1
|
Hosseini SM, Karimi-Abdolrezaee S. New insights on the role of chondroitin sulfate proteoglycans in neural stem cell-mediated repair in spinal cord injury. Neural Regen Res 2025; 20:1699-1700. [PMID: 39104100 PMCID: PMC11688555 DOI: 10.4103/nrr.nrr-d-24-00378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/25/2024] [Accepted: 06/04/2024] [Indexed: 08/07/2024] Open
Affiliation(s)
- Seyed Mojtaba Hosseini
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Manitoba Multiple Sclerosis Research Center, Winnipeg, MB, Canada
- Children Hospital Research Institute of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
2
|
Lv Y, Ji L, Dai H, Qiu S, Wang Y, Teng C, Yu B, Mi D, Yao C. Identification of key regulatory genes involved in myelination after spinal cord injury by GSEA analysis. Exp Neurol 2024; 382:114966. [PMID: 39326824 DOI: 10.1016/j.expneurol.2024.114966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/31/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024]
Abstract
Multilayer dense myelin tissue provides insulating space and nutritional support for axons in healthy spinal cord tissue. Oligodendrocyte precursor cells (OPCs) are the main glial cells that complement myelin loss in the central nervous system and play an important role in the repair of spinal cord injury (SCI). However, the regulation of axonal remyelination after SCI is still insufficient. In this study, we focused on the changes in genes related to myelin repair after rat hemisection SCI by gene set enrichment analysis (GSEA). Key genes proteolipid protein 1 (Plp1), hexosaminidase subunit alpha (Hexa), and hexosaminidase subunit beta (Hexb) during remyelination after SCI were found. Through quantitative real-time polymerase chain reaction (qPCR) experiments, we confirmed that within 28 days after rat hemisection SCI, the mRNA expression of gene Plp1 gradually decreased, while the expressions of gene Hexa and Hexb gradually increased, which was consistent with RNA sequencing results. In vitro, we performed EdU proliferation assays on OPC cell line OLN-93 and primary rat OPCs. We found that interference of Plp1 promoted OPC proliferation, while interference of Hexa and Hexb inhibited OPC proliferation. In addition, we performed in vitro differentiation experiments on primary rat OPCs. By measuring myelin sheath branch outgrowth and the fluorescence intensity of the mature myelin sheath marker myelin basic protein (MBP), we found that interference of Hexa or Hexb promoted OPC differentiation and maturation, but interference of Plp1 inhibited this process. Finally, we injected Hexb siRNA in vivo and found that interfering Hexb could improve motor movements and myelin regeneration after SCI in rats. Our results provide new target genes that can selectively regulate the proliferation and differentiation of endogenous OPCs, providing new ideas for promoting remyelination and functional recovery after SCI.
Collapse
Affiliation(s)
- Yehua Lv
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Lingyun Ji
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Hui Dai
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Shanru Qiu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Yu Wang
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Cheng Teng
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China
| | - Daguo Mi
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China.
| | - Chun Yao
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Affiliated Traditional Chinese Medicine Hospital and Medical School, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
3
|
Du S, Zhang XX, Gao X, He YB. Structure-based screening of FDA-approved drugs and molecular dynamics simulation to identify potential leukocyte antigen related protein (PTP-LAR) inhibitors. Comput Biol Chem 2024; 113:108264. [PMID: 39488935 DOI: 10.1016/j.compbiolchem.2024.108264] [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: 09/12/2024] [Revised: 10/20/2024] [Accepted: 10/20/2024] [Indexed: 11/05/2024]
Abstract
Leukocyte antigen related protein (LAR), a member of the PTP family, has become a potential target for exploring therapeutic interventions for various complex diseases, including neurodegenerative diseases. The reuse of FDA-approved drugs offers a promising approach for rapidly identifying potential LAR inhibitors. In this study, we conducted a structure-based virtual screening of FDA-approved drugs from ZINC database and selected candidate compounds based on their binding affinity and interactions with LAR. Our research revealed that the candidate compound ZINC6716957 exhibited excellent binding affinity to the binding pocket of LAR, formed interactions with key residues at the active site, and demonstrated low toxicity. To further understand the binding dynamics and interaction mechanisms, the 100-ns molecular dynamics simulations were performed. Post-dynamics analyses (RMSD, RMSF, SASA, hydrogen bond, binding free energy and free energy landscape) indicated that the compound ZINC6716957 stabilized the structure of LAR and the residues (Tyr1355, Arg1431, Lys1433, Arg1528, Tyr1563 and Thr1567) played a vital role in stabilizing the conformational changes of protein. In conclusion, the identified compound ZINC6716957 possessed robust inhibitory activity on LAR and merited extensive research, potentially unleashing its significant therapeutic potential in the treatment of complex diseases, particularly neurodegenerative disorders.
Collapse
Affiliation(s)
- Shan Du
- School of Pharmacy, Changzhi Medical College, 161 East Jiefang Street, Changzhi, Shanxi 046000, PR China
| | - Xin-Xin Zhang
- School of Pharmacy, Changzhi Medical College, 161 East Jiefang Street, Changzhi, Shanxi 046000, PR China
| | - Xiang Gao
- School of Pharmacy, Changzhi Medical College, 161 East Jiefang Street, Changzhi, Shanxi 046000, PR China
| | - Yan-Bin He
- School of Pharmacy, Changzhi Medical College, 161 East Jiefang Street, Changzhi, Shanxi 046000, PR China.
| |
Collapse
|
4
|
Hosseini SM, Nemati S, Karimi-Abdolrezaee S. Astrocytes originated from neural stem cells drive the regenerative remodeling of pathologic CSPGs in spinal cord injury. Stem Cell Reports 2024; 19:1451-1473. [PMID: 39303705 PMCID: PMC11561464 DOI: 10.1016/j.stemcr.2024.08.007] [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: 05/09/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/22/2024] Open
Abstract
Neural degeneration is a hallmark of spinal cord injury (SCI). Multipotent neural precursor cells (NPCs) have the potential to reconstruct the damaged neuron-glia network due to their tri-lineage capacity to generate neurons, astrocytes, and oligodendrocytes. However, astrogenesis is the predominant fate of resident or transplanted NPCs in the SCI milieu adding to the abundant number of resident astrocytes in the lesion. How NPC-derived astrocytes respond to the inflammatory milieu of SCI and the mechanisms by which they contribute to the post-injury recovery processes remain largely unknown. Here, we uncover that activated NPC-derived astrocytes exhibit distinct molecular signature that is immune modulatory and foster neurogenesis, neuronal maturity, and synaptogenesis. Mechanistically, NPC-derived astrocytes perform regenerative matrix remodeling by clearing inhibitory chondroitin sulfate proteoglycans (CSPGs) from the injury milieu through LAR and PTP-σ receptor-mediated endocytosis and the production of ADAMTS1 and ADAMTS9, while most resident astrocytes are pro-inflammatory and contribute to the pathologic deposition of CSPGs. These novel findings unravel critical mechanisms of NPC-mediated astrogenesis in SCI repair.
Collapse
Affiliation(s)
- Seyed Mojtaba Hosseini
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Manitoba Multiple Sclerosis Research Center, Winnipeg, MB, Canada
| | - Shiva Nemati
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Manitoba Multiple Sclerosis Research Center, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada; Manitoba Multiple Sclerosis Research Center, Winnipeg, MB, Canada; Children Hospital Research Institute of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
5
|
Poirier A, Picard C, Labonté A, Aubry I, Auld D, Zetterberg H, Blennow K, Tremblay ML, Poirier J. PTPRS is a novel marker for early Tau pathology and synaptic integrity in Alzheimer's disease. Sci Rep 2024; 14:14718. [PMID: 38926456 PMCID: PMC11208446 DOI: 10.1038/s41598-024-65104-2] [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: 03/21/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
We examined the role of protein tyrosine phosphatase receptor sigma (PTPRS) in the context of Alzheimer's disease and synaptic integrity. Publicly available datasets (BRAINEAC, ROSMAP, ADC1) and a cohort of asymptomatic but "at risk" individuals (PREVENT-AD) were used to explore the relationship between PTPRS and various Alzheimer's disease biomarkers. We identified that PTPRS rs10415488 variant C shows features of neuroprotection against early Tau pathology and synaptic degeneration in Alzheimer's disease. This single nucleotide polymorphism correlated with higher PTPRS transcript abundance and lower p(181)Tau and GAP-43 levels in the CSF. In the brain, PTPRS protein abundance was significantly correlated with the quantity of two markers of synaptic integrity: SNAP25 and SYT-1. We also found the presence of sexual dimorphism for PTPRS, with higher CSF concentrations in males than females. Male carriers for variant C were found to have a 10-month delay in the onset of AD. We thus conclude that PTPRS acts as a neuroprotective receptor in Alzheimer's disease. Its protective effect is most important in males, in whom it postpones the age of onset of the disease.
Collapse
Affiliation(s)
- Alexandre Poirier
- Division of Experimental Medicine, Faculty of Medicine and Health Science, McGill University, Montréal, QC, Canada
- Goodman Cancer Institute, McGill University, Montréal, Canada
| | - Cynthia Picard
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Anne Labonté
- Douglas Mental Health University Institute, Montréal, QC, Canada
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada
| | - Isabelle Aubry
- Goodman Cancer Institute, McGill University, Montréal, Canada
- McGill University, Montréal, QC, Canada
| | - Daniel Auld
- McGill University, Montréal, QC, Canada
- Victor Phillip Dahdaleh Institute of Genomic Medicine, McGill University, Montréal, QC, Canada
| | - Henrik Zetterberg
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- UK Dementia Research Institute at UCL, London, UK
- Hong Kong Center for Neurodegenerative Diseases, Clear Water Bay, Hong Kong, SAR, People's Republic of China
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA
- University of Science and Technology of China, Hefei, Anhui, People's Republic of China
| | - Kaj Blennow
- Department of Psychiatry and Neurochemistry, Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Clinical Neurochemistry Laboratory, Sahlgrenska University Hospital, Mölndal, Sweden
- Department of Neurodegenerative Disease, UCL Institute of Neurology, London, UK
- University of Science and Technology of China, Hefei, Anhui, People's Republic of China
- Institut du Cerveau et de la Moelle épinière (ICM), Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France
| | - Michel L Tremblay
- Division of Experimental Medicine, Faculty of Medicine and Health Science, McGill University, Montréal, QC, Canada.
- Goodman Cancer Institute, McGill University, Montréal, Canada.
- McGill University, Montréal, QC, Canada.
- Department of Biochemistry, McGill University, Montréal, Canada.
| | - Judes Poirier
- Douglas Mental Health University Institute, Montréal, QC, Canada.
- Centre for the Studies in the Prevention of Alzheimer's Disease, Montréal, QC, Canada.
- McGill University, Montréal, QC, Canada.
| |
Collapse
|
6
|
Poirier A, Picard C, Labonté A, Aubry I, Auld D, Zetterberg H, Blennow K, Tremblay ML, Poirier J. PTPRS is a novel marker for early tau pathology and synaptic integrity in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.12.593733. [PMID: 38766183 PMCID: PMC11100782 DOI: 10.1101/2024.05.12.593733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
We examined the role of protein tyrosine phosphatase receptor sigma (PTPRS) in the context of Alzheimer's disease and synaptic integrity. Publicly available datasets (BRAINEAC, ROSMAP, ADC1) and a cohort of asymptomatic but "at risk" individuals (PREVENT-AD) were used to explore the relationship between PTPRS and various Alzheimer's disease biomarkers. We identified that PTPRS rs10415488 variant C shows features of neuroprotection against early tau pathology and synaptic degeneration in Alzheimer's disease. This single nucleotide polymorphism correlated with higher PTPRS transcript abundance and lower P-tau181 and GAP-43 levels in the CSF. In the brain, PTPRS protein abundance was significantly correlated with the quantity of two markers of synaptic integrity: SNAP25 and SYT-1. We also found the presence of sexual dimorphism for PTPRS, with higher CSF concentrations in males than females. Male carriers for variant C were found to have a 10-month delay in the onset of AD. We thus conclude that PTPRS acts as a neuroprotective receptor in Alzheimer's disease. Its protective effect is most important in males, in whom it postpones the age of onset of the disease.
Collapse
|
7
|
Yang R, Zhang Y, Kang J, Zhang C, Ning B. Chondroitin Sulfate Proteoglycans Revisited: Its Mechanism of Generation and Action for Spinal Cord Injury. Aging Dis 2024; 15:153-168. [PMID: 37307832 PMCID: PMC10796098 DOI: 10.14336/ad.2023.0512] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/12/2023] [Indexed: 06/14/2023] Open
Abstract
Reactive astrocytes (RAs) produce chondroitin sulfate proteoglycans (CSPGs) in large quantities after spinal cord injury (SCI) and inhibit axon regeneration through the Rho-associated protein kinase (ROCK) pathway. However, the mechanism of producing CSPGs by RAs and their roles in other aspects are often overlooked. In recent years, novel generation mechanisms and functions of CSPGs have gradually emerged. Extracellular traps (ETs), a new recently discovered phenomenon in SCI, can promote secondary injury. ETs are released by neutrophils and microglia, which activate astrocytes to produce CSPGs after SCI. CSPGs inhibit axon regeneration and play an important role in regulating inflammation as well as cell migration and differentiation; some of these regulations are beneficial. The current review summarized the process of ET-activated RAs to generate CSPGs at the cellular signaling pathway level. Moreover, the roles of CSPGs in inhibiting axon regeneration, regulating inflammation, and regulating cell migration and differentiation were discussed. Finally, based on the above process, novel potential therapeutic targets were proposed to eliminate the adverse effects of CSPGs.
Collapse
Affiliation(s)
- Rui Yang
- Jinan Central Hospital, Shandong University, Jinan, Shandong, China.
| | - Ying Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jianning Kang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Ce Zhang
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bin Ning
- Jinan Central Hospital, Shandong University, Jinan, Shandong, China.
- Central Hospital Affiliated to Shandong First Medical University, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
8
|
Wang R, Li T, Diao S, Chen C. Inhibition of the proteoglycan receptor PTPσ promotes functional recovery on a rodent model of preterm hypoxic-ischemic brain injury. Exp Neurol 2023; 370:114564. [PMID: 37806512 DOI: 10.1016/j.expneurol.2023.114564] [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: 06/25/2023] [Revised: 09/25/2023] [Accepted: 10/05/2023] [Indexed: 10/10/2023]
Abstract
BACKGROUND Preterm white matter injury (WMI) is the most common brain injury in preterm infants and is associated with long-term adverse neurodevelopmental outcomes. Protein tyrosine phosphatase sigma (PTPσ) was discovered as chondroitin sulfate proteoglycan (CSPG) receptor that played roles in inhibiting myelin regeneration in spinal injury, experimental autoimmune encephalomyelitis, and stroke models. However, the role of PTPσ in perinatal WMI is not well understood. AIMS This study examines the effect of PTPσ inhibition on neurodevelopmental outcomes, myelination, and neuroinflammation in a mouse model of preterm WMI. MATERIALS AND METHODS Modified Rice-Vannucci model was performed on postnatal day 3 (P3) C57BL/6 mice. Intracellular Sigma Peptide (ISP) or vehicle was administrated subcutaneously one hour after injury for an additional 14 consecutive days. A battery of behavioral tests was performed to evaluate the short- and long-term effects of ISP on neurobehavioral deficit. Real time qPCR, western blot, immunofluorescence, and transmission electron microscopy were performed to assess white matter development. qPCR and flow cytometry were performed to evaluate neuroinflammation and microglia/macrophage phenotype. RESULTS The expression of PTPσ was increased after preterm WMI. ISP improved short-term neurological outcomes and ameliorated long-term motor and cognitive function of mice after preterm WMI. ISP promoted oligodendrocyte differentiation, maturation, myelination, and improved microstructure of myelin after preterm WMI. Furthermore, ISP administration fostered a beneficial inflammatory response in the acute phase after preterm WMI, inhibited the infiltration of peripheral macrophages, and promoted anti-inflammatory phenotype of microglia/macrophages. CONCLUSION PTPσ inhibition can ameliorate neurofunctional deficit, promote white matter development, modulate neuroinflammation and microglia/macrophage phenotype after preterm WMI. Thus, ISP administration may be a potential therapeutic strategy to improve neurodevelopmental outcomes of perinatal WMI.
Collapse
Affiliation(s)
- Ran Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China; Department of Developmental and Behavioral Pediatrics, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Tiantian Li
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Sihao Diao
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China
| | - Chao Chen
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China; Key Laboratory of Neonatal Diseases, National Health Commission, China.
| |
Collapse
|
9
|
Ganz T, Zveik O, Fainstein N, Lachish M, Rechtman A, Sofer L, Brill L, Ben-Hur T, Vaknin-Dembinsky A. Oligodendrocyte progenitor cells differentiation induction with MAPK/ERK inhibitor fails to support repair processes in the chronically demyelinated CNS. Glia 2023; 71:2815-2831. [PMID: 37610097 DOI: 10.1002/glia.24453] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 07/23/2023] [Accepted: 07/24/2023] [Indexed: 08/24/2023]
Abstract
Remyelination failure is considered a major obstacle in treating chronic-progressive multiple sclerosis (MS). Studies have shown blockage in the differentiation of resident oligodendrocyte progenitor cells (OPC) into myelin-forming cells, suggesting that pushing OPC into a differentiation program might be sufficient to overcome remyelination failure. Others stressed the need for a permissive environment to allow proper activation, migration, and differentiation of OPC. PD0325901, a MAPK/ERK inhibitor, was previously shown to induce OPC differentiation, non-specific immunosuppression, and a significant therapeutic effect in acute demyelinating MS models. We examined PD0325901 effects in the chronically inflamed central nervous system. Treatment with PD0325901 induced OPC differentiation into mature oligodendrocytes with high morphological complexity. However, treatment of Biozzi mice with chronic-progressive experimental autoimmune encephalomyelitis with PD0325901 showed no clinical improvement in comparison to the control group, no reduction in demyelination, nor induction of OPC migration into foci of demyelination. PD0325901 induced a direct general immunosuppressive effect on various cell populations, leading to a diminished phagocytic capability of microglia and less activation of lymph-node cells. It also significantly impaired the immune-modulatory functions of OPC. Our findings suggest OPC regenerative function depends on a permissive environment, which may include pro-regenerative inflammatory elements. Furthermore, they indicate that maintaining a delicate balance between the pro-myelinating and immune functions of OPC is of importance. Thus, the highly complex mission of creating a pro-regenerative environment depends upon an appropriate immune response controlled in time, place, and intensity. We suggest the need to employ a multi-systematic therapeutic approach, which cannot be achieved through a single molecule-based therapy.
Collapse
Affiliation(s)
- Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Marva Lachish
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Lihi Sofer
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Livnat Brill
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Tamir Ben-Hur
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
10
|
Tian T, Zhang S, Yang M. Recent progress and challenges in the treatment of spinal cord injury. Protein Cell 2023; 14:635-652. [PMID: 36856750 PMCID: PMC10501188 DOI: 10.1093/procel/pwad003] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 12/29/2022] [Indexed: 02/12/2023] Open
Abstract
Spinal cord injury (SCI) disrupts the structural and functional connectivity between the higher center and the spinal cord, resulting in severe motor, sensory, and autonomic dysfunction with a variety of complications. The pathophysiology of SCI is complicated and multifaceted, and thus individual treatments acting on a specific aspect or process are inadequate to elicit neuronal regeneration and functional recovery after SCI. Combinatory strategies targeting multiple aspects of SCI pathology have achieved greater beneficial effects than individual therapy alone. Although many problems and challenges remain, the encouraging outcomes that have been achieved in preclinical models offer a promising foothold for the development of novel clinical strategies to treat SCI. In this review, we characterize the mechanisms underlying axon regeneration of adult neurons and summarize recent advances in facilitating functional recovery following SCI at both the acute and chronic stages. In addition, we analyze the current status, remaining problems, and realistic challenges towards clinical translation. Finally, we consider the future of SCI treatment and provide insights into how to narrow the translational gap that currently exists between preclinical studies and clinical practice. Going forward, clinical trials should emphasize multidisciplinary conversation and cooperation to identify optimal combinatorial approaches to maximize therapeutic benefit in humans with SCI.
Collapse
Affiliation(s)
- Ting Tian
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Sensen Zhang
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
| | - Maojun Yang
- Ministry of Education Key Laboratory of Protein Science, Beijing Advanced Innovation Center for Structural Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing 100084, China
- Cryo-EM Facility Center, Southern University of Science and Technology, Shenzhen 518055, China
| |
Collapse
|
11
|
Lu W, Wen J. H 2S-RhoA/ROCK Pathway and Glial Cells in Axonal Remyelination After Ischemic Stroke. Mol Neurobiol 2023; 60:5493-5504. [PMID: 37322287 DOI: 10.1007/s12035-023-03422-8] [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: 12/05/2022] [Accepted: 06/03/2023] [Indexed: 06/17/2023]
Abstract
Ischemic stroke is one of the main reasons of disability and death. Stroke-induced functional deficits are mainly due to the secondary degeneration of the white matter characterized by axonal demyelination and injury of axon-glial integrity. Enhancement of the axonal regeneration and remyelination could promote the neural functional recovery. However, cerebral ischemia-induced activation of RhoA/Rho kinase (ROCK) pathway plays a crucial and harmful role in the process of axonal recovery and regeneration. Inhibition of this pathway could promote the axonal regeneration and remyelination. In addition, hydrogen sulfide (H2S) has the significant neuroprotective role during the recovery of ischemic stroke via inhibiting the inflammatory response and oxidative stress, regulating astrocyte function, promoting the differentiation of endogenous oligodendrocyte precursor cells (OPCs) to mature oligodendrocyte. Among all of these effects, promoting the formation of mature oligodendrocyte is a crucial part of axonal regeneration and remyelination. Furthermore, numerous studies have uncovered the crosstalk between astrocytes and oligodendrocyte, microglial cells and oligodendrocyte in the axonal remyelination following ischemic stroke. The purpose of this review was to discuss the relationship among H2S, RhoA/ROCK pathway, astrocytes, and microglial cells in the axonal remyelination following ischemic stroke to reveal new strategies for preventing and treating this devastating disease.
Collapse
Affiliation(s)
- Weizhuo Lu
- Medical Branch, Hefei Technology College, Hefei, China
| | - Jiyue Wen
- Department of Pharmacology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China.
| |
Collapse
|
12
|
John U, Patro N, Patro IK. Astrogliosis and associated CSPG upregulation adversely affect dendritogenesis, spinogenesis and synaptic activity in the cerebellum of a double-hit rat model of protein malnutrition (PMN) and lipopolysaccharide (LPS) induced bacterial infection. J Chem Neuroanat 2023; 131:102286. [PMID: 37169039 DOI: 10.1016/j.jchemneu.2023.102286] [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: 01/31/2023] [Revised: 05/07/2023] [Accepted: 05/08/2023] [Indexed: 05/13/2023]
Abstract
The extracellular matrix (ECM) plays a vital role in growth, guidance and survival of neurons in the central nervous system (CNS). The chondroitin sulphate proteoglycans (CSPGs) are a type of ECM proteins that are crucial for CNS homeostasis. The major goal of this study was to uncover the effects of astroglial activation and associated intensified expression of CSPGs on dendritogenesis, spinogenesis as well as on synaptic activity in cerebellum following protein malnutrition (PMN) and lipopolysaccharide (LPS) induced bacterial infection. Female Wistar albino rats (3 months old) were switched to control (20% protein) or low protein (LP, 8% protein) diet for 15 days followed by breeding. A set of pups born to control/LP mothers and maintained on respective diets throughout the experimental period constituted the control and LP groups, while a separate set of both control and LP group pups exposed to bacterial infection by a single intraperitoneal injection of LPS (0.3 mg/ kg body weight) on postnatal day-9 (P-9) constituted control+LPS and LP+LPS groups respectively. The consequences of astrogliosis induced CSPG upregulation on cerebellar cytoarchitecture and synaptic activity were studied using standard immunohistochemical and histological tools on P-21 and 6 months of age. The results revealed reactive astrogliosis and associated CSPG upregulation in a double-hit model of PMN and LPS induced bacterial infection resulted in disrupted dendritogenesis, reduced postsynaptic density protein (PSD-95) levels and a deleterious impact on normal spine growth. Such alterations frequently have the potential to cause synaptic dysregulation and inhibition of plasticity both during development as well as adulthood. At the light of our results, we can envision that upregulation of CSPGs in PMN and LPS co-challenged individuals might emerge as an important modulator of brain circuitry and a major causative factor for many neurological disorders.
Collapse
Affiliation(s)
- Urmilla John
- School of Studies in Neuroscience, Jiwaji University, Gwalior, India; School of Studies in Zoology, Jiwaji University, Gwalior, India
| | - Nisha Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior, India
| | - Ishan K Patro
- School of Studies in Neuroscience, Jiwaji University, Gwalior, India; School of Studies in Zoology, Jiwaji University, Gwalior, India.
| |
Collapse
|
13
|
Xiong Y, Fu Y, Li Z, Zheng Y, Cui M, Zhang C, Huang XY, Jian Y, Chen BH. Laquinimod Inhibits Microglial Activation, Astrogliosis, BBB Damage, and Infarction and Improves Neurological Damage after Ischemic Stroke. ACS Chem Neurosci 2023. [PMID: 37161270 DOI: 10.1021/acschemneuro.2c00740] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023] Open
Abstract
Glial activation is involved in neuroinflammation and blood-brain barrier (BBB) damage, which plays a key role in ischemic stroke-induced neuronal damage; therefore, regulating glial activation is an important way to inhibit ischemic brain injury. Effects of laquinimod (LAQ) include inhibiting axonal damage and neuroinflammation in multiple neuronal injury diseases. However, whether laquinimod can exert neuroprotective effects after ischemic stroke remains unknown. In this study, we investigated the effect of LAQ on glial activation, BBB damage, and neuronal damage in an ischemic stroke model. Adult ICR mice were used to create a photothrombotic stroke (PT) model. LAQ was administered orally at 30 min after ischemic injury. Neurobehavioral tests, Evans Blue, immunofluorescence, TUNEL, Nissl staining, and western blot were performed to evaluate the neurofunctional outcome. Quantification of immunofluorescence was evaluated by unbiased stereology. LAQ post-treatment significantly reduced infarction and improved forepaw function at 5 days after PT. Interestingly, LAQ treatment significantly promoted anti-inflammatory microglial activation. Moreover, LAQ treatment reduced astrocyte activation, glial scar formation, and BBB breakdown in ischemic brains. Therefore, this study demonstrated that LAQ post-treatment restricted microglial polarization, astrogliosis, and glial scar and improved BBB damage and behavioral function. LAQ may serve as a novel target to develop new therapeutic agents for ischemic stroke.
Collapse
Affiliation(s)
- Ye Xiong
- The First School of Clinical Medicine, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
| | - Yanqiong Fu
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
| | - Zhuoli Li
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
| | - Yu Zheng
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
| | - Maiyin Cui
- Department of Rehabilitation and Traditional Chinese Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou 310052, Zhejiang, P. R. China
| | - Chan Zhang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
| | - Xin Yi Huang
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
| | - Yong Jian
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, Zhejiang, P. R. China
| | - Bai Hui Chen
- Department of Histology and Embryology, Institute of Neuroscience, Wenzhou Medical University, Wenzhou 325035, Zhejiang, P. R. China
| |
Collapse
|
14
|
Neuregulin-1/PI3K signaling effects on oligodendrocyte proliferation, remyelination and behaviors deficit in a male mouse model of ischemic stroke. Exp Neurol 2023; 362:114323. [PMID: 36690057 DOI: 10.1016/j.expneurol.2023.114323] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 01/12/2023] [Accepted: 01/13/2023] [Indexed: 01/22/2023]
Abstract
In this study, we investigated the effect of neuregulin-1 (NRG1) on demyelination and neurological function in an ischemic stroke model, and further explored its neuroprotective mechanisms. Adult male ICR mice underwent photothrombotic ischemia surgery and were injected with NRG1 beginning 30 min after ischemia. Cylinder and grid walking tests were performed to evaluate the forepaw function. In addition, the effect of NRG1 on neuronal damage/death (Cresyl violet, CV), neuronal nuclei (NeuN), nestin, doublecortin (DCX), myelin basic protein (MBP), non-phosphorylated neurofilaments (SMI-32), adenomatous polyposis coli (APC), erythroblastic leukemia viral oncogene homolog (ErbB) 2, 4 and serine-threonine protein kinase (Akt) in cortex were evaluated using immunohistochemistry, immunofluorescence and western blot. The cylinder and grid walking tests exposed that treatment of NRG1 observably regained the forepaw function. NRG1 treatment reduced cerebral infarction, restored forepaw function, promoted proliferation and differentiation of neuron and increased oligodendrogliogenesis. The neuroprotective effect of NRG1 is involved in its activation of PI3K/Akt signaling pathway via ErbB2, as shown by the suppression of the effect of NRG1 by the PI3K inhibitor LY294002. Our results demonstrate that NRG1 is effective in ameliorating the both acute phase neuroprotection and long-term neurological functions via resumption of neuronal proliferation and differentiation and oligodendrogliogenesis in a male mouse model of ischemic stroke.
Collapse
|
15
|
Almeida F, Marques S, Santos A, Prins C, Cardoso F, Heringer L, Mendonça H, Martinez A. Molecular approaches for spinal cord injury treatment. Neural Regen Res 2023; 18:23-30. [PMID: 35799504 PMCID: PMC9241396 DOI: 10.4103/1673-5374.344830] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Injuries to the spinal cord result in permanent disabilities that limit daily life activities. The main reasons for these poor outcomes are the limited regenerative capacity of central neurons and the inhibitory milieu that is established upon traumatic injuries. Despite decades of research, there is still no efficient treatment for spinal cord injury. Many strategies are tested in preclinical studies that focus on ameliorating the functional outcomes after spinal cord injury. Among these, molecular compounds are currently being used for neurological recovery, with promising results. These molecules target the axon collapsed growth cone, the inhibitory microenvironment, the survival of neurons and glial cells, and the re-establishment of lost connections. In this review we focused on molecules that are being used, either in preclinical or clinical studies, to treat spinal cord injuries, such as drugs, growth and neurotrophic factors, enzymes, and purines. The mechanisms of action of these molecules are discussed, considering traumatic spinal cord injury in rodents and humans.
Collapse
|
16
|
John U, Patro N, Patro I. Perineuronal nets: Cruise from a honeycomb to the safety nets. Brain Res Bull 2022; 190:179-194. [DOI: 10.1016/j.brainresbull.2022.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 09/17/2022] [Accepted: 10/05/2022] [Indexed: 11/30/2022]
|
17
|
Luo F, Wang J, Zhang Z, You Z, Bedolla A, Okwubido-Williams F, Huang LF, Silver J, Luo Y. Inhibition of CSPG receptor PTPσ promotes migration of newly born neuroblasts, axonal sprouting, and recovery from stroke. Cell Rep 2022; 40:111137. [PMID: 35905716 PMCID: PMC9677607 DOI: 10.1016/j.celrep.2022.111137] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/16/2022] [Accepted: 07/05/2022] [Indexed: 12/12/2022] Open
Abstract
In addition to neuroprotective strategies, neuroregenerative processes could provide targets for stroke recovery. However, the upregulation of inhibitory chondroitin sulfate proteoglycans (CSPGs) impedes innate regenerative efforts. Here, we examine the regulatory role of PTPσ (a major proteoglycan receptor) in dampening post-stroke recovery. Use of a receptor modulatory peptide (ISP) or Ptprs gene deletion leads to increased neurite outgrowth and enhanced NSCs migration upon inhibitory CSPG substrates. Post-stroke ISP treatment results in increased axonal sprouting as well as neuroblast migration deeply into the lesion scar with a transcriptional signature reflective of repair. Lastly, peptide treatment post-stroke (initiated acutely or more chronically at 7 days) results in improved behavioral recovery in both motor and cognitive functions. Therefore, we propose that CSPGs induced by stroke play a predominant role in the regulation of neural repair and that blocking CSPG signaling pathways will lead to enhanced neurorepair and functional recovery in stroke.
Collapse
Affiliation(s)
- Fucheng Luo
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Jiapeng Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Zhen Zhang
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - Zhen You
- Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Alicia Bedolla
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - FearGod Okwubido-Williams
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA
| | - L Frank Huang
- Division of Experimental Hematology and Cancer Biology, Brain Tumor Center, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA; Department of Pediatrics, College of Medicine, University of Cincinnati, Cincinnati, OH 45221, USA
| | - Jerry Silver
- Department of Neurosciences, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yu Luo
- Department of Molecular Genetics, Biochemistry, and Microbiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45229, USA.
| |
Collapse
|
18
|
Zveik O, Fainstein N, Rechtman A, Haham N, Ganz T, Lavon I, Brill L, Vaknin‐Dembinsky A. Cerebrospinal fluid of progressive multiple sclerosis patients reduces differentiation and immune functions of oligodendrocyte progenitor cells. Glia 2022; 70:1191-1209. [PMID: 35266197 PMCID: PMC9314832 DOI: 10.1002/glia.24165] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 02/20/2022] [Accepted: 02/21/2022] [Indexed: 12/31/2022]
Abstract
Oligodendrocyte progenitor cells (OPCs) are responsible for remyelination in the central nervous system (CNS) in health and disease. For patients with multiple sclerosis (MS), remyelination is not always successful, and the mechanisms differentiating successful from failed remyelination are not well-known. Growing evidence suggests an immune role for OPCs, in addition to their regenerative role; however, it is not clear if this helps or hinders the regenerative process. We studied the effect of cerebrospinal fluid (CSF) from relapsing MS (rMS) and progressive MS (pMS) patients on primary OPC differentiation and immune gene expression and function. We observed that CSF from either rMS or pMS patients has a differential effect on the ability of mice OPCs to differentiate into mature oligodendrocytes and to express immune functions. CSF of pMS patients impaired differentiation into mature oligodendrocytes. In addition, it led to decreased major histocompatibility complex class (MHC)-II expression, tumor necrosis factor (TNF)-α secretion, nuclear factor kappa-B (NFκB) activation, and less activation and proliferation of T cells. Our findings suggest that OPCs are not only responsible for remyelination, but they may also play an active role as innate immune cells in the CNS.
Collapse
Affiliation(s)
- Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Nina Fainstein
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Nitzan Haham
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Iris Lavon
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
- Leslie and Michael Gaffin Center for Neuro‐OncologyHadassah‐Hebrew University Medical CenterJerusalemIsrael
| | - Livnat Brill
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| | - Adi Vaknin‐Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Department of Neurology and Laboratory of NeuroimmunologyThe Agnes‐Ginges Center for Neurogenetics, Hadassah – Hebrew University Medical CenterJerusalemIsrael
| |
Collapse
|
19
|
Yellajoshyula D, Pappas SS, Dauer WT. Oligodendrocyte and Extracellular Matrix Contributions to Central Nervous System Motor Function: Implications for Dystonia. Mov Disord 2022; 37:456-463. [PMID: 34989453 PMCID: PMC11152458 DOI: 10.1002/mds.28892] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 11/17/2021] [Accepted: 12/03/2021] [Indexed: 12/18/2022] Open
Abstract
The quest to elucidate nervous system function and dysfunction in disease has focused largely on neurons and neural circuits. However, fundamental aspects of nervous system development, function, and plasticity are regulated by nonneuronal elements, including glial cells and the extracellular matrix (ECM). The rapid rise of genomics and neuroimaging techniques in recent decades has highlighted neuronal-glial interactions and ECM as a key component of nervous system development, plasticity, and function. Abnormalities of neuronal-glial interactions have been understudied but are increasingly recognized to play a key role in many neurodevelopmental disorders. In this report, we consider the role of myelination and the ECM in the development and function of central nervous system motor circuits and the neurodevelopmental disease dystonia. © 2022 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
| | - Samuel S Pappas
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - William T Dauer
- Peter O'Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
20
|
Kang NU, Lee SJ, Gwak SJ. Fabrication Techniques of Nerve Guidance Conduits for Nerve Regeneration. Yonsei Med J 2022; 63:114-123. [PMID: 35083896 PMCID: PMC8819402 DOI: 10.3349/ymj.2022.63.2.114] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 11/26/2021] [Indexed: 11/27/2022] Open
Abstract
Neuronal loss and axonal degeneration after spinal cord injury or peripheral injury result in the loss of sensory and motor functions. Nerve regeneration is a complicated and medical challenge that requires suitable guides to bridge nerve injury gaps and restore nerve function. Due to the hostility of the microenvironment in the lesion, multiple conditions should be fulfilled to achieve improved functional recovery. Many nerve conduits have been fabricated using various natural and synthetic polymers. The design and material of the nerve guide conduits were carefully reviewed. A detailed review was conducted on the fabrication method of the nerve guide conduit for nerve regeneration. The typical fabrication methods used to fabricate nerve conduits are dip coating, solvent casting, micropatterning, electrospinning, and additive manufacturing. The advantages and disadvantages of the fabrication methods were reported, and research to overcome these limitations was reviewed. Extensive reviews have focused on the biological functions and in vivo performance of polymeric nerve conduits. In this paper, we emphasize the fabrication method of nerve conduits by polymers and their properties. By learning from the existing candidates, we can advance the strategies for designing novel polymeric systems with better properties for nerve regeneration.
Collapse
Affiliation(s)
- Nae-Un Kang
- Department of Mechanical Engineering, College of Engineering, Wonkwang University, Iksan, Korea
| | - Seung-Jae Lee
- Department of Mechanical Design Engineering, College of Engineering, Wonkwang University, Iksan, Korea.
| | - So-Jung Gwak
- Department of Chemical Engineering, College of Engineering, Wonkwang University, Iksan, Korea.
| |
Collapse
|
21
|
Shahsavani N, Alizadeh A, Kataria H, Karimi-Abdolrezaee S. Availability of neuregulin-1beta1 protects neurons in spinal cord injury and against glutamate toxicity through caspase dependent and independent mechanisms. Exp Neurol 2021; 345:113817. [PMID: 34314724 DOI: 10.1016/j.expneurol.2021.113817] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 07/06/2021] [Accepted: 07/20/2021] [Indexed: 12/27/2022]
Abstract
Spinal cord injury (SCI) causes sensorimotor and autonomic impairment that partly reflects extensive, permanent loss of neurons at the epicenter and penumbra of the injury. Strategies aimed at enhancing neuronal protection are critical to attenuate neurodegeneration and improve neurological recovery after SCI. In rat SCI, we previously uncovered that the tissue levels of neuregulin-1beta 1 (Nrg-1β1) are acutely and persistently downregulated in the injured spinal cord. Nrg-1β1 is well-known for its critical roles in the development, maintenance and physiology of neurons and glia in the developing and adult spinal cord. However, despite this pivotal role, Nrg-1β1 specific effects and mechanisms of action on neuronal injury remain largely unknown in SCI. In the present study, using a clinically-relevant model of compressive/contusive SCI in rats and an in vitro model of glutamate toxicity in primary neurons, we demonstrate Nrg-1β1 provides early neuroprotection through attenuation of reactive oxygen species, lipid peroxidation, necrosis and apoptosis in acute and subacute stages of SCI. Mechanistically, availability of Nrg-1β1 following glutamate challenge protects neurons from caspase-dependent and independent cell death that is mediated by modulation of mitochondria associated apoptotic cascades and MAP kinase and AKT signaling pathways. Altogether, our work provides novel insights into the role and mechanisms of Nrg-1β1 in neuronal injury after SCI and introduces its potential as a new neuroprotective target for this debilitating neurological condition.
Collapse
Affiliation(s)
- Narjes Shahsavani
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
22
|
Richard SA, Sackey M. Elucidating the Pivotal Neuroimmunomodulation of Stem Cells in Spinal Cord Injury Repair. Stem Cells Int 2021; 2021:9230866. [PMID: 34341666 PMCID: PMC8325586 DOI: 10.1155/2021/9230866] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2021] [Revised: 07/03/2021] [Accepted: 07/17/2021] [Indexed: 12/11/2022] Open
Abstract
Spinal cord injury (SCI) is a distressing incident with abrupt onset of the motor as well as sensory dysfunction, and most often, the injury occurs as result of high-energy or velocity accidents as well as contact sports and falls in the elderly. The key challenges associated with nerve repair are the lack of self-repair as well as neurotrophic factors and primary and secondary neuronal apoptosis, as well as factors that prevent the regeneration of axons locally. Neurons that survive the initial traumatic damage may be lost due to pathogenic activities like neuroinflammation and apoptosis. Implanted stem cells are capable of differentiating into neural cells that replace injured cells as well as offer local neurotrophic factors that aid neuroprotection, immunomodulation, axonal sprouting, axonal regeneration, and remyelination. At the microenvironment of SCI, stem cells are capable of producing growth factors like brain-derived neurotrophic factor and nerve growth factor which triggers neuronal survival as well as axonal regrowth. Although stem cells have proven to be of therapeutic value in SCI, the major disadvantage of some of the cell types is the risk for tumorigenicity due to the contamination of undifferentiated cells prior to transplantation. Local administration of stem cells via either direct cellular injection into the spinal cord parenchyma or intrathecal administration into the subarachnoid space is currently the best transplantation modality for stem cells during SCI.
Collapse
Affiliation(s)
- Seidu A. Richard
- Department of Medicine, Princefield University, P.O. Box MA128, Ho, Ghana
| | - Marian Sackey
- Department of Pharmacy, Ho Teaching Hospital, P.O. Box MA-374, Ho, Ghana
| |
Collapse
|
23
|
Hart CG, Karimi-Abdolrezaee S. Recent insights on astrocyte mechanisms in CNS homeostasis, pathology, and repair. J Neurosci Res 2021; 99:2427-2462. [PMID: 34259342 DOI: 10.1002/jnr.24922] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/06/2021] [Accepted: 06/24/2021] [Indexed: 12/20/2022]
Abstract
Astrocytes play essential roles in development, homeostasis, injury, and repair of the central nervous system (CNS). Their development is tightly regulated by distinct spatial and temporal cues during embryogenesis and into adulthood throughout the CNS. Astrocytes have several important responsibilities such as regulating blood flow and permeability of the blood-CNS barrier, glucose metabolism and storage, synapse formation and function, and axon myelination. In CNS pathologies, astrocytes also play critical parts in both injury and repair mechanisms. Upon injury, they undergo a robust phenotypic shift known as "reactive astrogliosis," which results in both constructive and deleterious outcomes. Astrocyte activation and migration at the site of injury provides an early defense mechanism to minimize the extent of injury by enveloping the lesion area. However, astrogliosis also contributes to the inhibitory microenvironment of CNS injury and potentiate secondary injury mechanisms, such as inflammation, oxidative stress, and glutamate excitotoxicity, which facilitate neurodegeneration in CNS pathologies. Intriguingly, reactive astrocytes are increasingly a focus in current therapeutic strategies as their activation can be modulated toward a neuroprotective and reparative phenotype. This review will discuss recent advancements in knowledge regarding the development and role of astrocytes in the healthy and pathological CNS. We will also review how astrocytes have been genetically modified to optimize their reparative potential after injury, and how they may be transdifferentiated into neurons and oligodendrocytes to promote repair after CNS injury and neurodegeneration.
Collapse
Affiliation(s)
- Christopher G Hart
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
24
|
Abstract
Spinal cord injury (SCI) destroys the sensorimotor pathway and blocks the information flow between the peripheral nerve and the brain, resulting in autonomic function loss. Numerous studies have explored the effects of obstructed information flow on brain structure and function and proved the extensive plasticity of the brain after SCI. Great progress has also been achieved in therapeutic strategies for SCI to restore the "re-innervation" of the cerebral cortex to the limbs to some extent. Although no thorough research has been conducted, the changes of brain structure and function caused by "re-domination" have been reported. This article is a review of the recent research progress on local structure, functional changes, and circuit reorganization of the cerebral cortex after SCI. Alterations of structure and electrical activity characteristics of brain neurons, features of brain functional reorganization, and regulation of brain functions by reconfigured information flow were also explored. The integration of brain function is the basis for the human body to exercise complex/fine movements and is intricately and widely regulated by information flow. Hence, its changes after SCI and treatments should be considered.
Collapse
Affiliation(s)
- Can Zhao
- Institute of Rehabilitation Engineering, China Rehabilitation Science Institute, Beijing, China
- School of Rehabilitation, Capital Medical University, Beijing, China
| | - Shu-Sheng Bao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Meng Xu
- Department of Orthopedics, The First Medical Center of PLA General Hospital, Beijing, China
| | - Jia-Sheng Rao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
25
|
Yao M, Fang J, Tao W, Feng G, Wei M, Gao Y, Xin W, Li Y, Du S. Modulation of proteoglycan receptor regulates RhoA/CRMP2 pathways and promotes axonal myelination. Neurosci Lett 2021; 760:136079. [PMID: 34166723 DOI: 10.1016/j.neulet.2021.136079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/08/2021] [Accepted: 06/18/2021] [Indexed: 11/15/2022]
Abstract
The function of the myelinating system is important because a defective myelin sheath results in various nervous disorders, including multiple sclerosis and peripheral neuropathies. The dorsal root entry zone (DREZ) is a transitional area between the central nervous system (CNS) and the peripheral nervous system (PNS) that is generated by two types of cells-oligodendrocytes and Schwann cells (SCs). It is well known that after injury the extracellular matrix, including the CSPG, impairs axonal myelination by activating protein tyrosine phosphatase-σ (PTPσ) in both cells. The Intracellular Sigma Peptide (ISP) is memetic of the PTPσ wedge region. It competitively binds to PTPσ and regulates the downstream signaling of RhoA. In the present study, we aimed to investigate whether the ISP increased myelination in vivo and in vitro. The in vitro assay was meant to further verify the in vivo mechanisms. We observed that ISP administration could increase axonal myelination both in vivo and in vitro. Furthermore, we provide evidence that, in oligodendrocytes and Schwann cells, the myelination-induced effects of ISP application entail an inverse expression of the RhoA/CRMP2 signaling pathway. Overall, our results indicate that the ISP modulation of PTPσ enhances axonal myelination via the RhoA/CRMP2 signaling pathways.
Collapse
Affiliation(s)
- Min Yao
- School of Pharmaceutical Sciences, Health Science Centre, Shenzhen University, Shenzhen 518060, China; Department of Surgery, The University of Hong Kong, Hong Kong SAR 999077, China; School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Jie Fang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Wei Tao
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen 518055, China
| | - Gang Feng
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen 518055, China
| | - Mingyi Wei
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen 518055, China
| | - Yuhao Gao
- Department of Neuroscience and Behavioral Biology, Emory College of Arts and Sciences, Emory University, Atlanta, GA 30322, USA
| | - Wen Xin
- Beijing TransGen Biotech Co., Ltd, Beijing 100192, China
| | - Yu Li
- Department of Neurosurgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China.
| | - Shiwei Du
- Department of Neurosurgery, Shenzhen University General Hospital, Shenzhen University Clinical Medical Academy, Shenzhen 518055, China.
| |
Collapse
|
26
|
Melrose J, Hayes AJ, Bix G. The CNS/PNS Extracellular Matrix Provides Instructive Guidance Cues to Neural Cells and Neuroregulatory Proteins in Neural Development and Repair. Int J Mol Sci 2021; 22:5583. [PMID: 34070424 PMCID: PMC8197505 DOI: 10.3390/ijms22115583] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 05/11/2021] [Accepted: 05/17/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The extracellular matrix of the PNS/CNS is unusual in that it is dominated by glycosaminoglycans, especially hyaluronan, whose space filling and hydrating properties make essential contributions to the functional properties of this tissue. Hyaluronan has a relatively simple structure but its space-filling properties ensure micro-compartments are maintained in the brain ultrastructure, ensuring ionic niches and gradients are maintained for optimal cellular function. Hyaluronan has cell-instructive, anti-inflammatory properties and forms macro-molecular aggregates with the lectican CS-proteoglycans, forming dense protective perineuronal net structures that provide neural and synaptic plasticity and support cognitive learning. AIMS To highlight the central nervous system/peripheral nervous system (CNS/PNS) and its diverse extracellular and cell-associated proteoglycans that have cell-instructive properties regulating neural repair processes and functional recovery through interactions with cell adhesive molecules, receptors and neuroregulatory proteins. Despite a general lack of stabilising fibrillar collagenous and elastic structures in the CNS/PNS, a sophisticated dynamic extracellular matrix is nevertheless important in tissue form and function. CONCLUSIONS This review provides examples of the sophistication of the CNS/PNS extracellular matrix, showing how it maintains homeostasis and regulates neural repair and regeneration.
Collapse
Affiliation(s)
- James Melrose
- Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
- Sydney Medical School, Northern, The University of Sydney, Sydney, NSW 2052, Australia
- Faculty of Medicine and Health, The University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
| | - Anthony J. Hayes
- Bioimaging Research Hub, Cardiff School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK;
| | - Gregory Bix
- Clinical Neuroscience Research Center, Departments of Neurosurgery and Neurology, Tulane University School of Medicine, New Orleans, LA 70112, USA;
| |
Collapse
|
27
|
Termini CM, Pang A, Batton DM, Chute JP. Proteoglycans regulate protein tyrosine phosphatase receptor σ organization on hematopoietic stem/progenitor cells. Exp Hematol 2021; 96:44-51. [PMID: 33515635 PMCID: PMC10838547 DOI: 10.1016/j.exphem.2021.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 01/07/2021] [Accepted: 01/22/2021] [Indexed: 01/08/2023]
Abstract
Protein tyrosine phosphatase receptor σ (PTPσ) is highly expressed by murine and human hematopoietic stem cells (HSCs) and negatively regulates HSC self-renewal and regeneration. Previous studies of the nervous system suggest that heparan sulfate proteoglycans can inactivate PTPσ by clustering PTPσ receptors on neurons, but this finding has yet to be visually verified with adequate resolution. Here, we sought to visualize and quantify how heparan sulfate proteoglycans regulate the organization and activation of PTPσ in hematopoietic stem/progenitor cells (HSPCs). Our study illustrates that syndecan-2 promotes PTPσ clustering, which sustains phospho-tyrosine and phospho-ezrin levels in association with augmentation of hematopoietic colony formation. Strategies that promote clustering of PTPσ on HSPCs may serve to powerfully augment hematopoietic function.
Collapse
Affiliation(s)
- Christina M Termini
- Division of Hematology/Oncology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA; Department of Orthopaedic Surgery, University of California at Los Angeles, Los Angeles, CA; Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Amara Pang
- Division of Hematology/Oncology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA; Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA
| | - Destiny M Batton
- Division of Hematology/Oncology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA
| | - John P Chute
- Division of Hematology/Oncology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA; Department of Orthopaedic Surgery, University of California at Los Angeles, Los Angeles, CA; Department of Medicine, Cedars Sinai Medical Center, Los Angeles, CA; Broad Stem Cell Research Center, University of California at Los Angeles, Los Angeles, CA; Jonsson Comprehensive Cancer Center, University of California at Los Angeles, Los Angeles, CA.
| |
Collapse
|
28
|
Li F, Song X, Xu J, Shi Y, Hu R, Ren Z, Qi Q, Lü H, Cheng X, Hu J. Morroniside protects OLN-93 cells against H 2O 2-induced injury through the PI3K/Akt pathway-mediated antioxidative stress and antiapoptotic activities. Cell Cycle 2021; 20:661-675. [PMID: 33734020 DOI: 10.1080/15384101.2021.1889186] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Neurodegenerative disorders, including spinal cord injury (SCI), result in oxidative stress-induced cell damage. Morroniside (MR), a major active ingredient of the Chinese herb Shan Zhu Yu, has been shown to ameliorate oxidative stress and inflammatory response. Our previous study also confirmed that morroniside protects SK-N-SH cell line (human neuroblastoma cells) against oxidative impairment. However, it remains unclear whether MR also plays a protective role for oligodendrocytes that are damaged following SCI. The present study investigated the protective effects of MR against hydrogen peroxide (H2O2)-induced cell death in OLN-93 cells. MR protected OLN-93 cells from H2O2-induced injury, attenuated H2O2-induced increase in reactive oxygen species (ROS) and malondialdehyde (MDA) levels, and blocked the reduction of mitochondrial membrane potential (MMP) induced by H2O2. MR enhanced the activity of the antioxidant enzyme superoxide dismutase (SOD) and suppressed H2O2-induced downregulation of the antiapoptotic protein Bcl-2 and activation of the proapoptotic protein caspase-3. Finally, we found that LY294002, a specific inhibitor of the PI3K/Akt pathway, inhibited the protective effect of MR against H2O2-induced OLN-93 cell injury in the MTT and TUNEL assays. LY294002 also inhibited the expression of SOD and Bcl-2, and increased the expression of iNOS and c-caspase-3 induced by MR treatment. MR exerts protective effects against H2O2-induced OLN-93 cell injury through the PI3K/Akt signaling pathway-mediated antioxidative stress and antiapoptotic activities. MR may provide a potential strategy for SCI treatment or other related neurodegeneration.
Collapse
Affiliation(s)
- Fengzhi Li
- Department of Cell Biology College of Basic Medical Sciences, Dalian Medical University, Dalian, P.R. China
| | - Xue Song
- Department of Central Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China
| | - Jiaxin Xu
- Department of Cell Biology College of Basic Medical Sciences, Dalian Medical University, Dalian, P.R. China
| | - Yujiao Shi
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Ruina Hu
- Department of Cell Biology College of Basic Medical Sciences, Dalian Medical University, Dalian, P.R. China
| | - Zhen Ren
- Department of Cell Biology College of Basic Medical Sciences, Dalian Medical University, Dalian, P.R. China
| | - Qi Qi
- Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Hezuo Lü
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| | - Xiaoxin Cheng
- Department of Cell Biology College of Basic Medical Sciences, Dalian Medical University, Dalian, P.R. China
| | - Jianguo Hu
- Department of Clinical Laboratory, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China.,Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China
| |
Collapse
|
29
|
Shahsavani N, Kataria H, Karimi-Abdolrezaee S. Mechanisms and repair strategies for white matter degeneration in CNS injury and diseases. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166117. [PMID: 33667627 DOI: 10.1016/j.bbadis.2021.166117] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 02/22/2021] [Accepted: 02/23/2021] [Indexed: 12/14/2022]
Abstract
White matter degeneration is an important pathophysiological event of the central nervous system that is collectively characterized by demyelination, oligodendrocyte loss, axonal degeneration and parenchymal changes that can result in sensory, motor, autonomic and cognitive impairments. White matter degeneration can occur due to a variety of causes including trauma, neurotoxic exposure, insufficient blood flow, neuroinflammation, and developmental and inherited neuropathies. Regardless of the etiology, the degeneration processes share similar pathologic features. In recent years, a plethora of cellular and molecular mechanisms have been identified for axon and oligodendrocyte degeneration including oxidative damage, calcium overload, neuroinflammatory events, activation of proteases, depletion of adenosine triphosphate and energy supply. Extensive efforts have been also made to develop neuroprotective and neuroregenerative approaches for white matter repair. However, less progress has been achieved in this area mainly due to the complexity and multifactorial nature of the degeneration processes. Here, we will provide a timely review on the current understanding of the cellular and molecular mechanisms of white matter degeneration and will also discuss recent pharmacological and cellular therapeutic approaches for white matter protection as well as axonal regeneration, oligodendrogenesis and remyelination.
Collapse
Affiliation(s)
- Narjes Shahsavani
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
30
|
Jeong HJ, Yun Y, Lee SJ, Ha Y, Gwak SJ. Biomaterials and strategies for repairing spinal cord lesions. Neurochem Int 2021; 144:104973. [PMID: 33497713 DOI: 10.1016/j.neuint.2021.104973] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 01/17/2021] [Accepted: 01/18/2021] [Indexed: 01/13/2023]
Abstract
Spinal cord injury (SCI) causes intractable disease and leads to inevitable physical, financial, and psychological burdens on patients and their families. SCI is commonly divided into primary and secondary injury. Primary injury occurs upon direct impact to the spinal cord, which leads to cell necrosis, axon disruption, and vascular loss. This triggers pathophysiological secondary injury, which has several phases: acute, subacute, intermediate, and chronic. These phases are dependent on post-injury time and pathophysiology and have various causes, such as the infiltration of inflammatory cells and release of cytokines that can act as a barrier to neural regeneration. Another unique feature of SCI is the glial scar produced from the reactive proliferation of astrocytes, which acts as a barrier to axonal regeneration. Interdisciplinary research is investigating the use of biomaterials and tissue-engineered fabrication to overcome SCI. In this review, we discuss representative biomaterials, including natural and synthetic polymers and nanomaterials. In addition, we describe several strategies to repair spinal cord injuries, such as fabrication and the delivery of therapeutic biocomponents. These biomaterials and strategies may offer beneficial information to enhance the repair of spinal cord lesions.
Collapse
Affiliation(s)
- Hun-Jin Jeong
- Department of Mechanical Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
| | - Yeomin Yun
- Department of Neurosurgery, Spine and Spinal Cord Institute, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemoon-gu, Seoul, Republic of Korea
| | - Seung-Jae Lee
- Department of Mechanical Engineering, Wonkwang University, 54538, Iksan, Republic of Korea; Department of Mechanical and Design Engineering, Wonkwang University, 54538, Iksan, Republic of Korea
| | - Yoon Ha
- Department of Neurosurgery, Spine and Spinal Cord Institute, Yonsei University College of Medicine, 134 Shinchon-dong, Seodaemoon-gu, Seoul, Republic of Korea; POSTECH Biotech Center, Pohang University of Science and Technology, San 31, Pohang, Gyeongbuk, Republic of Korea
| | - So-Jung Gwak
- Department of Chemical Engineering, Wonkwang University, 54538, Iksan, Republic of Korea.
| |
Collapse
|
31
|
Zhang Z, Zhou H, Zhou J. Heterogeneity and Proliferative and Differential Regulators of NG2-glia in Physiological and Pathological States. Curr Med Chem 2021; 27:6384-6406. [PMID: 31333083 DOI: 10.2174/0929867326666190717112944] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 06/12/2019] [Accepted: 06/20/2019] [Indexed: 12/16/2022]
Abstract
NG2-glia, also called Oligodendrocyte Precursor Cells (OPCs), account for approximately 5%-10% of the cells in the developing and adult brain and constitute the fifth major cell population in the central nervous system. NG2-glia express receptors and ion channels involved in rapid modulation of neuronal activities and signaling with neuronal synapses, which have functional significance in both physiological and pathological states. NG2-glia participate in quick signaling with peripheral neurons via direct synaptic touches in the developing and mature central nervous system. These distinctive glia perform the unique function of proliferating and differentiating into oligodendrocytes in the early developing brain, which is critical for axon myelin formation. In response to injury, NG2-glia can proliferate, migrate to the lesions, and differentiate into oligodendrocytes to form new myelin sheaths, which wrap around damaged axons and result in functional recovery. The capacity of NG2-glia to regulate their behavior and dynamics in response to neuronal activity and disease indicate their critical role in myelin preservation and remodeling in the physiological state and in repair in the pathological state. In this review, we provide a detailed summary of the characteristics of NG2-glia, including their heterogeneity, the regulators of their proliferation, and the modulators of their differentiation into oligodendrocytes.
Collapse
Affiliation(s)
- Zuo Zhang
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Hongli Zhou
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| | - Jiyin Zhou
- National Drug Clinical Trial Institution, the Second Affiliated Hospital, Army Medical University, Chongqing 400037, China
| |
Collapse
|
32
|
Edwar-Mickael M, Kubick N. CD4 + Tregs may be essential for solving astrocyte glial scar deadlock. Neural Regen Res 2021; 16:2563. [PMID: 33907049 PMCID: PMC8374566 DOI: 10.4103/1673-5374.313069] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Affiliation(s)
- Michel Edwar-Mickael
- Department of Experimental Genomics, Institute of Animal Biotechnology and Genetics, Polish Academy of Science, Jastrzebiec, Poland; Department of Immunology, PM Forskningscentreum, Ekerö Stockholm, Sweden
| | - Norwin Kubick
- Institute of Biochemistry, Molecular Cell Biology, University Clinic Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
33
|
Cheng L, Sami A, Ghosh B, Urban MW, Heinsinger NM, Liang SS, Smith GM, Wright MC, Li S, Lepore AC. LAR inhibitory peptide promotes recovery of diaphragm function and multiple forms of respiratory neural circuit plasticity after cervical spinal cord injury. Neurobiol Dis 2020; 147:105153. [PMID: 33127470 PMCID: PMC7726004 DOI: 10.1016/j.nbd.2020.105153] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Revised: 09/14/2020] [Accepted: 10/25/2020] [Indexed: 12/18/2022] Open
Abstract
Chondroitin sulfate proteoglycans (CSPGs), up-regulated in and around the lesion after traumatic spinal cord injury (SCI), are key extracellular matrix inhibitory molecules that limit axon growth and consequent recovery of function. CSPG-mediated inhibition occurs via interactions with axonal receptors, including leukocyte common antigen- related (LAR) phosphatase. We tested the effects of a novel LAR inhibitory peptide in rats after hemisection at cervical level 2, a SCI model in which bulbospinal inspiratory neural circuitry originating in the medullary rostral ventral respiratory group (rVRG) becomes disconnected from phrenic motor neuron (PhMN) targets in cervical spinal cord, resulting in persistent partial-to-complete diaphragm paralysis. LAR peptide was delivered by a soaked gelfoam, which was placed directly over the injury site immediately after C2 hemisection and replaced at 1 week post-injury. Axotomized rVRG axons originating in ipsilateral medulla or spared rVRG fibers originating in contralateral medulla were separately assessed by anterograde tracing via AAV2-mCherry injection into rVRG. At 8 weeks post-hemisection, LAR peptide significantly improved ipsilateral hemidiaphragm function, as assessed in vivo with electromyography recordings. LAR peptide promoted robust regeneration of ipsilateral-originating rVRG axons into and through the lesion site and into intact caudal spinal cord to reach PhMNs located at C3-C5 levels. Furthermore, regenerating rVRG axons re-established putative monosynaptic connections with their PhMNs targets. In addition, LAR peptide stimulated robust sprouting of both modulatory serotonergic axons and contralateral-originating rVRG fibers within the PhMN pool ipsilateral/ caudal to the hemisection. Our study demonstrates that targeting LAR-based axon growth inhibition promotes multiple forms of respiratory neural circuit plasticity and provides a new peptide-based therapeutic strategy to ameliorate the devastating respiratory consequences of SCI.
Collapse
Affiliation(s)
- Lan Cheng
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Armin Sami
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Biswarup Ghosh
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Mark W Urban
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Nicolette M Heinsinger
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - Sophia S Liang
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, United States of America
| | - George M Smith
- Department of Neuroscience, Shriners Hospitals for Pediatric Research Center, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140-5104, United States of America
| | - Megan C Wright
- Department of Biology, Arcadia University, Glenside, PA 19038, United States of America
| | - Shuxin Li
- Department of Anatomy and Cell Biology, Shriners Hospitals for Pediatric Research Center, Temple University School of Medicine, 3500 North Broad Street, Philadelphia, PA 19140-5104, United States of America
| | - Angelo C Lepore
- Department of Neuroscience, Vickie and Jack Farber Institute for Neuroscience, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, PA 19107, United States of America.
| |
Collapse
|
34
|
Hart CG, Karimi-Abdolrezaee S. Bone morphogenetic proteins: New insights into their roles and mechanisms in CNS development, pathology and repair. Exp Neurol 2020; 334:113455. [PMID: 32877654 DOI: 10.1016/j.expneurol.2020.113455] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 08/18/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023]
Abstract
Bone morphogenetic proteins (BMPs) are a highly conserved and diverse family of proteins that play essential roles in various stages of development including the formation and patterning of the central nervous system (CNS). Bioavailability and function of BMPs are regulated by input from a plethora of transcription factors and signaling pathways. Intriguingly, recent literature has uncovered novel roles for BMPs in regulating homeostatic and pathological responses in the adult CNS. Basal levels of BMP ligands and receptors are widely expressed in the adult brain and spinal cord with differential expression patterns across CNS regions, cell types and subcellular locations. Recent evidence indicates that several BMP isoforms are transiently or chronically upregulated in the aged or pathological CNS. Genetic knockout and pharmacological studies have elucidated that BMPs regulate several aspects of CNS injury and repair including cell survival and differentiation, reactive astrogliosis and glial scar formation, axon regeneration, and myelin preservation and repair. Several BMP isoforms can be upregulated in the injured or diseased CNS simultaneously yet exert complementary or opposing effects on the endogenous cell responses after injury. Emerging studies also show that dysregulation of BMPs is associated with various CNS pathologies. Interestingly, modulation of BMPs can lead to beneficial or detrimental effects on CNS injury and repair mechanisms in a ligand, temporally or spatially specific manner, which reflect the complexity of BMP signaling. Given the significance of BMPs in neurodevelopment, a better understanding of their role in the context of injury may provide new therapeutic targets for the pathologic CNS. This review will provide a timely overview on the foundation and recent advancements in knowledge regarding the role and mechanisms of BMP signaling in the developing and adult CNS, and their implications in pathological responses and repair processes after injury or diseases.
Collapse
Affiliation(s)
- Christopher G Hart
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Children's Hospital Research Institute of Manitoba, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
35
|
Yang T, Dai Y, Chen G, Cui S. Dissecting the Dual Role of the Glial Scar and Scar-Forming Astrocytes in Spinal Cord Injury. Front Cell Neurosci 2020; 14:78. [PMID: 32317938 PMCID: PMC7147295 DOI: 10.3389/fncel.2020.00078] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 03/18/2020] [Indexed: 12/19/2022] Open
Abstract
Recovery from spinal cord injury (SCI) remains an unsolved problem. As a major component of the SCI lesion, the glial scar is primarily composed of scar-forming astrocytes and plays a crucial role in spinal cord regeneration. In recent years, it has become increasingly accepted that the glial scar plays a dual role in SCI recovery. However, the underlying mechanisms of this dual role are complex and need further clarification. This dual role also makes it difficult to manipulate the glial scar for therapeutic purposes. Here, we briefly discuss glial scar formation and some representative components associated with scar-forming astrocytes. Then, we analyze the dual role of the glial scar in a dynamic perspective with special attention to scar-forming astrocytes to explore the underlying mechanisms of this dual role. Finally, taking the dual role of the glial scar into account, we provide several pieces of advice on novel therapeutic strategies targeting the glial scar and scar-forming astrocytes.
Collapse
Affiliation(s)
- Tuo Yang
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.,Medical School of Nantong University, Nantong, China
| | - YuJuan Dai
- Medical School of Nantong University, Nantong, China
| | - Gang Chen
- Department of Tissue and Embryology, Medical School of Nantong University, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, China.,Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - ShuSen Cui
- Department of Hand Surgery, China-Japan Union Hospital of Jilin University, Changchun, China
| |
Collapse
|
36
|
Rodemer W, Zhang G, Sinitsa I, Hu J, Jin LQ, Li S, Selzer ME. PTPσ Knockdown in Lampreys Impairs Reticulospinal Axon Regeneration and Neuronal Survival After Spinal Cord Injury. Front Cell Neurosci 2020; 14:61. [PMID: 32265663 PMCID: PMC7096546 DOI: 10.3389/fncel.2020.00061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/02/2020] [Indexed: 01/10/2023] Open
Abstract
Traumatic spinal cord injury (SCI) results in persistent functional deficits due to the lack of axon regeneration within the mammalian CNS. After SCI, chondroitin sulfate proteoglycans (CSPGs) inhibit axon regrowth via putative interactions with the LAR-family protein tyrosine phosphatases, PTPσ and LAR, localized on the injured axon tips. Unlike mammals, the sea lamprey, Petromyzon marinus, robustly recovers locomotion after complete spinal cord transection (TX). Behavioral recovery is accompanied by heterogeneous yet predictable anatomical regeneration of the lamprey's reticulospinal (RS) system. The identified RS neurons can be categorized as "good" or "bad" regenerators based on the likelihood that their axons will regenerate. Those neurons that fail to regenerate their axons undergo a delayed form of caspase-mediated cell death. Previously, this lab reported that lamprey PTPσ mRNA is selectively expressed in "bad regenerator" RS neurons, preceding SCI-induced caspase activation. Consequently, we hypothesized that PTPσ deletion would reduce retrograde cell death and promote axon regeneration. Using antisense morpholino oligomers (MOs), we knocked down PTPσ expression after TX and assessed the effects on axon regeneration, caspase activation, intracellular signaling, and behavioral recovery. Unexpectedly, PTPσ knockdown significantly impaired RS axon regeneration at 10 weeks post-TX, primarily due to reduced long-term neuron survival. Interestingly, cell loss was not preceded by an increase in caspase or p53 activation. Behavioral recovery was largely unaffected, although PTPσ knockdowns showed mild deficits in the recovery of swimming distance and latency to immobility during open field swim assays. Although the mechanism underlying the cell death following TX and PTPσ knockdown remains unknown, this study suggests that PTPσ is not a net negative regulator of long tract axon regeneration in lampreys.
Collapse
Affiliation(s)
- William Rodemer
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Guixin Zhang
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Isabelle Sinitsa
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- College of Science and Technology, Temple University, Philadelphia, PA, United States
| | - Jianli Hu
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Li-qing Jin
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Shuxin Li
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Michael E. Selzer
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
- Department of Neurology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
37
|
Feliu A, Mestre L, Carrillo-Salinas FJ, Yong VW, Mecha M, Guaza C. 2-arachidonoylglycerol reduces chondroitin sulphate proteoglycan production by astrocytes and enhances oligodendrocyte differentiation under inhibitory conditions. Glia 2020; 68:1255-1273. [PMID: 31894889 DOI: 10.1002/glia.23775] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 11/22/2019] [Accepted: 12/17/2019] [Indexed: 01/21/2023]
Abstract
The failure to remyelinate and regenerate is a critical impediment to recovery in multiple sclerosis (MS), resulting in severe dysfunction and disability. The chondroitin sulfate proteoglycans (CSPGs) that accumulate in MS lesions are thought to be linked to the failure to regenerate, impeding oligodendrocyte precursor cell (OPC) differentiation and neuronal growth. The potential of endocannabinoids to influence MS progression may reflect their capacity to enhance repair processes. Here, we investigated how 2-arachidonoylglycerol (2-AG) may affect the production of the CSPGs neurocan and brevican by astrocytes in culture. In addition, we studied whether 2-AG promotes oligodendrocyte differentiation under inhibitory conditions in vitro. Following treatment with 2-AG or by enhancing its endogenous tone through the use of inhibitors of its hydrolytic enzymes, CSPG production by rat and human TGF-β1 stimulated astrocytes was reduced. These effects of 2-AG might reflect its influence on TGF-β1/SMAD pathway, signaling that is involved in CSPG upregulation. The matrix generated from 2-AG-treated astrocytes is less inhibitory to oligodendrocyte differentiation and significantly, 2-AG administration directly promotes the differentiation of rat and human oligodendrocytes cultured under inhibitory conditions. Overall, the data obtained favor targeting the endocannabinoid system to neutralize CSPG accumulation and to enhance oligodendrocyte differentiation.
Collapse
Affiliation(s)
- Ana Feliu
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Instituto Cajal, CSIC, Madrid, Spain
| | - Leyre Mestre
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Instituto Cajal, CSIC, Madrid, Spain
| | | | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences and Oncology, University of Calgary, Calgary, Alberta, Canada
| | - Miriam Mecha
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Instituto Cajal, CSIC, Madrid, Spain
| | - Carmen Guaza
- Functional and Systems Neurobiology Department, Neuroimmunology Group, Instituto Cajal, CSIC, Madrid, Spain
| |
Collapse
|
38
|
McCullough BS, Batsomboon P, Hutchinson KB, Dudley GB, Barrios AM. Synthesis and PTP Inhibitory Activity of Illudalic Acid and Its Methyl Ether, with Insights into Selectivity for LAR PTP over Other Tyrosine Phosphatases under Physiologically Relevant Conditions. JOURNAL OF NATURAL PRODUCTS 2019; 82:3386-3393. [PMID: 31809044 DOI: 10.1021/acs.jnatprod.9b00663] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The protein tyrosine phosphatase (PTP) family of enzymes includes many attractive therapeutic targets, such as those in the leukocyte common antigen-related (LAR) subfamily of receptor PTPs. Synthesis and PTP inhibitory activity of illudalic acid and its methyl ether are described, with a focus on selective inhibition of LAR PTP relative to a small collection of other representative PTPs. The synthesis comprises 16 steps and provides illudalic acid in up to 12% overall yield from neopentylene-fused benzoate 1 (20 steps from commercial materials). Illudalic acid dose-dependently (measured IC50 = 2.1 ± 0.2 μM) and time-dependently inhibits LAR consistent with previous reports of covalent binding. The kinetics of LAR inhibition by illudalic acid are consistent with a two-step mechanism in which the inhibitor and enzyme first interact noncovalently (KI = 130 ± 50 μM), followed by covalent ligation at a rate kinact = 1.3 ± 0.4 min-1. The kinact/KI ratio of 104 corresponds to a t∞1/2 of 0.5 min, as discussed herein. The phenol methyl ether of illudalic acid was found to be less potent in our dose-response assays (measured IC50 = 55 ± 6 μM) but more selective for LAR, with a weaker initial noncovalent interaction and faster covalent ligation of LAR as compared to illudalic acid itself. A truncated analogue of illudalic acid that lacks the neopentylene ring fusion was found to be devoid of significant activity under our assay conditions, in contrast to previous reports. These observations collectively help inform further development of illudalic acid analogues as potent and selective inhibitors of the LAR subfamily of tyrosine phosphatases.
Collapse
Affiliation(s)
- Brandon S McCullough
- Department of Medicinal Chemistry , University of Utah , Salt Lake City , Utah 84112 , United States
| | - Paratchata Batsomboon
- C. Eugene Bennett Department of Chemistry , West Virginia University , Morgantown , West Virginia 26506 , United States
| | - Kacey B Hutchinson
- Department of Medicinal Chemistry , University of Utah , Salt Lake City , Utah 84112 , United States
| | - Gregory B Dudley
- C. Eugene Bennett Department of Chemistry , West Virginia University , Morgantown , West Virginia 26506 , United States
| | - Amy M Barrios
- Department of Medicinal Chemistry , University of Utah , Salt Lake City , Utah 84112 , United States
| |
Collapse
|
39
|
Hart CG, Dyck SM, Kataria H, Alizadeh A, Nagakannan P, Thliveris JA, Eftekharpour E, Karimi-Abdolrezaee S. Acute upregulation of bone morphogenetic protein-4 regulates endogenous cell response and promotes cell death in spinal cord injury. Exp Neurol 2019; 325:113163. [PMID: 31881217 DOI: 10.1016/j.expneurol.2019.113163] [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: 08/27/2019] [Revised: 12/10/2019] [Accepted: 12/24/2019] [Indexed: 01/11/2023]
Abstract
Traumatic spinal cord injury (SCI) elicits a cascade of secondary injury mechanisms that induce profound changes in glia and neurons resulting in their activation, injury or cell death. The resultant imbalanced microenvironment of acute SCI also negatively impacts regenerative processes in the injured spinal cord. Thus, it is imperative to uncover endogenous mechanisms that drive these acute injury events. Here, we demonstrate that the active form of bone morphogenetic protein-4 (BMP4) is robustly and transiently upregulated in acute SCI in rats. BMP4 is a key morphogen in neurodevelopment; however, its role in SCI is not fully defined. Thus, we elucidated the ramification of BMP4 upregulation in a preclinical model of compressive/contusive SCI in the rat by employing noggin, an endogenous antagonist of BMP ligands, and LDN193189, an intracellular inhibitor of BMP signaling. In parallel, we studied cell-specific effects of BMP4 on neural precursor cells (NPCs), oligodendrocyte precursor cells (OPCs), neurons and astrocytes in vitro. We demonstrate that activation of BMP4 inhibits differentiation of spinal cord NPCs and OPCs into mature myelin-expressing oligodendrocytes, and acute blockade of BMPs promotes oligodendrogenesis, oligodendrocyte preservation and remyelination after SCI. Importantly, we report for the first time that BMP4 directly induces caspase-3 mediated apoptosis in neurons and oligodendrocytes in vitro, and noggin and LDN193189 remarkably attenuate caspase-3 activation and lipid peroxidation in acute SCI. BMP4 also enhances the production of inhibitory chondroitin sulfate proteoglycans (CSPGs) in activated astrocytes in vitro and after SCI. Interestingly, our work reveals that despite the beneficial effects of BMP inhibition in acute SCI, neither noggin nor LDN193189 treatment resulted in long-term functional recovery. Collectively, our findings suggest a role for BMP4 in regulating acute secondary injury mechanisms following SCI, and a potential target for combinatorial approaches to improve endogenous cell response and remyelination.
Collapse
Affiliation(s)
- Christopher G Hart
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Scott M Dyck
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Pandian Nagakannan
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - James A Thliveris
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Eftekhar Eftekharpour
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada; Children's Hospital Research Institute of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
40
|
Zhou L, Ma YC, Tang X, Li WY, Ma Y, Wang RL. Identification of the potential dual inhibitor of protein tyrosine phosphatase sigma and leukocyte common antigen-related phosphatase by virtual screen, molecular dynamic simulations and post-analysis. J Biomol Struct Dyn 2019; 39:45-62. [PMID: 31842717 DOI: 10.1080/07391102.2019.1705913] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Owing to their inhibitory role in regulating oligodendrocyte differentiation and apoptosis, protein tyrosine phosphatase sigma (PTPσ) and leukocyte common antigen-related phosphatase (LAR) play a crucial potential role in treating spinal cord injury (SCI) disease. In this research, the computer aided drug design (CADD) methods were applied to discover the potential dual-target drug involving virtual screen, molecular docking and molecular dynamic simulation. Initially, the top 20 compounds with higher docking score than the positive controls (ZINC13749892, ZINC14516161) were virtually screened out from NCI and ZINC databases, and then were submitted in ADMET to predict their drug properties. Among these potential compounds, ZINC72417086 showed a higher docking score and satisfied Lipinski's rule of five. In addition, the post-analysis demonstrated that when ZINC72417086 bound to PTPσ and LAR, it could stable proteins conformations and destroy the residues interactions between P-loop and other loop regions in active pocket. Meanwhile, residue ARG1595 and ARG1528 could play a crucial role in in the inhibition of PTPσ and LAR, respectively. This research offered a novel approach for rapid discovery of dual-target leads compounds to treat SCI.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Liang Zhou
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Yang-Chun Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Xue Tang
- Tasly Research Institute, Tasly Holding Group Co., Ltd, Tianjin, China
| | - Wei-Ya Li
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Ying Ma
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| | - Run-Ling Wang
- Tianjin Key Laboratory on Technologies Enabling Development of Clinical Therapeutics and Diagnostics (Theranostics), School of Pharmacy, Tianjin Medical University, Tianjin, China
| |
Collapse
|
41
|
Shi G, Zhou X, Wang X, Zhang X, Zhang P, Feng S. Signatures of altered DNA methylation gene expression after central and peripheral nerve injury. J Cell Physiol 2019; 235:5171-5181. [PMID: 31691285 DOI: 10.1002/jcp.29393] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 10/07/2019] [Indexed: 01/09/2023]
Abstract
Nerve damage can lead to movement and sensory dysfunction, with high morbidity and disability rates causing severe burdens on patients, families, and society. DNA methylation is a kind of epigenetics, and a great number of previous studies have demonstrated that DNA methylation plays an important role in the process of nerve regeneration and remodeling. However, compared with the central nervous system, the peripheral nervous system shows stronger recovery after injury, which is related to the complex microenvironment and epigenetic changes occurring at the site of injury. Therefore, what common epigenetic changes between the central and peripheral nervous systems remain to be elucidated. We first screened differential methylation genes after spinal cord injury and sciatic nerve injury using whole-genome bisulfite sequencing and methylated DNA immunoprecipitation sequencing, respectively. Subsequently, a total of 16 genes had the same epigenetic changes after spinal cord injury and sciatic nerve injury. The Gene Ontology analysis and Kyoto Encyclopedia of Genes and Genomes enrichment analysis were performed to identify the critical biological processes and pathways. Furthermore, a protein-protein interaction network analysis indicated that Dnm3, Ntrk3, Smurf1, Dpysl2, Kalrn, Shank1, Dlg2, Arsb, Reln, Bmp5, Numbl, Prickle2, Map6, and Htr7 were the core genes. These outcomes may provide novel insights into the molecular mechanism of the subacute phase of nerve injury. These verified genes can offer potential diagnostic and therapeutic targets for nerve injury.
Collapse
Affiliation(s)
- Guidong Shi
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China.,Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xianhu Zhou
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xu Wang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaolei Zhang
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.,Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin, China
| | - Shiqing Feng
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China.,International Science and Technology Cooperation Base of Spinal Cord Injury, Tianjin Key Laboratory of Spine and Spinal Cord Injury, Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
42
|
Beyer F, Jadasz J, Samper Agrelo I, Schira-Heinen J, Groh J, Manousi A, Bütermann C, Estrada V, Reiche L, Cantone M, Vera J, Viganò F, Dimou L, Müller HW, Hartung HP, Küry P. Heterogeneous fate choice of genetically modulated adult neural stem cells in gray and white matter of the central nervous system. Glia 2019; 68:393-406. [PMID: 31633850 DOI: 10.1002/glia.23724] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/16/2022]
Abstract
Apart from dedicated oligodendroglial progenitor cells, adult neural stem cells (aNSCs) can also give rise to new oligodendrocytes in the adult central nervous system (CNS). This process mainly confers myelinating glial cell replacement in pathological situations and can hence contribute to glial heterogeneity. Our previous studies demonstrated that the p57kip2 gene encodes an intrinsic regulator of glial fate acquisition and we here investigated to what degree its modulation can affect stem cell-dependent oligodendrogenesis in different CNS environments. We therefore transplanted p57kip2 knockdown aNSCs into white and gray matter (WM and GM) regions of the mouse brain, into uninjured spinal cords as well as in the vicinity of spinal cord injuries and evaluated integration and differentiation in vivo. Our experiments revealed that under healthy conditions intrinsic suppression of p57kip2 as well as WM localization promote differentiation toward myelinating oligodendrocytes at the expense of astrocyte generation. Moreover, p57kip2 knockdown conferred a strong benefit on cell survival augmenting net oligodendrocyte generation. In the vicinity of hemisectioned spinal cords, the gene knockdown led to a similar induction of oligodendroglial features; however, newly generated oligodendrocytes appeared to suffer more from the hostile environment. This study contributes to our understanding of mechanisms of adult oligodendrogenesis and glial heterogeneity and further reveals critical factors when considering aNSC mediated cell replacement in injury and disease.
Collapse
Affiliation(s)
- Felix Beyer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Janusz Jadasz
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Iria Samper Agrelo
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jessica Schira-Heinen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Janos Groh
- Department of Neurology, Developmental Neurobiology, University Hospital Würzburg, Würzburg, Germany
| | - Anastasia Manousi
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Christine Bütermann
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Veronica Estrada
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Laura Reiche
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Martina Cantone
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Julio Vera
- Laboratory of Systems Tumor Immunology, Department of Dermatology, Universitätsklinikum Erlangen, Erlangen, Germany
| | - Francesca Viganò
- Physiological Genomics, Institute of Physiology, Ludwig-Maximilians Universität München, München, Germany
| | - Leda Dimou
- Physiological Genomics, Institute of Physiology, Ludwig-Maximilians Universität München, München, Germany
| | - Hans Werner Müller
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Patrick Küry
- Department of Neurology, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
43
|
Duncan GJ, Manesh SB, Hilton BJ, Assinck P, Plemel JR, Tetzlaff W. The fate and function of oligodendrocyte progenitor cells after traumatic spinal cord injury. Glia 2019; 68:227-245. [PMID: 31433109 DOI: 10.1002/glia.23706] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 07/24/2019] [Accepted: 08/01/2019] [Indexed: 12/27/2022]
Abstract
Oligodendrocyte progenitor cells (OPCs) are the most proliferative and dispersed population of progenitor cells in the adult central nervous system, which allows these cells to rapidly respond to damage. Oligodendrocytes and myelin are lost after traumatic spinal cord injury (SCI), compromising efficient conduction and, potentially, the long-term health of axons. In response, OPCs proliferate and then differentiate into new oligodendrocytes and Schwann cells to remyelinate axons. This culminates in highly efficient remyelination following experimental SCI in which nearly all intact demyelinated axons are remyelinated in rodent models. However, myelin regeneration comprises only one role of OPCs following SCI. OPCs contribute to scar formation after SCI and restrict the regeneration of injured axons. Moreover, OPCs alter their gene expression following demyelination, express cytokines and perpetuate the immune response. Here, we review the functional contribution of myelin regeneration and other recently uncovered roles of OPCs and their progeny to repair following SCI.
Collapse
Affiliation(s)
- Greg J Duncan
- Department of Neurology, Jungers Center for Neurosciences Research, Oregon Health and Science University, Portland, Oregon
| | - Sohrab B Manesh
- Graduate Program in Neuroscience, International Collaboration on Repair Discoveries (ICORD), University of British Columbia (UBC), Vancouver, British Columbia, Canada
| | - Brett J Hilton
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
| | - Peggy Assinck
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh, UK
| | - Jason R Plemel
- Department of Medicine, Division of Neurology, Neuroscience and Mental Health Institute, University of Alberta, Calgary, Alberta, Canada
| | - Wolfram Tetzlaff
- Graduate Program in Neuroscience, International Collaboration on Repair Discoveries (ICORD), University of British Columbia (UBC), Vancouver, British Columbia, Canada.,Departments of Zoology and Surgery, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
44
|
Kataria H, Alizadeh A, Karimi-Abdolrezaee S. Neuregulin-1/ErbB network: An emerging modulator of nervous system injury and repair. Prog Neurobiol 2019; 180:101643. [PMID: 31229498 DOI: 10.1016/j.pneurobio.2019.101643] [Citation(s) in RCA: 66] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 06/07/2019] [Accepted: 06/11/2019] [Indexed: 12/20/2022]
Abstract
Neuregulin-1 (Nrg-1) is a member of the Neuregulin family of growth factors with essential roles in the developing and adult nervous system. Six different types of Nrg-1 (Nrg-1 type I-VI) and over 30 isoforms have been discovered; however, their specific roles are not fully determined. Nrg-1 signals through a complex network of protein-tyrosine kinase receptors, ErbB2, ErbB3, ErbB4 and multiple intracellular pathways. Genetic and pharmacological studies of Nrg-1 and ErbB receptors have identified a critical role for Nrg-1/ErbB network in neurodevelopment including neuronal migration, neural differentiation, myelination as well as formation of synapses and neuromuscular junctions. Nrg-1 signaling is best known for its characterized role in development and repair of the peripheral nervous system (PNS) due to its essential role in Schwann cell development, survival and myelination. However, our knowledge of the impact of Nrg-1/ErbB on the central nervous system (CNS) has emerged in recent years. Ongoing efforts have uncovered a multi-faceted role for Nrg-1 in regulating CNS injury and repair processes. In this review, we provide a timely overview of the most recent updates on Nrg-1 signaling and its role in nervous system injury and diseases. We will specifically highlight the emerging role of Nrg-1 in modulating the glial and immune responses and its capacity to foster neuroprotection and remyelination in CNS injury. Nrg-1/ErbB network is a key regulatory pathway in the developing nervous system; therefore, unraveling its role in neuropathology and repair can aid in development of new therapeutic approaches for nervous system injuries and associated disorders.
Collapse
Affiliation(s)
- Hardeep Kataria
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Arsalan Alizadeh
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Regenerative Medicine Program, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| |
Collapse
|
45
|
Katoh H, Yokota K, Fehlings MG. Regeneration of Spinal Cord Connectivity Through Stem Cell Transplantation and Biomaterial Scaffolds. Front Cell Neurosci 2019; 13:248. [PMID: 31244609 PMCID: PMC6563678 DOI: 10.3389/fncel.2019.00248] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/17/2019] [Indexed: 12/20/2022] Open
Abstract
Significant progress has been made in the treatment of spinal cord injury (SCI). Advances in post-trauma management and intensive rehabilitation have significantly improved the prognosis of SCI and converted what was once an “ailment not to be treated” into a survivable injury, but the cold hard fact is that we still do not have a validated method to improve the paralysis of SCI. The irreversible functional impairment of the injured spinal cord is caused by the disruption of neuronal transduction across the injury lesion, which is brought about by demyelination, axonal degeneration, and loss of synapses. Furthermore, refractory substrates generated in the injured spinal cord inhibit spontaneous recovery. The discovery of the regenerative capability of central nervous system neurons in the proper environment and the verification of neural stem cells in the spinal cord once incited hope that a cure for SCI was on the horizon. That hope was gradually replaced with mounting frustration when neuroprotective drugs, cell transplantation, and strategies to enhance remyelination, axonal regeneration, and neuronal plasticity demonstrated significant improvement in animal models of SCI but did not translate into a cure in human patients. However, recent advances in SCI research have greatly increased our understanding of the fundamental processes underlying SCI and fostered increasing optimism that these multiple treatment strategies are finally coming together to bring about a new era in which we will be able to propose encouraging therapies that will lead to appreciable improvements in SCI patients. In this review, we outline the pathophysiology of SCI that makes the spinal cord refractory to regeneration and discuss the research that has been done with cell replacement and biomaterial implantation strategies, both by itself and as a combined treatment. We will focus on the capacity of these strategies to facilitate the regeneration of neural connectivity necessary to achieve meaningful functional recovery after SCI.
Collapse
Affiliation(s)
- Hiroyuki Katoh
- Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada.,Department of Orthopaedic Surgery - Surgical Sciences, School of Medicine, Tokai University, Tokyo, Japan
| | - Kazuya Yokota
- Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada.,Department of Orthopaedic Surgery, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, Toronto, ON, Canada.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada.,Division of Neurosurgery, University of Toronto, Toronto, ON, Canada.,Spine Program, Toronto Western Hospital, University Health Network, Toronto, ON, Canada
| |
Collapse
|
46
|
Alizadeh A, Dyck SM, Karimi-Abdolrezaee S. Traumatic Spinal Cord Injury: An Overview of Pathophysiology, Models and Acute Injury Mechanisms. Front Neurol 2019; 10:282. [PMID: 30967837 PMCID: PMC6439316 DOI: 10.3389/fneur.2019.00282] [Citation(s) in RCA: 749] [Impact Index Per Article: 124.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Accepted: 03/05/2019] [Indexed: 12/11/2022] Open
Abstract
Traumatic spinal cord injury (SCI) is a life changing neurological condition with substantial socioeconomic implications for patients and their care-givers. Recent advances in medical management of SCI has significantly improved diagnosis, stabilization, survival rate and well-being of SCI patients. However, there has been small progress on treatment options for improving the neurological outcomes of SCI patients. This incremental success mainly reflects the complexity of SCI pathophysiology and the diverse biochemical and physiological changes that occur in the injured spinal cord. Therefore, in the past few decades, considerable efforts have been made by SCI researchers to elucidate the pathophysiology of SCI and unravel the underlying cellular and molecular mechanisms of tissue degeneration and repair in the injured spinal cord. To this end, a number of preclinical animal and injury models have been developed to more closely recapitulate the primary and secondary injury processes of SCI. In this review, we will provide a comprehensive overview of the recent advances in our understanding of the pathophysiology of SCI. We will also discuss the neurological outcomes of human SCI and the available experimental model systems that have been employed to identify SCI mechanisms and develop therapeutic strategies for this condition.
Collapse
Affiliation(s)
- Arsalan Alizadeh
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| | - Scott Matthew Dyck
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| | - Soheila Karimi-Abdolrezaee
- Regenerative Medicine Program, Department of Physiology and Pathophysiology, Rady Faculty of Health Sciences, Spinal Cord Research Center, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|