8-week protocols of resistance exercise and endurance exercise. The results indicate that irisin contributes to the regulation of nerve metabolism via overexpression of Ckm and ATP5j2 proteins. Irisin administration may improve sciatic nerve function by maintaining the architecture, enhancing axonal transport, and promoting synapse plasticity through increased structural and regulatory proteins and NO production. We also showed that irisin has the potential to induce neurotrophic support on the sciatic nerve by maintaining cell redox homeostasis, and responses to oxidative stress via the upregulation of disulfide-isomerase and superoxide dismutase enzymes. Comparing with exercise groups, these effects are somewhat exercise-like responses. These data suggest that irisin can be a promising therapeutic candidate for specific targeting of defects in peripheral neuropathies and nerve injuries as an alternative for physical therapy.
Collapse
Affiliation(s)
- Sedigheh Momenzadeh
- Department of Medical Biotechnology, School of Advanced Technologies, Shahrekord University of Medical Sciences, Shahrekord, Iran.,Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Saeed Zamani
- Department of Anatomical Sciences, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fariba Dehghan
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Carlos Barreiro
- 5 INBIOTEC (Instituto de Biotecnología de León), León, Spain.,Departamento de Biología Molecular, Universidad de León, Ponferrada, Spain
| | - Mohammad-Saeid Jami
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.,Department of Neurology, David Geffen School of Medicine, University of California Los Angeles (UCLA), Los Angeles, CA, USA
| |
Collapse
|
8
|
Jagaraj CJ, Parakh S, Atkin JD. Emerging Evidence Highlighting the Importance of Redox Dysregulation in the Pathogenesis of Amyotrophic Lateral Sclerosis (ALS). Front Cell Neurosci 2021; 14:581950. [PMID: 33679322 PMCID: PMC7929997 DOI: 10.3389/fncel.2020.581950] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 12/21/2020] [Indexed: 12/20/2022] Open
Abstract
The cellular redox state, or balance between cellular oxidation and reduction reactions, serves as a vital antioxidant defence system that is linked to all important cellular activities. Redox regulation is therefore a fundamental cellular process for aerobic organisms. Whilst oxidative stress is well described in neurodegenerative disorders including amyotrophic lateral sclerosis (ALS), other aspects of redox dysfunction and their contributions to pathophysiology are only just emerging. ALS is a fatal neurodegenerative disease affecting motor neurons, with few useful treatments. Hence there is an urgent need to develop more effective therapeutics in the future. Here, we discuss the increasing evidence for redox dysregulation as an important and primary contributor to ALS pathogenesis, which is associated with multiple disease mechanisms. Understanding the connection between redox homeostasis, proteins that mediate redox regulation, and disease pathophysiology in ALS, may facilitate a better understanding of disease mechanisms, and lead to the design of better therapeutic strategies.
Collapse
Affiliation(s)
- Cyril Jones Jagaraj
- Department of Biomedical Sciences, Macquarie University Centre for MND Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Sonam Parakh
- Department of Biomedical Sciences, Macquarie University Centre for MND Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia
| | - Julie D Atkin
- Department of Biomedical Sciences, Macquarie University Centre for MND Research, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW, Australia.,Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, Australia
| |
Collapse
|
9
|
Rozas P, Pinto C, Martínez Traub F, Díaz R, Pérez V, Becerra D, Ojeda P, Ojeda J, Wright MT, Mella J, Plate L, Henríquez JP, Hetz C, Medinas DB. Protein disulfide isomerase ERp57 protects early muscle denervation in experimental ALS. Acta Neuropathol Commun 2021; 9:21. [PMID: 33541434 PMCID: PMC7863244 DOI: 10.1186/s40478-020-01116-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a progressive fatal neurodegenerative disease that affects motoneurons. Mutations in superoxide dismutase 1 (SOD1) have been described as a causative genetic factor for ALS. Mice overexpressing ALS-linked mutant SOD1 develop ALS symptoms accompanied by histopathological alterations and protein aggregation. The protein disulfide isomerase family member ERp57 is one of the main up-regulated proteins in tissue of ALS patients and mutant SOD1 mice, whereas point mutations in ERp57 were described as possible risk factors to develop the disease. ERp57 catalyzes disulfide bond formation and isomerization in the endoplasmic reticulum (ER), constituting a central component of protein quality control mechanisms. However, the actual contribution of ERp57 to ALS pathogenesis remained to be defined. Here, we studied the consequences of overexpressing ERp57 in experimental ALS using mutant SOD1 mice. Double transgenic SOD1G93A/ERp57WT animals presented delayed deterioration of electrophysiological activity and maintained muscle innervation compared to single transgenic SOD1G93A littermates at early-symptomatic stage, along with improved motor performance without affecting survival. The overexpression of ERp57 reduced mutant SOD1 aggregation, but only at disease end-stage, dissociating its role as an anti-aggregation factor from the protection of neuromuscular junctions. Instead, proteomic analysis revealed that the neuroprotective effects of ERp57 overexpression correlated with increased levels of synaptic and actin cytoskeleton proteins in the spinal cord. Taken together, our results suggest that ERp57 operates as a disease modifier at early stages by maintaining motoneuron connectivity.
Collapse
Affiliation(s)
- Pablo Rozas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, 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
| | - Cristina Pinto
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Francisca Martínez Traub
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, 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
| | - Rodrigo Díaz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, 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
| | - Viviana Pérez
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Daniela Becerra
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, 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
| | - Patricia Ojeda
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, 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
| | - Jorge Ojeda
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Madison T Wright
- Department of Chemistry and Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Jessica Mella
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Lars Plate
- Department of Chemistry and Department of Biological Sciences, Vanderbilt University, Nashville, TN, USA
| | - Juan Pablo Henríquez
- Neuromuscular Studies Laboratory (NeSt Lab), Department of Cell Biology, Faculty of Biological Sciences, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, 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.
| | - Danilo B Medinas
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Independencia 1027, P.O. Box 70086, 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.
| |
Collapse
|
10
|
Shin YH, Cho H, Choi BY, Kim J, Ha J, Suh SW, Park SB. Phenotypic Discovery of Neuroprotective Agents by Regulation of Tau Proteostasis via Stress-Responsive Activation of PERK Signaling. Angew Chem Int Ed Engl 2021; 60:1831-1838. [PMID: 33210431 PMCID: PMC7898623 DOI: 10.1002/anie.202013915] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Indexed: 02/06/2023]
Abstract
Tau protein aggregates are a recognized neuropathological feature in Alzheimer's disease as well as many other neurodegenerative disorders, known as tauopathies. The development of tau-targeting therapies is therefore extremely important but efficient strategies or protein targets are still unclear. Here, we performed a cell-based phenotypic screening under endoplasmic reticulum (ER) stress conditions and identified a small molecule, SB1617, capable of suppressing abnormal tau protein aggregation. By applying label-free target identification technology, we revealed that the transient enhancement of protein kinase-like endoplasmic reticulum kinase (PERK) signaling pathway through the inhibition of stress-responsive SB1617 targets, PDIA3 and DNAJC3, is an effective strategy for regulating proteostasis in tauopathies. The molecular mechanism and the promising efficacy of SB1617 were demonstrated in neuronal cells and a mouse model with traumatic brain injury, a tauopathy known to involve ER stress.
Collapse
Affiliation(s)
- Young-Hee Shin
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826, Korea
| | - Hana Cho
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826, Korea
| | - Bo Young Choi
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, 24252, Korea
| | - Jonghoon Kim
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826, Korea.,Present address: Department of Chemistry, Soongsil University, Seoul, 06978, Korea
| | - Jaeyoung Ha
- Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826, Korea
| | - Sang Won Suh
- Department of Physiology, College of Medicine, Hallym University, Chuncheon, 24252, Korea
| | - Seung Bum Park
- CRI Center for Chemical Proteomics, Department of Chemistry, Seoul National University, Seoul, 08826, Korea.,Department of Biophysics and Chemical Biology, Seoul National University, Seoul, 08826, Korea
| |
Collapse
|
11
|
Shin Y, Cho H, Choi BY, Kim J, Ha J, Suh SW, Park SB. Phenotypic Discovery of Neuroprotective Agents by Regulation of Tau Proteostasis via Stress‐Responsive Activation of PERK Signaling. Angew Chem Int Ed Engl 2020. [DOI: 10.1002/ange.202013915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Young‐Hee Shin
- CRI Center for Chemical Proteomics Department of Chemistry Seoul National University Seoul 08826 Korea
| | - Hana Cho
- Department of Biophysics and Chemical Biology Seoul National University Seoul 08826 Korea
| | - Bo Young Choi
- Department of Physiology College of Medicine Hallym University Chuncheon 24252 Korea
| | - Jonghoon Kim
- CRI Center for Chemical Proteomics Department of Chemistry Seoul National University Seoul 08826 Korea
- Present address: Department of Chemistry Soongsil University Seoul 06978 Korea
| | - Jaeyoung Ha
- Department of Biophysics and Chemical Biology Seoul National University Seoul 08826 Korea
| | - Sang Won Suh
- Department of Physiology College of Medicine Hallym University Chuncheon 24252 Korea
| | - Seung Bum Park
- CRI Center for Chemical Proteomics Department of Chemistry Seoul National University Seoul 08826 Korea
- Department of Biophysics and Chemical Biology Seoul National University Seoul 08826 Korea
| |
Collapse
|
12
|
The transcriptome of anterior regeneration in earthworm Eudrilus eugeniae. Mol Biol Rep 2020; 48:259-283. [PMID: 33306150 DOI: 10.1007/s11033-020-06044-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/28/2020] [Indexed: 12/25/2022]
Abstract
The oligochaete earthworm, Eudrilus eugeniae is capable of regenerating both anterior and posterior segments. The present study focuses on the transcriptome analysis of earthworm E. eugeniae to identify and functionally annotate the key genes supporting the anterior blastema formation and regulating the anterior regeneration of the worm. The Illumina sequencing generated a total of 91,593,182 raw reads which were assembled into 105,193 contigs using CLC genomics workbench. In total, 40,946 contigs were annotated against the NCBI nr and SwissProt database and among them, 15,702 contigs were assigned to 14,575 GO terms. Besides a total of 9389 contigs were mapped to 416 KEGG biological pathways. The RNA-Seq comparison study identified 10,868 differentially expressed genes (DEGs) and of them, 3986 genes were significantly upregulated in the anterior regenerated blastema tissue samples of the worm. The GO enrichment analysis showed angiogenesis and unfolded protein binding as the top enriched functions and the pathway enrichment analysis denoted TCA cycle as the most significantly enriched pathway associated with the upregulated gene dataset of the worm. The identified DEGs and their function and pathway information can be effectively utilized further to interpret the key cellular, genetic and molecular events associated with the regeneration of the worm.
Collapse
|
13
|
Hasmatali JCD, De Guzman J, Zhai R, Yang L, McLean NA, Hutchinson C, Johnston JM, Misra V, Verge VMK. Axotomy Induces Phasic Alterations in Luman/CREB3 Expression and Nuclear Localization in Injured and Contralateral Uninjured Sensory Neurons: Correlation With Intrinsic Axon Growth Capacity. J Neuropathol Exp Neurol 2020; 78:348-364. [PMID: 30863858 DOI: 10.1093/jnen/nlz008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Luman/CREB3 is an important early retrograde axotomy signal regulating acute axon outgrowth in sensory neurons through the adaptive unfolded protein response. As the injury response is transcriptionally multiphasic, a spatiotemporal analysis of Luman/CREB3 localization in rat dorsal root ganglion (DRG) with unilateral L4-L6 spinal nerve injury was conducted to determine if Luman/CREB3 expression was similarly regulated. Biphasic alterations in Luman/CREB3 immunofluorescence and nuclear localization occurred in neurons ipsilateral to 1-hour, 1-day, 2-day, 4-day, and 1-week injury, with a largely parallel, but less avid response contralaterally. This biphasic response was not observed at the transcript level. To assess whether changes in neuronal Luman expression corresponded with an altered intrinsic capacity to grow an axon/neurite in vitro, injury-conditioned and contralateral uninjured DRG neurons underwent a 24-hour axon growth assay. Two-day injury-conditioned neurons exhibited maximal outgrowth capacity relative to naïve, declining at later injury-conditioned timepoints. Only neurons contralateral to 1-week injury exhibited significantly higher axon growth capacity than naïve. In conclusion, alterations in neuronal injury-associated Luman/CREB3 expression support that a multiphasic cell body response occurs and reveal a novel contralateral plasticity in axon growth capacity at 1-week post-injury. These adaptive responses have the potential to inform when repair or therapeutic intervention may be most effective.
Collapse
Affiliation(s)
- Jovan C D Hasmatali
- Department of Anatomy, Physiology and Pharmacology.,Cameco MS Neuroscience Research Center.,Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Department of Critical Care Medicine, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Jolly De Guzman
- Department of Anatomy, Physiology and Pharmacology.,Cameco MS Neuroscience Research Center
| | - Ruiling Zhai
- Department of Anatomy, Physiology and Pharmacology.,Cameco MS Neuroscience Research Center
| | - Lisa Yang
- Cameco MS Neuroscience Research Center
| | - Nikki A McLean
- Department of Anatomy, Physiology and Pharmacology.,Cameco MS Neuroscience Research Center
| | - Catherine Hutchinson
- Department of Anatomy, Physiology and Pharmacology.,Cameco MS Neuroscience Research Center
| | - Jayne M Johnston
- Department of Anatomy, Physiology and Pharmacology.,Cameco MS Neuroscience Research Center
| | - Vikram Misra
- Department of Veterinary Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Valerie M K Verge
- Department of Anatomy, Physiology and Pharmacology.,Cameco MS Neuroscience Research Center
| |
Collapse
|
14
|
Yoo DY, Cho SB, Jung HY, Kim W, Nam SM, Kim JW, Moon SM, Yoon YS, Kim DW, Choi SY, Hwang IK. Differential roles of exogenous protein disulfide isomerase A3 on proliferating cell and neuroblast numbers in the normal and ischemic gerbils. Brain Behav 2020; 10:e01534. [PMID: 31957985 PMCID: PMC7066343 DOI: 10.1002/brb3.1534] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 12/19/2019] [Accepted: 12/23/2019] [Indexed: 01/02/2023] Open
Abstract
INTRODUCTION We examined the effects of exogenous protein disulfide isomerase A3 (PDIA3) on hippocampal neurogenesis in gerbils under control and ischemic damage. METHODS To facilitate the delivery of PDIA3 to the brain, we constructed Tat-PDIA3 protein and administered vehicle (10% glycerol) or Tat-PDIA3 protein once a day for 28 days. On day 24 of vehicle or Tat-PDIA3 treatment, ischemia was transiently induced by occlusion of both common carotid arteries for 5 min. RESULTS Administration of Tat-PDIA3 significantly reduced ischemia-induced spontaneous motor activity, and the number of NeuN-positive nuclei in the Tat-PDIA3-treated ischemic group was significantly increased in the CA1 region compared to that in the vehicle-treated ischemic group. Ki67- and DCX-immunoreactive cells were significantly higher in the Tat-PDIA3-treated group compared to the vehicle-treated control group. In vehicle- and Tat-PDIA3-treated ischemic groups, the number of Ki67- and DCX-immunoreactive cells was significantly higher as compared to those in the vehicle- and Tat-PDIA3-treated control groups, respectively. In the dentate gyrus, the numbers of Ki67-immunoreactive cells were comparable between vehicle- and Tat-PDIA3-treated ischemic groups, while more DCX-immunoreactive cells were observed in the Tat-PDIA3-treated group. Transient forebrain ischemia increased the expression of phosphorylated cAMP-response element-binding protein (pCREB) in the dentate gyrus, but the administration of Tat-PDIA3 robustly increased pCREB-positive nuclei in the normal gerbils, but not in the ischemic gerbils. Brain-derived neurotrophic factor (BDNF) mRNA expression was significantly increased in the Tat-PDIA3-treated group compared to that in the vehicle-treated group. Transient forebrain ischemic increased BDNF mRNA levels in both vehicle- and Tat-PDIA3-treated groups, and there were no significant differences between groups. CONCLUSIONS These results suggest that Tat-PDIA3 enhances cell proliferation and neuroblast numbers in the dentate gyrus in normal, but not in ischemic gerbils, by increasing BDNF mRNA and phosphorylation of pCREB.
Collapse
Affiliation(s)
- Dae Young Yoo
- Department of Anatomy and Cell BiologyCollege of Veterinary Medicine, and Research Institute for Veterinary ScienceSeoul National UniversitySeoulSouth Korea
- Department of AnatomyCollege of MedicineSoonchunhyang UniversityCheonanSouth Korea
| | - Su Bin Cho
- Department of Biomedical Sciences, and Research Institute for Bioscience and BiotechnologyHallym UniversityChuncheonSouth Korea
| | - Hyo Young Jung
- Department of Anatomy and Cell BiologyCollege of Veterinary Medicine, and Research Institute for Veterinary ScienceSeoul National UniversitySeoulSouth Korea
| | - Woosuk Kim
- Department of Anatomy and Cell BiologyCollege of Veterinary Medicine, and Research Institute for Veterinary ScienceSeoul National UniversitySeoulSouth Korea
| | - Sung Min Nam
- Department of AnatomyCollege of Veterinary MedicineKonkuk UniversitySeoulSouth Korea
| | - Jong Whi Kim
- Department of Anatomy and Cell BiologyCollege of Veterinary Medicine, and Research Institute for Veterinary ScienceSeoul National UniversitySeoulSouth Korea
| | - Seung Myung Moon
- Department of NeurosurgeryDongtan Sacred Heart HospitalCollege of MedicineHallym UniversityHwaseongSouth Korea
- Research Institute for Complementary & Alternative MedicineHallym UniversityChuncheonSouth Korea
| | - Yeo Sung Yoon
- Department of Anatomy and Cell BiologyCollege of Veterinary Medicine, and Research Institute for Veterinary ScienceSeoul National UniversitySeoulSouth Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular BiologyResearch Institute of Oral SciencesCollege of DentistryGangneung‐Wonju National UniversityGangneungSouth Korea
| | - Soo Young Choi
- Department of Biomedical Sciences, and Research Institute for Bioscience and BiotechnologyHallym UniversityChuncheonSouth Korea
| | - In Koo Hwang
- Department of Anatomy and Cell BiologyCollege of Veterinary Medicine, and Research Institute for Veterinary ScienceSeoul National UniversitySeoulSouth Korea
| |
Collapse
|
15
|
Yousuf MS, Maguire AD, Simmen T, Kerr BJ. Endoplasmic reticulum-mitochondria interplay in chronic pain: The calcium connection. Mol Pain 2020; 16:1744806920946889. [PMID: 32787562 PMCID: PMC7427143 DOI: 10.1177/1744806920946889] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/26/2020] [Indexed: 12/14/2022] Open
Abstract
Chronic pain is a debilitating condition that affects roughly a third to a half of the world's population. Despite its substantial effect on society, treatment for chronic pain is modest, at best, notwithstanding its side effects. Hence, novel therapeutics are direly needed. Emerging evidence suggests that calcium plays an integral role in mediating neuronal plasticity that underlies sensitization observed in chronic pain states. The endoplasmic reticulum and the mitochondria are the largest calcium repositories in a cell. Here, we review how stressors, like accumulation of misfolded proteins and oxidative stress, influence endoplasmic reticulum and mitochondria function and contribute to chronic pain. We further examine the shuttling of calcium across the mitochondrial-associated membrane as a mechanism of cross-talk between the endoplasmic reticulum and the mitochondria. In addition, we discuss how endoplasmic reticulum stress, mitochondrial impairment, and calcium dyshomeostasis are implicated in various models of neuropathic pain. We propose a novel framework of endoplasmic reticulum-mitochondria signaling in mediating pain hypersensitivity. These observations require further investigation in order to develop novel therapies for chronic pain.
Collapse
Affiliation(s)
- Muhammad Saad Yousuf
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Aislinn D Maguire
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Thomas Simmen
- Department of Cell Biology, University of Alberta, Edmonton, Canada
| | - Bradley J Kerr
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
- Department of Pharmacology, University of Alberta, Edmonton, Canada
- Department of Anesthesiology and Pain Medicine, University of Alberta, Edmonton, Canada
| |
Collapse
|
16
|
The necroptosis machinery mediates axonal degeneration in a model of Parkinson disease. Cell Death Differ 2019; 27:1169-1185. [PMID: 31591470 DOI: 10.1038/s41418-019-0408-4] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 07/14/2019] [Accepted: 08/06/2019] [Indexed: 11/09/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative condition, characterized by motor impairment due to the progressive degeneration of dopaminergic neurons in the substantia nigra and depletion of dopamine release in the striatum. Accumulating evidence suggest that degeneration of axons is an early event in the disease, involving destruction programs that are independent of the survival of the cell soma. Necroptosis, a programmed cell death process, is emerging as a mediator of neuronal loss in models of neurodegenerative diseases. Here, we demonstrate activation of necroptosis in postmortem brain tissue from PD patients and in a toxin-based mouse model of the disease. Inhibition of key components of the necroptotic pathway resulted in a significant delay of 6-hydroxydopamine-dependent axonal degeneration of dopaminergic and cortical neurons in vitro. Genetic ablation of necroptosis mediators MLKL and RIPK3, as well as pharmacological inhibition of RIPK1 in preclinical models of PD, decreased dopaminergic neuron degeneration, improving motor performance. Together, these findings suggest that axonal degeneration in PD is mediated by the necroptosis machinery, a process here referred to as necroaxoptosis, a druggable pathway to target dopaminergic neuronal loss.
Collapse
|
17
|
Larsson S, Voss U. Neuroprotective effects of vitamin D on high fat diet- and palmitic acid-induced enteric neuronal loss in mice. BMC Gastroenterol 2018; 18:175. [PMID: 30463517 PMCID: PMC6249721 DOI: 10.1186/s12876-018-0905-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 11/08/2018] [Indexed: 01/26/2023] Open
Abstract
Background The role of vitamin D in obesity and diabetes is debated. Obese and/or diabetic patients have elevated levels of free fatty acids, increased susceptibility to gastrointestinal symptoms and are suggested to have altered vitamin D balance. The enteric nervous system is pivotal in regulating gastrointestinal activity and high fat diet (HFD) has been shown to cause loss of enteric neurons in ileum and colon. This study investigates the effect of vitamin D on HFD- and palmitic acid-induced enteric neuronal loss in vivo and in vitro. Methods Mice were fed either a normal diet (ND) or HFD supplemented with varying levels of vitamin D (from 0x to 20x normal vitamin D level) for 19 weeks. Ileum and colon were analyzed for neuronal numbers and remodeling. Primary cultures of myenteric neurons from mouse small intestine were treated with palmitic acid (4x10-4M) and/or 1α,25-hydroxy-vitamin D3 (VD, 10-11- 10-7M) with or without modulators of lipid metabolism and VD pathways. Cultures were analyzed by immunocyto- and histochemical methods. Results Vitamin D supplementation had no effect on enteric neuronal survival in the ND group. HFD caused substantial loss of myenteric neurons in ileum and colon. Vitamin D supplementation between 0-2x normal had no effect on HFD-induced neuronal loss. Supplementation with 20x normal, prevented the HFD-induced neuronal loss. In vitro supplementation of VD prevented the palmitic acid-induced neuronal loss. The VD receptor (VDR) was not identified in enteric neurons. Enteric glia expressed the alternative VD receptor, protein disulphide isomerase family A member 3 (PDIA3), but PDIA3 was not found to mediate the VD response in vitro. Inhibition of peroxisome proliferator-activated receptor gamma (PPARγ) and immune neutralization of isocitrate lyase prevented the VD mediated neuroprotection to palmitic acid exposure. Conclusions Results show that VD protect enteric neurons against HFD and palmitic acid induced neuronal loss. The mechanism behind is suggested to be through activation of PPARγ leading to improved neuronal peroxisome function and metabolism of neuronal lipid intermediates.
Collapse
Affiliation(s)
- Sara Larsson
- Unit of Molecular Endocrinology, Department of Experimental Medical Science, Lund University, Sölvegatan 19, BMC C11, 22184, Lund, Sweden
| | - Ulrikke Voss
- Unit of Neurogastroenterology, Department of Experimental Medical Science, Lund University, Sölvegatan 19, BMC B11, 22184, Lund, Sweden.
| |
Collapse
|
18
|
Thapa S, Abdulrahman B, Abdelaziz DH, Lu L, Ben Aissa M, Schatzl HM. Overexpression of quality control proteins reduces prion conversion in prion-infected cells. J Biol Chem 2018; 293:16069-16082. [PMID: 30154245 PMCID: PMC6187620 DOI: 10.1074/jbc.ra118.002754] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 08/09/2018] [Indexed: 11/06/2022] Open
Abstract
Prion diseases are fatal infectious neurodegenerative disorders in humans and other animals and are caused by misfolding of the cellular prion protein (PrPC) into the pathological isoform PrPSc. These diseases have the potential to transmit within or between species, including zoonotic transmission to humans. Elucidating the molecular and cellular mechanisms underlying prion propagation and transmission is therefore critical for developing molecular strategies for disease intervention. We have shown previously that impaired quality control mechanisms directly influence prion propagation. In this study, we manipulated cellular quality control pathways in vitro by stably and transiently overexpressing selected quality control folding (ERp57) and cargo (VIP36) proteins and investigated the effects of this overexpression on prion propagation. We found that ERp57 or VIP36 overexpression in persistently prion-infected neuroblastoma cells significantly reduces the amount of PrPSc in immunoblots and prion-seeding activity in the real-time quaking-induced conversion (RT-QuIC) assay. Using different cell lines infected with various prion strains confirmed that this effect is not cell type– or prion strain–specific. Moreover, de novo prion infection revealed that the overexpression significantly reduced newly formed PrPSc in acutely infected cells. ERp57-overexpressing cells significantly overcame endoplasmic reticulum stress, as revealed by expression of lower levels of the stress markers BiP and CHOP, accompanied by a decrease in PrP aggregates. Furthermore, application of ERp57-expressing lentiviruses prolonged the survival of prion-infected mice. Taken together, improved cellular quality control via ERp57 or VIP36 overexpression impairs prion propagation and could be utilized as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Simrika Thapa
- From the Calgary Prion Research Unit, University of Calgary, Calgary, Alberta T2N 4Z6, Canada.,the Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta T2N 4Z6, Canada
| | - Basant Abdulrahman
- From the Calgary Prion Research Unit, University of Calgary, Calgary, Alberta T2N 4Z6, Canada.,the Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta T2N 4Z6, Canada.,the Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt, and
| | - Dalia H Abdelaziz
- From the Calgary Prion Research Unit, University of Calgary, Calgary, Alberta T2N 4Z6, Canada.,the Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta T2N 4Z6, Canada.,the Department of Biochemistry and Molecular Biology, Faculty of Pharmacy, Helwan University, 11795 Cairo, Egypt, and
| | - Li Lu
- From the Calgary Prion Research Unit, University of Calgary, Calgary, Alberta T2N 4Z6, Canada.,the Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta T2N 4Z6, Canada
| | - Manel Ben Aissa
- From the Calgary Prion Research Unit, University of Calgary, Calgary, Alberta T2N 4Z6, Canada.,the Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta T2N 4Z6, Canada
| | - Hermann M Schatzl
- From the Calgary Prion Research Unit, University of Calgary, Calgary, Alberta T2N 4Z6, Canada, .,the Department of Comparative Biology and Experimental Medicine, Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta T2N 4Z6, Canada.,the Departments of Veterinary Sciences and of Molecular Biology, University of Wyoming, Laramie, Wyoming 82071
| |
Collapse
|
19
|
Chen X, Bai J, Liu X, Song Z, Zhang Q, Wang X, Jiang P. Nsp1α of Porcine Reproductive and Respiratory Syndrome Virus Strain BB0907 Impairs the Function of Monocyte-Derived Dendritic Cells via the Release of Soluble CD83. J Virol 2018; 92:e00366-18. [PMID: 29793955 PMCID: PMC6052304 DOI: 10.1128/jvi.00366-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/08/2018] [Indexed: 12/21/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV), a virulent pathogen of swine, suppresses the innate immune response and induces persistent infection. One mechanism used by viruses to evade the immune system is to cripple the antigen-processing machinery in monocyte-derived dendritic cells (MoDCs). In this study, we show that MoDCs infected by PRRSV express lower levels of the major histocompatibility complex (MHC)-peptide complex proteins TAP1 and ERp57 and are impaired in their ability to stimulate T cell proliferation and increase their production of CD83. Neutralization of sCD83 removes the inhibitory effects of PRRSV on MoDCs. When MoDCs are incubated with exogenously added sCD83 protein, TAP1 and ERp57 expression decreases and T lymphocyte activation is impaired. PRRSV nonstructural protein 1α (Nsp1α) enhances CD83 promoter activity. Mutations in the ZF domain of Nsp1α abolish its ability to activate the CD83 promoter. We generated recombinant PRRSVs with mutations in Nsp1α and the corresponding repaired PRRSVs. Viruses with Nsp1α mutations did not decrease levels of TAP1 and ERp57, impair the ability of MoDCs to stimulate T cell proliferation, or increase levels of sCD83. We show that the ZF domain of Nsp1α stimulates the secretion of CD83, which in turn inhibits MoDC function. Our study provides new insights into the mechanisms of immune suppression by PRRSV.IMPORTANCE PRRSV has a severe impact on the swine industry throughout the world. Understanding the mechanisms by which PRRSV infection suppresses the immune system is essential for a robust and sustainable swine industry. Here, we demonstrated that PRRSV infection manipulates MoDCs by interfering with their ability to produce proteins in the MHC-peptide complex. The virus also impairs the ability of MoDCs to stimulate cell proliferation, due in large part to the enhanced release of soluble CD83 from PRRSV-infected MoDCs. The viral nonstructural protein 1 (Nsp1) is responsible for upregulating CD83 promoter activity. Amino acids in the ZF domain of Nsp1α (L5-2A, rG45A, G48A, and L61-6A) are essential for CD83 promoter activation. Viruses with mutations at these sites no longer inhibit MoDC-mediated T cell proliferation. These findings provide novel insights into the mechanism by which the adaptive immune response is suppressed during PRRSV infection.
Collapse
Affiliation(s)
- Xi Chen
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Juan Bai
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xuewei Liu
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Zhongbao Song
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Qiaoya Zhang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xianwei Wang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ping Jiang
- Key Laboratory of Animal Diseases Diagnostic and Immunology, Ministry of Agriculture, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
- Jiangsu Coinnovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| |
Collapse
|
20
|
Axonal Activation of the Unfolded Protein Response Promotes Axonal Regeneration Following Peripheral Nerve Injury. Neuroscience 2018; 375:34-48. [PMID: 29438804 DOI: 10.1016/j.neuroscience.2018.02.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2017] [Revised: 02/01/2018] [Accepted: 02/02/2018] [Indexed: 01/04/2023]
Abstract
Adult mammalian peripheral neurons have an intrinsic regrowth capacity in response to axonal injury. The induction of calcium ion (Ca2+) oscillations at an injured site is critical for the regulation of regenerative responses. In polarized neurons, distal axonal segments contain a well-developed endoplasmic reticulum (ER) network that is responsible for Ca2+ homeostasis. Although these characteristics implicate the relevance among injury-induced Ca2+ dynamics, axonal ER-derived signaling, and regenerative responses propagated along the axons, the details are not fully understood. In the present study, we found that Ca2+ release from the axonal ER was accelerated in response to injury. Additionally, axonal injury-dependent Ca2+ release from the ER activated unfolded protein response (UPR) signaling at injured sites. Inhibition of axonal UPR signaling led to fragmentation of the axonal ER and disrupted growth cone formation, suggesting that activation of axonal UPR branches following axonal injury promotes regeneration via regulation of ER reconstruction and formation of growth cones. Our studies revealed that local activation of axonal UPR signaling by injury-induced Ca2+ release from the ER is critical for regeneration. These findings provide a new concept for the link between injury-induced signaling at a distant location and regulation of organelle and cytoskeletal formation in the orchestration of axonal regeneration.
Collapse
|
21
|
Parakh S, Jagaraj CJ, Vidal M, Ragagnin AMG, Perri ER, Konopka A, Toth RP, Galper J, Blair IP, Thomas CJ, Walker AK, Yang S, Spencer DM, Atkin JD. ERp57 is protective against mutant SOD1-induced cellular pathology in amyotrophic lateral sclerosis. Hum Mol Genet 2018; 27:1311-1331. [DOI: 10.1093/hmg/ddy041] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/29/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Sonam Parakh
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Cyril J Jagaraj
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Marta Vidal
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Audrey M G Ragagnin
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Emma R Perri
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Anna Konopka
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Reka P Toth
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Jasmin Galper
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Ian P Blair
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Colleen J Thomas
- Department of Physiology, Anatomy and Microbiology, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Adam K Walker
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Shu Yang
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
| | - Damian M Spencer
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| | - Julie D Atkin
- Centre for MND Research, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Macquarie University, Sydney, NSW 2109, Australia
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC 3086, Australia
| |
Collapse
|
22
|
Maffioli E, Schulte C, Nonnis S, Grassi Scalvini F, Piazzoni C, Lenardi C, Negri A, Milani P, Tedeschi G. Proteomic Dissection of Nanotopography-Sensitive Mechanotransductive Signaling Hubs that Foster Neuronal Differentiation in PC12 Cells. Front Cell Neurosci 2018; 11:417. [PMID: 29354032 PMCID: PMC5758595 DOI: 10.3389/fncel.2017.00417] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 12/12/2017] [Indexed: 12/11/2022] Open
Abstract
Neuronal cells are competent in precisely sensing nanotopographical features of their microenvironment. The perceived microenvironmental information will be “interpreted” by mechanotransductive processes and impacts on neuronal functioning and differentiation. Attempts to influence neuronal differentiation by engineering substrates that mimic appropriate extracellular matrix (ECM) topographies are hampered by the fact that profound details of mechanosensing/-transduction complexity remain elusive. Introducing omics methods into these biomaterial approaches has the potential to provide a deeper insight into the molecular processes and signaling cascades underlying mechanosensing/-transduction but their exigence in cellular material is often opposed by technical limitations of major substrate top-down fabrication methods. Supersonic cluster beam deposition (SCBD) allows instead the bottom-up fabrication of nanostructured substrates over large areas characterized by a quantitatively controllable ECM-like nanoroughness that has been recently shown to foster neuron differentiation and maturation. Exploiting this capacity of SCBD, we challenged mechanosensing/-transduction and differentiative behavior of neuron-like PC12 cells with diverse nanotopographies and/or changes of their biomechanical status, and analyzed their phosphoproteomic profiles in these settings. Versatile proteins that can be associated to significant processes along the mechanotransductive signal sequence, i.e., cell/cell interaction, glycocalyx and ECM, membrane/f-actin linkage and integrin activation, cell/substrate interaction, integrin adhesion complex, actomyosin organization/cellular mechanics, nuclear organization, and transcriptional regulation, were affected. The phosphoproteomic data suggested furthermore an involvement of ILK, mTOR, Wnt, and calcium signaling in these nanotopography- and/or cell mechanics-related processes. Altogether, potential nanotopography-sensitive mechanotransductive signaling hubs participating in neuronal differentiation were dissected.
Collapse
Affiliation(s)
- Elisa Maffioli
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy
| | - Carsten Schulte
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Simona Nonnis
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Francesca Grassi Scalvini
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Claudio Piazzoni
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Cristina Lenardi
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Armando Negri
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| | - Paolo Milani
- Centre for Nanostructured Materials and Interfaces, Università degli Studi di Milano, Milan, Italy
| | - Gabriella Tedeschi
- Department of Veterinary Medicine, Università degli Studi di Milano, Milan, Italy.,Fondazione Filarete, Milan, Italy
| |
Collapse
|
23
|
Abstract
The clinical manifestation of neurodegenerative diseases is initiated by the selective alteration in the functionality of distinct neuronal populations. The pathology of many neurodegenerative diseases includes accumulation of misfolded proteins in the brain. In physiological conditions, the proteostasis network maintains normal protein folding, trafficking and degradation; alterations in this network - particularly disturbances to the function of endoplasmic reticulum (ER) - are thought to contribute to abnormal protein aggregation. ER stress triggers a signalling reaction known as the unfolded protein response (UPR), which induces adaptive programmes that improve protein folding and promote quality control mechanisms and degradative pathways or can activate apoptosis when damage is irreversible. In this Review, we discuss the latest advances in defining the functional contribution of ER stress to brain diseases, including novel evidence that relates the UPR to synaptic function, which has implications for cognition and memory. A complex concept is emerging wherein the consequences of ER stress can differ drastically depending on the disease context and the UPR signalling pathway that is altered. Strategies to target specific components of the UPR using small molecules and gene therapy are in development, and promise interesting avenues for future interventions to delay or stop neurodegeneration.
Collapse
|
24
|
Abstract
The protein disulfide isomerase (PDI) gene family is a protein family classically characterized by endoplasmic reticulum (ER) localization and isomerase and redox activity. ERp57, a prominent multifunctional member of the PDI family, is detected at various levels in multiple cellular localizations outside of the ER. ERp57 has been functionally linked to a host of physiological processes and numerous studies have demonstrated altered expression and aberrant functionality of ERp57 in association with diverse pathological states. Here, we summarize available knowledge of ERp57's functions in subcellular compartments and the roles of dysregulated ERp57 in various diseases toward an emphasis on the potential utility of therapeutic development of ERp57.
Collapse
Affiliation(s)
- Aubryanna Hettinghouse
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA
| | - Ronghan Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA
| | - Chuan-Ju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA; Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
25
|
Sepulveda M, Rozas P, Hetz C, Medinas DB. ERp57 as a novel cellular factor controlling prion protein biosynthesis: Therapeutic potential of protein disulfide isomerases. Prion 2017; 10:50-6. [PMID: 26864548 DOI: 10.1080/19336896.2015.1129485] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Disturbance of endoplasmic reticulum (ER) proteostasis is observed in Prion-related disorders (PrDs). The protein disulfide isomerase ERp57 is a stress-responsive ER chaperone up-regulated in the brain of Creutzfeldt-Jakob disease patients. However, the actual role of ERp57 in prion protein (PrP) biogenesis and the ER stress response remained poorly defined. We have recently addressed this question using gain- and loss-of-function approaches in vitro and animal models, observing that ERp57 regulates steady-state levels of PrP. Our results revealed that ERp57 modulates the biosynthesis and maturation of PrP but, surprisingly, does not contribute to the global cellular reaction against ER stress in neurons. Here we discuss the relevance of ERp57 as a possible therapeutic target in PrDs and other protein misfolding disorders.
Collapse
Affiliation(s)
- Martin Sepulveda
- a Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile , Santiago , Chile.,b Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile , Santiago , Chile.,c Center for Geroscience, Brain Health and Metabolism, University of Chile , Santiago , Chile
| | - Pablo Rozas
- a Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile , Santiago , Chile.,b Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile , Santiago , Chile.,c Center for Geroscience, Brain Health and Metabolism, University of Chile , Santiago , Chile
| | - Claudio Hetz
- a Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile , Santiago , Chile.,b Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile , Santiago , Chile.,c Center for Geroscience, Brain Health and Metabolism, University of Chile , Santiago , Chile.,d Harvard School of Public Health , Boston , MA , USA
| | - Danilo B Medinas
- a Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile , Santiago , Chile.,b Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile , Santiago , Chile.,c Center for Geroscience, Brain Health and Metabolism, University of Chile , Santiago , Chile
| |
Collapse
|
26
|
Rozas P, Bargsted L, Martínez F, Hetz C, Medinas DB. The ER proteostasis network in ALS: Determining the differential motoneuron vulnerability. Neurosci Lett 2017; 636:9-15. [DOI: 10.1016/j.neulet.2016.04.066] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Revised: 04/17/2016] [Accepted: 04/29/2016] [Indexed: 12/13/2022]
|
27
|
Perri E, Parakh S, Atkin J. Protein Disulphide Isomerases: emerging roles of PDI and ERp57 in the nervous system and as therapeutic targets for ALS. Expert Opin Ther Targets 2016; 21:37-49. [PMID: 27786579 DOI: 10.1080/14728222.2016.1254197] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION There is increasing evidence that endoplasmic reticulum (ER) chaperones Protein Disulphide Isomerase (PDI) and ERp57 (endoplasmic reticulum protein 57) are protective against neurodegenerative diseases related to protein misfolding, including Amyotrophic Lateral Sclerosis (ALS). PDI and ERp57 also possess disulphide interchange activity, in which protein disulphide bonds are oxidized, reduced and isomerized, to form their native conformation. Recently, missense and intronic variants of PDI and ERp57 were associated with ALS, implying that PDI proteins are relevant to ALS pathology. Areas covered: Here, we discuss possible implications of the PDI and ERp57 variants, as well as recent studies describing previously unrecognized roles for PDI and ERp57 in the nervous system. Therapeutics based on PDI may therefore be attractive candidates for ALS. However, in addition to its protective functions, aberrant, toxic roles for PDI have recently been described. These functions need to be fully characterized before effective therapeutic strategies can be designed. Expert opinion: These disease-associated variants of PDI and ERp57 provide additional evidence for an important role for PDI proteins in ALS. However, there are many questions remaining unanswered that need to be addressed before the potential of the PDI family in relation to ALS can be fully realized.
Collapse
Affiliation(s)
- Emma Perri
- a Department of Biomedical Sciences, Faculty of Medicine and Health Sciences , Macquarie University , Sydney , Australia
| | - Sonam Parakh
- a Department of Biomedical Sciences, Faculty of Medicine and Health Sciences , Macquarie University , Sydney , Australia
| | - Julie Atkin
- a Department of Biomedical Sciences, Faculty of Medicine and Health Sciences , Macquarie University , Sydney , Australia.,b Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science , La Trobe University , Melbourne , Australia
| |
Collapse
|
28
|
Oñate M, Court FA, Hetz C. Bursting the unfolded protein response accelerates axonal regeneration. Neural Regen Res 2016; 11:892-3. [PMID: 27482204 PMCID: PMC4962573 DOI: 10.4103/1673-5374.184453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Affiliation(s)
- Maritza Oñate
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago, Chile; Millenium Nucleus for Regenerative Biology, Santiago, Chile
| | - Felipe A Court
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Center for Integrative Biology, Universidad Mayor, Santiago, Chile; Millenium Nucleus for Regenerative Biology, Santiago, Chile
| | - Claudio Hetz
- Geroscience Center for Brain Health and Metabolism, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA, 94945, USA; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|
29
|
Garcia-Huerta P, Bargsted L, Rivas A, Matus S, Vidal RL. ER chaperones in neurodegenerative disease: Folding and beyond. Brain Res 2016; 1648:580-587. [PMID: 27134034 DOI: 10.1016/j.brainres.2016.04.070] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 04/27/2016] [Accepted: 04/28/2016] [Indexed: 12/13/2022]
Abstract
Proteins along the secretory pathway are co-translationally translocated into the lumen of the endoplasmic reticulum (ER) as unfolded polypeptide chains. Afterwards, they are usually modified with N-linked glycans, correctly folded and stabilized by disulfide bonds. ER chaperones and folding enzymes control these processes. The accumulation of unfolded proteins in the ER activates a signaling response, termed the unfolded protein response (UPR). The hallmark of this response is the coordinated transcriptional up-regulation of ER chaperones and folding enzymes. In order to discuss the importance of the proper folding of certain substrates we will address the role of ER chaperones in normal physiological conditions and examine different aspects of its contribution in neurodegenerative disease. This article is part of a Special Issue entitled SI:ER stress.
Collapse
Affiliation(s)
- 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, Center for Molecular Studies of the Cell Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Leslie Bargsted
- 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, Center for Molecular Studies of the Cell Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Alexis Rivas
- 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, Center for Molecular Studies of the Cell Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Soledad Matus
- Neurounion Biomedical Foundation, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; CENPAR, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.
| | - Rene L Vidal
- Neurounion Biomedical Foundation, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; CENPAR, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.
| |
Collapse
|
30
|
Valenzuela V, Martínez G, Duran-Aniotz C, Hetz C. Gene therapy to target ER stress in brain diseases. Brain Res 2016; 1648:561-570. [PMID: 27131987 DOI: 10.1016/j.brainres.2016.04.064] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/12/2016] [Accepted: 04/26/2016] [Indexed: 02/07/2023]
Abstract
Gene therapy based on the use of Adeno-associated viruses (AAVs) is emerging as a safe and stable strategy to target molecular pathways involved in a variety of brain diseases. Endoplasmic reticulum (ER) stress is proposed as a transversal feature of most animal models and clinical samples from patients affected with neurodegenerative diseases. Manipulation of the unfolded protein response (UPR), a major homeostatic reaction under ER stress conditions, had proved beneficial in diverse models of neurodegeneration. Although increasing number of drugs are available to target ER stress, the use of small molecules to treat chronic brain diseases is challenging because of poor blood brain barrier permeability and undesirable side effects due to the role of the UPR in the physiology of peripheral organs. Gene therapy is currently considered a possible future alternative to circumvent these problems by the delivery of therapeutic agents to selective regions and cell types of the nervous system. Here we discuss current efforts to design gene therapy strategies to alleviate ER stress on a disease context. This article is part of a Special Issue entitled SI:ER stress.
Collapse
Affiliation(s)
- Vicente Valenzuela
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Gabriela Martínez
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Claudia Duran-Aniotz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, 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, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Immunology and Infectious diseases, Harvard School of Public Health, 02115 Boston, MA, USA.
| |
Collapse
|
31
|
Injury to the nervous system: A look into the ER. Brain Res 2016; 1648:617-625. [PMID: 27117870 DOI: 10.1016/j.brainres.2016.04.053] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 12/12/2022]
Abstract
Injury to the central or peripheral nervous systems leads to the loss of cognitive and/or sensorimotor capabilities that still lack an effective treatment. Although injury to the nervous system involves multiple and complex molecular factors, alteration to protein homeostasis is emerging as a relevant pathological mechanism. In particular, chronic endoplasmic reticulum (ER) stress is proposed as a possible driver of neuronal dysfunction in conditions such as spinal cord injury, stroke and damage to peripheral nerves. Importantly, manipulation of the unfolded protein response (UPR), a homeostatic pathway engaged by ER stress, has proved effective in improving cognitive and motor recovery after nervous system injury. Here we provide an overview on recent findings depicting a functional role of the UPR to the functional recovery after injury in the peripheral and central nervous systems. This article is part of a Special Issue entitled SI:ER stress.
Collapse
|
32
|
Woehlbier U, Colombo A, Saaranen MJ, Pérez V, Ojeda J, Bustos FJ, Andreu CI, Torres M, Valenzuela V, Medinas DB, Rozas P, Vidal RL, Lopez-Gonzalez R, Salameh J, Fernandez-Collemann S, Muñoz N, Matus S, Armisen R, Sagredo A, Palma K, Irrazabal T, Almeida S, Gonzalez-Perez P, Campero M, Gao FB, Henny P, van Zundert B, Ruddock LW, Concha ML, Henriquez JP, Brown RH, Hetz C. ALS-linked protein disulfide isomerase variants cause motor dysfunction. EMBO J 2016; 35:845-65. [PMID: 26869642 PMCID: PMC4972141 DOI: 10.15252/embj.201592224] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2015] [Revised: 12/27/2015] [Accepted: 01/05/2016] [Indexed: 12/11/2022] Open
Abstract
Disturbance of endoplasmic reticulum (ER) proteostasis is a common feature of amyotrophic lateral sclerosis (ALS). Protein disulfide isomerases (PDIs) areERfoldases identified as possibleALSbiomarkers, as well as neuroprotective factors. However, no functional studies have addressed their impact on the disease process. Here, we functionally characterized fourALS-linked mutations recently identified in two majorPDIgenes,PDIA1 andPDIA3/ERp57. Phenotypic screening in zebrafish revealed that the expression of thesePDIvariants induce motor defects associated with a disruption of motoneuron connectivity. Similarly, the expression of mutantPDIs impaired dendritic outgrowth in motoneuron cell culture models. Cellular and biochemical studies identified distinct molecular defects underlying the pathogenicity of thesePDImutants. Finally, targetingERp57 in the nervous system led to severe motor dysfunction in mice associated with a loss of neuromuscular synapses. This study identifiesERproteostasis imbalance as a risk factor forALS, driving initial stages of the disease.
Collapse
Affiliation(s)
- Ute Woehlbier
- 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 Genomics and Bioinformatics, Universidad Mayor, Santiago, Chile
| | - Alicia Colombo
- Program of Anatomy and Developmental Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile Department of Pathological Anatomy, Hospital Clínico, University of Chile, Santiago, Chile
| | - Mirva J Saaranen
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Viviana Pérez
- Department of Cell Biology, Faculty of Biological Sciences, Millennium Nucleus of Regenerative Biology, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Jorge Ojeda
- Department of Cell Biology, Faculty of Biological Sciences, Millennium Nucleus of Regenerative Biology, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Fernando J Bustos
- Faculty of Biological Sciences and Faculty of Medicine, Center for Biomedical Research, Universidad Andres Bello, Santiago, Chile
| | - Catherine I Andreu
- 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
| | - Mauricio Torres
- 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
| | - Vicente Valenzuela
- 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
| | - 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
| | - Pablo Rozas
- 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
| | - Rene L Vidal
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile Center for Geroscience, Brain Health and Metabolism, Santiago, Chile Neurounion Biomedical Foundation, CENPAR, Santiago, Chile
| | | | - Johnny Salameh
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Natalia Muñoz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile Center for Geroscience, Brain Health and Metabolism, Santiago, Chile Neurounion Biomedical Foundation, CENPAR, Santiago, Chile
| | - Soledad Matus
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile Center for Geroscience, Brain Health and Metabolism, Santiago, Chile Neurounion Biomedical Foundation, CENPAR, Santiago, Chile
| | - Ricardo Armisen
- Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Alfredo Sagredo
- Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| | - Karina Palma
- 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
| | - Thergiory Irrazabal
- 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
| | - Sandra Almeida
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Paloma Gonzalez-Perez
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Mario Campero
- Department of Neurology and Neurosurgery, Faculty of Medicine, University of Chile, Santiago, Chile Faculty of Medicine, Clínica Alemana, Universidad del Desarrollo, Santiago, Chile
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Pablo Henny
- Department of Anatomy, Medical School, Universidad Católica de Chile, Santiago, Chile
| | - Brigitte van Zundert
- Faculty of Biological Sciences and Faculty of Medicine, Center for Biomedical Research, Universidad Andres Bello, Santiago, Chile
| | - Lloyd W Ruddock
- Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Miguel L Concha
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile Program of Anatomy and Developmental Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Juan P Henriquez
- Department of Cell Biology, Faculty of Biological Sciences, Millennium Nucleus of Regenerative Biology, Center for Advanced Microscopy (CMA Bio-Bio), Universidad de Concepción, Concepción, Chile
| | - Robert H Brown
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - 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 Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| |
Collapse
|
33
|
Affiliation(s)
- Leslie Bargsted
- Neurounion Biomedical Foundation, CENPAR, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Claudio Hetz
- Neurounion Biomedical Foundation, CENPAR, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, Program of Cellular and Molecular Biology, University of Chile, Santiago, Chile; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA
| | - Soledad Matus
- Neurounion Biomedical Foundation, CENPAR, Santiago, Chile; Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| |
Collapse
|
34
|
Activation of the unfolded protein response promotes axonal regeneration after peripheral nerve injury. Sci Rep 2016; 6:21709. [PMID: 26906090 PMCID: PMC4764858 DOI: 10.1038/srep21709] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 01/12/2016] [Indexed: 12/13/2022] Open
Abstract
Although protein-folding stress at the endoplasmic reticulum (ER) is emerging as a driver of neuronal dysfunction in models of spinal cord injury and neurodegeneration, the contribution of this pathway to peripheral nerve damage remains poorly explored. Here we targeted the unfolded protein response (UPR), an adaptive reaction against ER stress, in mouse models of sciatic nerve injury and found that ablation of the transcription factor XBP1, but not ATF4, significantly delay locomotor recovery. XBP1 deficiency led to decreased macrophage recruitment, a reduction in myelin removal and axonal regeneration. Conversely, overexpression of XBP1s in the nervous system in transgenic mice enhanced locomotor recovery after sciatic nerve crush, associated to an improvement in key pro-regenerative events. To assess the therapeutic potential of UPR manipulation to axonal regeneration, we locally delivered XBP1s or an shRNA targeting this transcription factor to sensory neurons of the dorsal root ganglia using a gene therapy approach and found an enhancement or reduction of axonal regeneration in vivo, respectively. Our results demonstrate a functional role of specific components of the ER proteostasis network in the cellular changes associated to regeneration and functional recovery after peripheral nerve injury.
Collapse
|
35
|
Perri ER, Thomas CJ, Parakh S, Spencer DM, Atkin JD. The Unfolded Protein Response and the Role of Protein Disulfide Isomerase in Neurodegeneration. Front Cell Dev Biol 2016; 3:80. [PMID: 26779479 PMCID: PMC4705227 DOI: 10.3389/fcell.2015.00080] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 12/03/2015] [Indexed: 12/11/2022] Open
Abstract
The maintenance and regulation of proteostasis is a critical function for post-mitotic neurons and its dysregulation is increasingly implicated in neurodegenerative diseases. Despite having different clinical manifestations, these disorders share similar pathology; an accumulation of misfolded proteins in neurons and subsequent disruption to cellular proteostasis. The endoplasmic reticulum (ER) is an important component of proteostasis, and when the accumulation of misfolded proteins occurs within the ER, this disturbs ER homeostasis, giving rise to ER stress. This triggers the unfolded protein response (UPR), distinct signaling pathways that whilst initially protective, are pro-apoptotic if ER stress is prolonged. ER stress is increasingly implicated in neurodegenerative diseases, and emerging evidence highlights the complexity of the UPR in these disorders, with both protective and detrimental components being described. Protein Disulfide Isomerase (PDI) is an ER chaperone induced during ER stress that is responsible for the formation of disulfide bonds in proteins. Whilst initially considered to be protective, recent studies have revealed unconventional roles for PDI in neurodegenerative diseases, distinct from its normal function in the UPR and the ER, although these mechanisms remain poorly defined. However, specific aspects of PDI function may offer the potential to be exploited therapeutically in the future. This review will focus on the evidence linking ER stress and the UPR to neurodegenerative diseases, with particular emphasis on the emerging functions ascribed to PDI in these conditions.
Collapse
Affiliation(s)
- Emma R Perri
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Melbourne, VIC, Australia
| | - Colleen J Thomas
- Department of Physiology, Anatomy and Microbiology, School of Life Sciences, La Trobe University Melbourne, VIC, Australia
| | - Sonam Parakh
- Department of Biomedical Sciences, Faculty of Medicine and Human Science, Macquarie University Sydney, NSW, Australia
| | - Damian M Spencer
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University Melbourne, VIC, Australia
| | - Julie D Atkin
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe UniversityMelbourne, VIC, Australia; Department of Biomedical Sciences, Faculty of Medicine and Human Science, Macquarie UniversitySydney, NSW, Australia
| |
Collapse
|
36
|
Khachatoorian R, French SW. Chaperones in hepatitis C virus infection. World J Hepatol 2016; 8:9-35. [PMID: 26783419 PMCID: PMC4705456 DOI: 10.4254/wjh.v8.i1.9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 10/01/2015] [Accepted: 12/18/2015] [Indexed: 02/06/2023] Open
Abstract
The hepatitis C virus (HCV) infects approximately 3% of the world population or more than 185 million people worldwide. Each year, an estimated 350000-500000 deaths occur worldwide due to HCV-associated diseases including cirrhosis and hepatocellular carcinoma. HCV is the most common indication for liver transplantation in patients with cirrhosis worldwide. HCV is an enveloped RNA virus classified in the genus Hepacivirus in the Flaviviridae family. The HCV viral life cycle in a cell can be divided into six phases: (1) binding and internalization; (2) cytoplasmic release and uncoating; (3) viral polyprotein translation and processing; (4) RNA genome replication; (5) encapsidation (packaging) and assembly; and (6) virus morphogenesis (maturation) and secretion. Many host factors are involved in the HCV life cycle. Chaperones are an important group of host cytoprotective molecules that coordinate numerous cellular processes including protein folding, multimeric protein assembly, protein trafficking, and protein degradation. All phases of the viral life cycle require chaperone activity and the interaction of viral proteins with chaperones. This review will present our current knowledge and understanding of the role of chaperones in the HCV life cycle. Analysis of chaperones in HCV infection will provide further insights into viral/host interactions and potential therapeutic targets for both HCV and other viruses.
Collapse
|
37
|
|
38
|
Torres M, Medinas DB, Matamala JM, Woehlbier U, Cornejo VH, Solda T, Andreu C, Rozas P, Matus S, Muñoz N, Vergara C, Cartier L, Soto C, Molinari M, Hetz C. The Protein-disulfide Isomerase ERp57 Regulates the Steady-state Levels of the Prion Protein. J Biol Chem 2015; 290:23631-45. [PMID: 26170458 DOI: 10.1074/jbc.m114.635565] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Indexed: 12/19/2022] Open
Abstract
Although the accumulation of a misfolded and protease-resistant form of the prion protein (PrP) is a key event in prion pathogenesis, the cellular factors involved in its folding and quality control are poorly understood. PrP is a glycosylated and disulfide-bonded protein synthesized at the endoplasmic reticulum (ER). The ER foldase ERp57 (also known as Grp58) is highly expressed in the brain of sporadic and infectious forms of prion-related disorders. ERp57 is a disulfide isomerase involved in the folding of a subset of glycoproteins in the ER as part of the calnexin/calreticulin cycle. Here, we show that levels of ERp57 increase mainly in neurons of Creutzfeldt-Jacob patients. Using gain- and loss-of-function approaches in cell culture, we demonstrate that ERp57 expression controls the maturation and total levels of wild-type PrP and mutant forms associated with human disease. In addition, we found that PrP physically interacts with ERp57, and also with the closest family member PDIA1, but not ERp72. Furthermore, we generated a conditional knock-out mouse for ERp57 in the nervous system and detected a reduction in the steady-state levels of the mono- and nonglycosylated forms of PrP in the brain. In contrast, ERp57 transgenic mice showed increased levels of endogenous PrP. Unexpectedly, ERp57 expression did not affect the susceptibility of cells to ER stress in vitro and in vivo. This study identifies ERp57 as a new modulator of PrP levels and may help with understanding the consequences of ERp57 up-regulation observed in human disease.
Collapse
Affiliation(s)
- Mauricio Torres
- From the Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile, the Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago 8380453, Chile
| | - Danilo B Medinas
- From the Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile, the Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago 8380453, Chile
| | - José Manuel Matamala
- From the Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile, the Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago 7500691, Chile
| | - Ute Woehlbier
- From the Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile, the Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago 8380453, Chile
| | - Víctor Hugo Cornejo
- From the Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile, the Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago 8380453, Chile
| | - Tatiana Solda
- the Institute for Research in Biomedicine, Bellinzona CH6500, Switzerland
| | - Catherine Andreu
- From the Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile, the Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago 8380453, Chile
| | - Pablo Rozas
- From the Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile, the Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago 8380453, Chile
| | - Soledad Matus
- From the Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile, the Neurounion Biomedical Foundation, CENPAR, Santiago 7630614, Chile
| | - Natalia Muñoz
- From the Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile, the Neurounion Biomedical Foundation, CENPAR, Santiago 7630614, Chile
| | - Carmen Vergara
- the Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago 7500691, Chile
| | - Luis Cartier
- the Department of Neurological Sciences, Faculty of Medicine, University of Chile, Santiago 7500691, Chile
| | - Claudio Soto
- the Department of Neurology, University of Texas Medical School, Houston, Texas 77030, and
| | - Maurizio Molinari
- the Institute for Research in Biomedicine, Bellinzona CH6500, Switzerland, the Università della Svizzera Italiana, Lugano CH6900, Switzerland, the Ecole Polytechnique Fédérale de Lausanne, School of Life Sciences, Lausanne CH1015, Switzerland
| | - Claudio Hetz
- From the Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago 8380453, Chile, the Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Center for Molecular Studies of the Cell, University of Chile, Santiago 8380453, Chile, the Harvard School of Public Health, Boston, Massachusetts 02115
| |
Collapse
|