51
|
Sun ZY, Ma DL, Gu LH, Chen X, Zhang L, Li L. DHF-7 Ameliorates Behavioral Disorders and White Matter Lesions by Regulating BDNF and Fyn in a Mouse Model of Schizophrenia Induced by Cuprizone and MK-801. Int J Neuropsychopharmacol 2022; 25:600-612. [PMID: 35353146 PMCID: PMC9352181 DOI: 10.1093/ijnp/pyac022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 02/24/2022] [Accepted: 03/24/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Schizophrenia is a psychiatric disorder including multiple clinical symptoms such as severe psychosis and cognitive dysfunction. DHF-7 is a novel dihydroflavanone derivative that was designed and synthesized to treat schizophrenia. This study aimed to investigate the effects and mechanisms of DHF-7 in a mouse model of schizophrenia induced by a combination of cuprizone and MK-801. METHODS After intragastric administration of DHF-7 for 7 weeks, open field, Y-maze, and novel object recognition tests were performed to detect behavioral changes in the mouse model. White matter lesions and myelin loss were determined using transmission electron microscopy and oil red O staining. Western blotting and immunohistochemistry were used to detect the expression of the related proteins. RESULTS The results showed that DHF-7 treatment significantly improved cognitive impairment and positive symptoms in the model mice. Moreover, DHF-7 alleviated white matter lesions and demyelination and promoted the differentiation and maturation of oligodendrocytes for remyelination in the corpus callosum of model mice. The mechanistic study showed that DHF-7 increased the expression of brain-derived neurotrophic factor and phosphorylated Fyn, thus activating the tyrosine kinase receptor B (Trk B)/Fyn/N-methyl-D-aspartate receptor subunit 2 B (NMDAR2B) and Raf/mitogen-activated protein kinase (MEK)/ extracellular signal-related kinase (ERK) signaling pathways. CONCLUSIONS Our results provide an experimental basis for the development of DHF-7 as a novel therapeutic agent for schizophrenia.
Collapse
Affiliation(s)
| | | | - Li-Hong Gu
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing, China,National Center for Neurological Disorders, Beijing, China,National Clinical Research Center for Geriatric Diseases, Beijing, China,Beijing Institute for Brain Disorders, Beijing, China,Beijing Engineering Research Center for Nerve System Drugs, Beijing, China,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, China
| | - Xi Chen
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing, China,National Center for Neurological Disorders, Beijing, China,National Clinical Research Center for Geriatric Diseases, Beijing, China,Beijing Institute for Brain Disorders, Beijing, China,Beijing Engineering Research Center for Nerve System Drugs, Beijing, China,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, China
| | - Lan Zhang
- Department of Pharmacy, Xuanwu Hospital, Capital Medical University, Beijing, China,National Center for Neurological Disorders, Beijing, China,National Clinical Research Center for Geriatric Diseases, Beijing, China,Beijing Institute for Brain Disorders, Beijing, China,Beijing Engineering Research Center for Nerve System Drugs, Beijing, China,Key Laboratory for Neurodegenerative Diseases of Ministry of Education, Beijing, China
| | - Lin Li
- Correspondence: Lin Li, MD, PhD, Department of Pharmacy, Xuanwu Hospital, Capital Medical University, 45 Chang-chun Street, Beijing 100053, China ()
| |
Collapse
|
52
|
Pappaianni E, Borsarini B, Doucet GE, Hochman A, Frangou S, Micali N. Initial evidence of abnormal brain plasticity in anorexia nervosa: an ultra-high field study. Sci Rep 2022; 12:2589. [PMID: 35173174 PMCID: PMC8850617 DOI: 10.1038/s41598-022-06113-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 01/05/2022] [Indexed: 11/09/2022] Open
Abstract
Anorexia Nervosa has been associated with white matter abnormalities implicating subcortical abnormal myelination. Extending these findings to intracortical myelin has been challenging but ultra-high field neuroimaging offers new methodological opportunities. To test the integrity of intracortical myelin in AN we used 7 T neuroimaging to acquire T1-weighted images optimized for intracortical myelin from seven females with AN (age range: 18-33) and 11 healthy females (age range: 23-32). Intracortical T1 values (inverse index of myelin concentration) were extracted from 148 cortical regions at ten depth-levels across the cortical ribbon. Across all cortical regions, these levels were averaged to generate estimates of total intracortical myelin concentration and were clustered using principal component analyses into two clusters; the outer cluster comprised T1 values across depth-levels ranging from the CSF boundary to the middle of the cortical regions and the inner cluster comprised T1 values across depth-levels ranging from the middle of the cortical regions to the gray/white matter boundary. Individuals with AN exhibited higher T1 values (i.e., decreased intracortical myelin concentration) in all three metrics. It remains to be established if these abnormalities result from undernutrition or specific lipid nutritional imbalances, or are trait markers; and whether they may contribute to neurobiological deficits seen in AN.
Collapse
Affiliation(s)
- Edoardo Pappaianni
- Department of Psychiatry, Faculty of Medicine, University of Geneva, 2 Rue Verte, 1205, Geneva, Switzerland
| | - Bianca Borsarini
- Department of Psychiatry, Faculty of Medicine, University of Geneva, 2 Rue Verte, 1205, Geneva, Switzerland
| | | | - Ayelet Hochman
- Department of Psychology, St. John's University, Queens, NY, USA
| | - Sophia Frangou
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada
| | - Nadia Micali
- Department of Psychiatry, Faculty of Medicine, University of Geneva, 2 Rue Verte, 1205, Geneva, Switzerland. .,Great Ormond Street Institute of Child Health, University College London, London, UK. .,Department of Pediatrics, Gynecology and Obstetrics, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| |
Collapse
|
53
|
Suzuki H, Imajo Y, Funaba M, Nishida N, Sakamoto T, Sakai T. Current Concepts of Neural Stem/Progenitor Cell Therapy for Chronic Spinal Cord Injury. Front Cell Neurosci 2022; 15:794692. [PMID: 35185471 PMCID: PMC8850278 DOI: 10.3389/fncel.2021.794692] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Accepted: 12/20/2021] [Indexed: 11/13/2022] Open
Abstract
Chronic spinal cord injury (SCI) is a devastating condition that results in major neurological deficits and social burden. It continues to be managed symptomatically, and no real therapeutic strategies have been devised for its treatment. Neural stem/neural progenitor cells (NSCs/NPCs) being used for the treatment of chronic SCI in experimental SCI models can not only replace the lost cells and remyelinate axons in the injury site but also support their growth and provide neuroprotective factors. Currently, several clinical studies using NSCs/NPCs are underway worldwide. NSCs/NPCs also have the potential to differentiate into all three neuroglial lineages to regenerate neural circuits, demyelinate denuded axons, and provide trophic support to endogenous cells. This article explains the challenging pathophysiology of chronic SCI and discusses key NSC/NPC-based techniques having the greatest potential for translation over the next decade.
Collapse
|
54
|
Carvalho E, Morais M, Ferreira H, Silva M, Guimarães S, Pêgo A. A paradigm shift: Bioengineering meets mechanobiology towards overcoming remyelination failure. Biomaterials 2022; 283:121427. [DOI: 10.1016/j.biomaterials.2022.121427] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 01/31/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022]
|
55
|
Basu S, Choudhury IN, Nazareth L, Chacko A, Shelper T, Vial ML, Ekberg JAK, St John JA. In vitro modulation of Schwann cell behavior by VEGF and PDGF in an inflammatory environment. Sci Rep 2022; 12:662. [PMID: 35027585 PMCID: PMC8758747 DOI: 10.1038/s41598-021-04222-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 12/17/2021] [Indexed: 01/19/2023] Open
Abstract
Peripheral glial cell transplantation with Schwann cells (SCs) is a promising approach for treating spinal cord injury (SCI). However, improvements are needed and one avenue to enhance regenerative functional outcomes is to combine growth factors with cell transplantation. Vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) are neuroprotective, and a combination of these factors has improved outcomes in rat SCI models. Thus, transplantation of SCs combined with VEGF and PDGF may further improve regenerative outcomes. First, however, we must understand how the two factors modulate SCs. In this in vitro study, we show that an inflammatory environment decreased the rate of SC-mediated phagocytosis of myelin debris but the addition of VEGF and PDGF (alone and combined) improved phagocytosis. Cytokine expression by SCs in the inflammatory environment revealed that addition of PDGF led to significantly lower level of pro-inflammatory cytokine, TNF-α, but IL-6 and anti-inflammatory cytokines (TGF-β and IL-10), remained unaltered. Further, PDGF was able to decrease the expression of myelination associated gene Oct6 in the presence of inflammatory environment. Overall, these results suggest that the use of VEGF and/or PDGF combined with SC transplantation may be beneficial in SCI therapy.
Collapse
Affiliation(s)
- Souptik Basu
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Indra N Choudhury
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Lynn Nazareth
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Anu Chacko
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Todd Shelper
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Marie-Laure Vial
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia
| | - Jenny A K Ekberg
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia.,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia.,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia
| | - James A St John
- Clem Jones Centre for Neurobiology and Stem Cell Research, Griffith University, Nathan, QLD, Australia. .,Menzies Health Institute Queensland, Griffith University, Southport, QLD, Australia. .,Griffith Institute for Drug Discovery, Griffith University, Nathan, QLD, Australia.
| |
Collapse
|
56
|
Abstract
Traumatic injury of the central nervous system (CNS) is a worldwide health problem affecting millions of people. Trauma of the CNS, that is, traumatic brain injury (TBI) and spinal cord injury (SCI), lead to massive and progressive cell loss and axonal degeneration, usually with very limited regeneration. At present, there are no treatments to protect injured CNS tissue or to replace the lost tissue. Stem cells are a cell type that by definition can self-renew and give rise to multiple cell lineages. In recent years, therapies using stem and progenitor cells have shown promising effects in experimental CNS trauma, particularly in the acute-subacute stage, but also in chronic injuries. However, the therapeutic mechanisms by which transplanted cells achieve the structural and/or functional improvements are often not clear. Stem cell therapies for CNS trauma can be categorized into 2 main concepts, transplantation of exogenous neural stem cells and neural progenitor cells and recruitment of endogenous stem and progenitor cells. In this review, focusing on the advances during the last decade, we will discuss the major cell therapies, the pros and cons of these 2 concepts for TBI and SCI, and the treatment strategies we believe will be successful.
Collapse
Affiliation(s)
- Xiaofei Li
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Erik Sundström
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Corresponding author: Erik Sundström, Department of Neurobiology, Care Sciences and Society (NVS), BioClinicum J9:20, Karolinska University Hospital, S17164 Solna, Sweden.
| |
Collapse
|
57
|
Remyelination in PNS and CNS: current and upcoming cellular and molecular strategies to treat disabling neuropathies. Mol Biol Rep 2021; 48:8097-8110. [PMID: 34731366 DOI: 10.1007/s11033-021-06755-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 09/15/2021] [Indexed: 10/19/2022]
Abstract
Myelin is a lipid-rich nerve cover that consists of glial cell's plasmalemma layers and accelerates signal conduction. Axon-myelin contact is a source for many developmental and regenerative signals of myelination. Intra- or extracellular factors including both enhancers and inhibitors are other factors affecting the myelination process. Myelin damages are observed in several congenital and hereditary diseases, physicochemical conditions, infections, or traumatic insults, and remyelination is known as an intrinsic response to injuries. Here we discuss some molecular events and conditions involved in de- and remyelination and compare the phenomena of remyelination in CNS and PNS. We have explained applying some of these molecular events in myelin restoration. Finally, the current and upcoming treatment strategies for myelin restoration are explained in three groups of immunotherapy, endogenous regeneration enhancement, and cell therapy to give a better insight for finding the more effective rehabilitation strategies considering the underlying molecular events of a lesion formation and its current condition.
Collapse
|
58
|
Kim Y, Roh EJ, Joshi HP, Shin HE, Choi H, Kwon SY, Sohn S, Han I. Bazedoxifene, a Selective Estrogen Receptor Modulator, Promotes Functional Recovery in a Spinal Cord Injury Rat Model. Int J Mol Sci 2021; 22:ijms222011012. [PMID: 34681670 PMCID: PMC8537911 DOI: 10.3390/ijms222011012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/06/2021] [Accepted: 10/08/2021] [Indexed: 12/14/2022] Open
Abstract
In research on various central nervous system injuries, bazedoxifene acetate (BZA) has shown two main effects: neuroprotection by suppressing the inflammatory response and remyelination by enhancing oligodendrocyte precursor cell differentiation and oligodendrocyte proliferation. We examined the effects of BZA in a rat spinal cord injury (SCI) model. Anti-inflammatory and anti-apoptotic effects were investigated in RAW 264.7 cells, and blood-spinal cord barrier (BSCB) permeability and angiogenesis were evaluated in a human brain endothelial cell line (hCMEC/D3). In vivo experiments were carried out on female Sprague Dawley rats subjected to moderate static compression SCI. The rats were intraperitoneally injected with either vehicle or BZA (1mg/kg pre-SCI and 3 mg/kg for 7 days post-SCI) daily. BZA decreased the lipopolysaccharide-induced production of proinflammatory cytokines and nitric oxide in RAW 264.7 cells and preserved BSCB disruption in hCMEC/D3 cells. In the rats, BZA reduced caspase-3 activity at 1 day post-injury (dpi) and suppressed phosphorylation of MAPK (p38 and ERK) at dpi 2, hence reducing the expression of IL-6, a proinflammatory cytokine. BZA also led to remyelination at dpi 20. BZA contributed to improvements in locomotor recovery after compressive SCI. This evidence suggests that BZA may have therapeutic potential to promote neuroprotection, remyelination, and functional outcomes following SCI.
Collapse
Affiliation(s)
- Yiyoung Kim
- School of Medicine, CHA University, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea;
| | - Eun Ji Roh
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (E.J.R.); (H.E.S.); (H.C.); (S.Y.K.); (S.S.)
| | - Hari Prasad Joshi
- Department of Physiology and Pathophysiology, Spinal Cord Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0W2, Canada;
| | - Hae Eun Shin
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (E.J.R.); (H.E.S.); (H.C.); (S.Y.K.); (S.S.)
| | - Hyemin Choi
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (E.J.R.); (H.E.S.); (H.C.); (S.Y.K.); (S.S.)
| | - Su Yeon Kwon
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (E.J.R.); (H.E.S.); (H.C.); (S.Y.K.); (S.S.)
| | - Seil Sohn
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (E.J.R.); (H.E.S.); (H.C.); (S.Y.K.); (S.S.)
| | - Inbo Han
- Department of Neurosurgery, CHA University School of Medicine, CHA Bundang Medical Center, Seongnam-si 13496, Gyeonggi-do, Korea; (E.J.R.); (H.E.S.); (H.C.); (S.Y.K.); (S.S.)
- Correspondence:
| |
Collapse
|
59
|
Jeffries MA, McLane LE, Khandker L, Mather ML, Evangelou AV, Kantak D, Bourne JN, Macklin WB, Wood TL. mTOR Signaling Regulates Metabolic Function in Oligodendrocyte Precursor Cells and Promotes Efficient Brain Remyelination in the Cuprizone Model. J Neurosci 2021; 41:8321-8337. [PMID: 34417330 PMCID: PMC8496195 DOI: 10.1523/jneurosci.1377-20.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 06/21/2021] [Accepted: 08/10/2021] [Indexed: 02/02/2023] Open
Abstract
In demyelinating diseases, such as multiple sclerosis, primary loss of myelin and subsequent neuronal degeneration throughout the CNS impair patient functionality. While the importance of mechanistic target of rapamycin (mTOR) signaling during developmental myelination is known, no studies have yet directly examined the function of mTOR signaling specifically in the oligodendrocyte (OL) lineage during remyelination. Here, we conditionally deleted Mtor from adult oligodendrocyte precursor cells (OPCs) using Ng2-CreERT in male adult mice to test its function in new OLs responsible for remyelination. During early remyelination after cuprizone-induced demyelination, mice lacking mTOR in adult OPCs had unchanged OL numbers but thinner myelin. Myelin thickness recovered by late-stage repair, suggesting a delay in myelin production when Mtor is deleted from adult OPCs. Surprisingly, loss of mTOR in OPCs had no effect on efficiency of remyelination after lysophosphatidylcholine lesions in either the spinal cord or corpus callosum, suggesting that mTOR signaling functions specifically in a pathway dysregulated by cuprizone to promote remyelination efficiency. We further determined that cuprizone and inhibition of mTOR cooperatively compromise metabolic function in primary rat OLs undergoing differentiation. Together, our results support the conclusion that mTOR signaling in OPCs is required to overcome the metabolic dysfunction in the cuprizone-demyelinated adult brain.SIGNIFICANCE STATEMENT Impaired remyelination by oligodendrocytes contributes to the progressive pathology in multiple sclerosis, so it is critical to identify mechanisms of improving remyelination. The goal of this study was to examine mechanistic target of rapamycin (mTOR) signaling in remyelination. Here, we provide evidence that mTOR signaling promotes efficient remyelination of the brain after cuprizone-mediated demyelination but has no effect on remyelination after lysophosphatidylcholine demyelination in the spinal cord or brain. We also present novel data revealing that mTOR inhibition and cuprizone treatment additively affect the metabolic profile of differentiating oligodendrocytes, supporting a mechanism for the observed remyelination delay. These data suggest that altered metabolic function may underlie failure of remyelination in multiple sclerosis lesions and that mTOR signaling may be of therapeutic potential for promoting remyelination.
Collapse
Affiliation(s)
- Marisa A Jeffries
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Lauren E McLane
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Luipa Khandker
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Marie L Mather
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Angelina V Evangelou
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Divyangi Kantak
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| | - Jennifer N Bourne
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Wendy B Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, Colorado 80045
| | - Teresa L Wood
- Department of Pharmacology, Physiology, and Neuroscience and Center for Cell Signaling, Rutgers New Jersey Medical School, Newark, New Jersey 07103
| |
Collapse
|
60
|
Hashem HR. Evaluation of the postnatal effects induced by Diazinon on the Growth of the mice offspring and the development of their cerebellar cortex. Cells Tissues Organs 2021; 211:539-554. [PMID: 34425578 DOI: 10.1159/000518993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 08/05/2021] [Indexed: 11/19/2022] Open
Affiliation(s)
- Heba R Hashem
- Anatomy and Embryology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
61
|
A Time-Course Study of the Expression Level of Synaptic Plasticity-Associated Genes in Un-Lesioned Spinal Cord and Brain Areas in a Rat Model of Spinal Cord Injury: A Bioinformatic Approach. Int J Mol Sci 2021; 22:ijms22168606. [PMID: 34445312 PMCID: PMC8395345 DOI: 10.3390/ijms22168606] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/27/2021] [Accepted: 08/06/2021] [Indexed: 12/27/2022] Open
Abstract
"Neuroplasticity" is often evoked to explain adaptation and compensation after acute lesions of the Central Nervous System (CNS). In this study, we investigated the modification of 80 genes involved in synaptic plasticity at different times (24 h, 8 and 45 days) from the traumatic spinal cord injury (SCI), adopting a bioinformatic analysis. mRNA expression levels were analyzed in the motor cortex, basal ganglia, cerebellum and in the spinal segments rostral and caudal to the lesion. The main results are: (i) a different gene expression regulation is observed in the Spinal Cord (SC) segments rostral and caudal to the lesion; (ii) long lasting changes in the SC includes the extracellular matrix (ECM) enzymes Timp1, transcription regulators (Egr, Nr4a1), second messenger associated proteins (Gna1, Ywhaq); (iii) long-lasting changes in the Motor Cortex includes transcription regulators (Cebpd), neurotransmitters/neuromodulators and receptors (Cnr1, Gria1, Nos1), growth factors and related receptors (Igf1, Ntf3, Ntrk2), second messenger associated proteins (Mapk1); long lasting changes in Basal Ganglia and Cerebellum include ECM protein (Reln), growth factors (Ngf, Bdnf), transcription regulators (Egr, Cebpd), neurotransmitter receptors (Grin2c). These data suggest the molecular mapping as a useful tool to investigate the brain and SC reorganization after SCI.
Collapse
|
62
|
Jahandideh A, Noori H, Rahimi B, Hamblin MR, Behroozi Z, Ramezani M, Ramezani F. Alginate scaffolds improve functional recovery after spinal cord injury. Eur J Trauma Emerg Surg 2021; 48:1711-1721. [PMID: 34363487 DOI: 10.1007/s00068-021-01760-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 07/27/2021] [Indexed: 12/21/2022]
Abstract
PURPOSE In this systematic review and meta-analysis, the use of alginate for the repair of the damaged spinal cord was investigated. METHODS After an extensive search of databases including MEDLINE, SCOPUS, EMBASE and Web of Science, an initial screening was performed based on inclusion and exclusion criteria. The full text of related articles was reviewed and data mining was performed. Data were analyzed by calculating the mean of ratios between treated and untreated groups using STATA software. Subgroup analysis was also performed due to heterogeneity. Articles were subjected to quality control and PRISMA guidelines were followed. RESULTS Twelve studies and 17 experiments were included in the study. After SCI, alginate hydrogel had a moderate effect on motor function recovery (SMD = 0.64; 95% CI 0.28-1.00; p < 0.0001) and alginate scaffolds loaded with drugs, growth factors, or cells on the SCI group compared with untreated SCI animals showed has a strong effect in the treatment of SCI (SMD = 2.82; 95% CI 1.49-4.145; p < 0.0001). Treatment with drug/cell in combination with alginate was more strongly significant compared to the groups treated with drug/cell alone (SMD = 4.55; 95% CI 1.42-7.69; p < 0.0001). Alginate alone or in combination therapy when used as an implant, had a more significant effect than injection. CONCLUSION These findings suggest that alginate is an efficient scaffold for functional recovery and even a much better scaffold for drug/cell delivery after SCI.
Collapse
Affiliation(s)
- Atefeh Jahandideh
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hamid Noori
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Behnaz Rahimi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, 2028, South Africa
| | - Zahra Behroozi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Fatemeh Ramezani
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
63
|
Wang Z, Baharani A, Wei Z, Truong D, Bi X, Wang F, Li XM, Verge VMK, Zhang Y. Low field magnetic stimulation promotes myelin repair and cognitive recovery in chronic cuprizone mouse model. Clin Exp Pharmacol Physiol 2021; 48:1090-1102. [PMID: 33638234 DOI: 10.1111/1440-1681.13490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/09/2021] [Accepted: 02/23/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Multiple sclerosis (MS) is an inflammatory demyelinating disease featured with neuroinflammation, demyelination, and the loss of oligodendrocytes. Cognitive impairment and depression are common neuropsychiatric symptoms in MS that are poorly managed with the present interventions. OBJECTIVE This study aimed to investigate the effects of low field magnetic stimulation (LFMS), a novel non-invasive neuromodulation technology, on cognitive impairment and depressive symptoms associated with MS using a mouse model of demyelination. METHODS C57BL female mice were fed with a 0.2% cuprizone diet for 12 weeks to induce a chronic demyelinating model followed by 4 weeks of cuprizone withdrawal with either sham or LFMS treatment. RESULTS Improved cognition and depression-like behaviour and restored weight gain were observed in mice with LFMS treatment. Immunohistochemical and immunoblotting data showed enhanced myelin basic protein (MBP) and myelin oligodendrocyte glycoprotein expressions (MOG) in the prefrontal cortex of mice with LFMS treatment, supporting that myelin repair was promoted. LFMS also increased the protein expression of mature oligodendrocyte biomarker glutathione-S-transferase (GST-π). In addition, expression of TGF-β and associated receptors were elevated with LFMS treatment, implicating this pathway in the response. CONCLUSION Results from the present study revealed LFMS to have neuroprotective effects, suggesting that LFMS has potential therapeutic value for treating cognitive impairment and depression related to demyelination disorders.
Collapse
Affiliation(s)
- Zitong Wang
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Akanksha Baharani
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Zelan Wei
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Davin Truong
- College of Dentistry, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiaoying Bi
- Department of Neurology, Shanghai Changhai Hospital, the Second Military Medical University, Shanghai, China
| | - Fei Wang
- Nanjing Brain Hospital, Nanjing Medical University, Nanjing, China
| | - Xin-Min Li
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Valerie M K Verge
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
- Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Yanbo Zhang
- Department of Psychiatry, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
- Cameco MS Neuroscience Research Centre, College of Medicine, University of Saskatchewan, Saskatoon, SK, Canada
- Department of Psychiatry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
64
|
Franklin ZG, Ladagu AD, Germain Jean Magloire KW, Folarin OR, Sefirin D, Tashara TG, Dieudonne N, Olopade JO. Ameliorative effects of the aqueous extract of Khaya anthotheca (Welw.) C.DC (Meliaceae) in vanadium induced anxiety, memory loss and pathologies in the brain and ovary of mice. JOURNAL OF ETHNOPHARMACOLOGY 2021; 275:114099. [PMID: 33831470 DOI: 10.1016/j.jep.2021.114099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 02/08/2021] [Accepted: 04/01/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ethnobotanical enquiries have revealed that Khaya anthotheca (Welw.) C.DC (Meliaceae) has anxiolytic properties and is used to alleviate vaginal dryness in postmenopausal women in Cameroon. The aim of this study was to evaluate the ameliorative effects of the aqueous extract of K. anthotheca in vanadium induced anxiety, memory loss and pathologies in the brain and ovary of mice. MATERIAL AND METHODS Forty neonatal female mice were used in this study. All animals received vanadium (3 mg/kg BW/72 h, by lactation and i.p.) for 20 weeks except the Control group. At 16 weeks old, mice were divided into 5 groups (n = 8): Control group received distilled water; V-group received vanadium (V) (3 mg/kg BW every 72 h i.p.), V + Vit E group received vitamin E (500 mg/kg BW/72 h) and vanadium (V) (3 mg/kg BW/72 h i.p, simultaneously). V + KA 125 and V + KA 250 groups received K. anthotheca extract at the doses of 125 and 250 mg/kg BW/day respectively and vanadium (V) (3 mg/kg BW/72 h i.p, simultaneously).The treatment was done per os at 10 mL/kg of volume of administration for 4 weeks. To evalute anxiolytic effects and spatial working memory improved by the extract in mice, the elevated open space test and Y maze test were used respectively. After sacrifice, brains were harvested and pathologies were assessed using cresyl violet stainning and immunohistochemistry (GFAP, Iba-1 and MBP), while pathologies in the ovaries were assessed using immunohistochemistry (Collagen type 1) and H&E stainning. RESULTS Results in the three sessions of elevated open space test showed that vanadium exposure resulted in a significant (p < 0.05; p < 0.01) increase of the latency of first entry in the slopes and a significant (p < 0.05; p < 0.01; p < 0.001) decrease of the time spent and number of entries in the slopes however, Khaya anthotheca treatment induced a significant (p < 0.05; p < 0.01) decrease of the latency of first entry in the slopes and a significant (p < 0.05; p < 0.01) increase of the time spent and number of entries in the slopes. In the Y maze test, vanadium exposure resulted in a significant decrease (p < 0.01) in the percentage of correct alternation, K. anthotheca extract at the dose of 250 mg/kg BW however induced a significant (p < 0.05) increase of this percentage of correct spontaneous alternation. In the brain, degeneration induced by vanadium exposure was marked by an increase of GFAP-immunoreactive cells, microgliosis and demyelination. The treatment with Khaya anthotheca extract at the dose of 250 mg/kg BW resulted in the preservation of cellular integrity in the same anatomical regions with reduced astroglial and microglial activation and prevented demyelination. In addition, vanadium exposure decreased Collagen type 1 expression in the ovaries and induced a deterioration of tertiary follicle. Khaya anthotheca treatment at the dose of 250 mg/kg BW induced an increase of expression of Collagen type 1 and alleviated deterioration of the microarchitecture of tertiary follicle induced by vanadium. CONCLUSION These effects induced by K. anthotheca extract could justify the traditional use of this plant in Cameroonian traditional medicine to manage anxiety. Therefore, to minimise vanadium induced toxicity, the plant should be given more emphasis as a candidate in developing a modern phytodrug.
Collapse
Affiliation(s)
- Zemo Gamo Franklin
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon; Department of Psychology, Faculty of Arts, Letters and Social Science, University of Yaounde I, P.O. Box 7011, Yaounde, Cameroon.
| | - Amany Digal Ladagu
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, 20001, Nigeria.
| | | | - Oluwabusayo Rachael Folarin
- Department of Biomedical Laboratory Science, Faculty of Basic Medical Sciences, College of Medicine, University of Ibadan, Ibadan, 200284, Nigeria.
| | - Djiogue Sefirin
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon.
| | - Taidinda Gilbert Tashara
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, 20001, Nigeria.
| | - Njamen Dieudonne
- Department of Animal Biology and Physiology, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon.
| | - James Olukayode Olopade
- Department of Veterinary Anatomy, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, 20001, Nigeria.
| |
Collapse
|
65
|
Suzuki H, Sakai T. Current Concepts of Stem Cell Therapy for Chronic Spinal Cord Injury. Int J Mol Sci 2021; 22:ijms22147435. [PMID: 34299053 PMCID: PMC8308009 DOI: 10.3390/ijms22147435] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic spinal cord injury (SCI) is a catastrophic condition associated with significant neurological deficit and social and financial burdens. It is currently being managed symptomatically with no real therapeutic strategies available. In recent years, a number of innovative regenerative strategies have emerged and have been continuously investigated in clinical trials. In addition, several more are coming down the translational pipeline. Among ongoing and completed trials are those reporting the use of mesenchymal stem cells, neural stem/progenitor cells, induced pluripotent stem cells, olfactory ensheathing cells, and Schwann cells. The advancements in stem cell technology, combined with the powerful neuroimaging modalities, can now accelerate the pathway of promising novel therapeutic strategies from bench to bedside. Various combinations of different molecular therapies have been combined with supportive scaffolds to facilitate favorable cell–material interactions. In this review, we summarized some of the most recent insights into the preclinical and clinical studies using stem cells and other supportive drugs to unlock the microenvironment in chronic SCI to treat patients with this condition. Successful future therapies will require these stem cells and other synergistic approaches to address the persistent barriers to regeneration, including glial scarring, loss of structural framework, and immunorejection.
Collapse
|
66
|
Liu H, Yang X, Yang J, Yuan Y, Wang Y, Zhang R, Xiong H, Xu Y. IL-17 Inhibits Oligodendrocyte Progenitor Cell Proliferation and Differentiation by Increasing K + Channel Kv1.3. Front Cell Neurosci 2021; 15:679413. [PMID: 34239419 PMCID: PMC8258110 DOI: 10.3389/fncel.2021.679413] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/20/2021] [Indexed: 12/02/2022] Open
Abstract
Interleukin 17 (IL-17) is a signature cytokine of Th17 cells. IL-17 level is significantly increased in inflammatory conditions of the CNS, including but not limited to post-stroke and multiple sclerosis. IL-17 has been detected direct toxicity on oligodendrocyte (Ol) lineage cells and inhibition on oligodendrocyte progenitor cell (OPC) differentiation, and thus promotes myelin damage. The cellular mechanism of IL-17 in CNS inflammatory diseases remains obscure. Voltage-gated K+ (Kv) channel 1.3 is the predominant Kv channel in Ol and potentially involved in Ol function and cell cycle regulation. Kv1.3 of T cells involves in immunomodulation of inflammatory progression, but the role of Ol Kv1.3 in inflammation-related pathogenesis has not been fully investigated. We hypothesized that IL-17 induces myelin injury through Kv1.3 activation. To test the hypothesis, we studied the involvement of OPC/Ol Kv1.3 in IL-17-induced Ol/myelin injury in vitro and in vivo. Kv1.3 currents and channel expression gradually decreased during the OPC development. Application of IL-17 to OPC culture increased Kv1.3 expression, leading to a decrease of AKT activation, inhibition of proliferation and myelin basic protein reduction, which were prevented by a specific Kv1.3 blocker 5-(4-phenoxybutoxy) psoralen. IL-17-caused myelin injury was validated in LPC-induced demyelination mouse model, particularly in corpus callosum, which was also mitigated by aforementioned Kv1.3 antagonist. IL-17 altered Kv1.3 expression and resultant inhibitory effects on OPC proliferation and differentiation may by interrupting AKT phosphorylating activation. Taken together, our results suggested that IL-17 impairs remyelination and promotes myelin damage by Kv1.3-mediated Ol/myelin injury. Thus, blockade of Kv1.3 as a potential therapeutic strategy for inflammatory CNS disease may partially attribute to the direct protection on OPC proliferation and differentiation other than immunomodulation.
Collapse
Affiliation(s)
- Han Liu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xueke Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanpeng Yuan
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanlin Wang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Rui Zhang
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huangui Xiong
- Neurophysiology Laboratory, Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE, United States
| | - Yuming Xu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
67
|
Fang S, Zhong L, Wang AQ, Zhang H, Yin ZS. Identification of Regeneration and Hub Genes and Pathways at Different Time Points after Spinal Cord Injury. Mol Neurobiol 2021; 58:2643-2662. [PMID: 33484404 DOI: 10.1007/s12035-021-02289-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 01/11/2021] [Indexed: 12/19/2022]
Abstract
Spinal cord injury (SCI) is a neurological injury that can cause neuronal loss around the lesion site and leads to locomotive and sensory deficits. However, the underlying molecular mechanisms remain unclear. This study aimed to verify differential gene time-course expression in SCI and provide new insights for gene-level studies. We downloaded two rat expression profiles (GSE464 and GSE45006) from the Gene Expression Omnibus database, including 1 day, 3 days, 7 days, and 14 days post-SCI, along with thoracic spinal cord data for analysis. At each time point, gene integration was performed using "batch normalization." The raw data were standardized, and differentially expressed genes at the different time points versus the control were analyzed by Gene Ontology enrichment analysis, the Kyoto Encyclopedia of Genes and Genomes pathway analysis, and gene set enrichment analysis. A protein-protein interaction network was then built and visualized. In addition, ten hub genes were identified at each time point. Among them, Gnb5, Gng8, Agt, Gnai1, and Psap lack correlation studies in SCI and deserve further investigation. Finally, we screened and analyzed genes for tissue repair, reconstruction, and regeneration and found that Anxa1, Snap25, and Spp1 were closely related to repair and regeneration after SCI. In conclusion, hub genes, signaling pathways, and regeneration genes involved in secondary SCI were identified in our study. These results may be useful for understanding SCI-related biological processes and the development of targeted intervention strategies.
Collapse
Affiliation(s)
- Sheng Fang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Lin Zhong
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui Province, China
- Department of Orthopedics, The Third Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - An-Quan Wang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Hui Zhang
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Zong-Sheng Yin
- Department of Orthopedics, The First Affiliated Hospital of Anhui Medical University, #218 Jixi Road, Hefei, 230022, Anhui Province, China.
| |
Collapse
|
68
|
GluA2 overexpression in oligodendrocyte progenitors promotes postinjury oligodendrocyte regeneration. Cell Rep 2021; 35:109147. [PMID: 34010640 PMCID: PMC8185898 DOI: 10.1016/j.celrep.2021.109147] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 01/28/2020] [Accepted: 04/27/2021] [Indexed: 01/18/2023] Open
Abstract
Oligodendrocyte precursor cells (OPCs) are essential for developmental myelination and oligodendrocyte regeneration after CNS injury. These progenitors express calcium-permeable AMPA receptors (AMPARs) and form direct synapses with neurons throughout the CNS, but the roles of this signaling are unclear. To enable selective alteration of the properties of AMPARs in oligodendroglia, we generate mice that allow cell-specific overexpression of EGFP-GluA2 in vivo. In healthy conditions, OPC-specific GluA2 overexpression significantly increase their proliferation in an age-dependent manner but did not alter their rate of differentiation into oligodendrocytes. In contrast, after demyelinating brain injury in neonates or adults, higher GluA2 levels promote both OPC proliferation and oligodendrocyte regeneration, but do not prevent injury-induced initial cell loss. These findings indicate that AMPAR GluA2 content regulates the proliferative and regenerative behavior of adult OPCs, serving as a putative target for better myelin repair.
Collapse
|
69
|
González P, González-Fernández C, Javier Rodríguez F. Effects of Wnt5a overexpression in spinal cord injury. J Cell Mol Med 2021; 25:5150-5163. [PMID: 33939286 PMCID: PMC8178287 DOI: 10.1111/jcmm.16507] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 03/05/2021] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
Accordingly to its known function in corticospinal tract (CST) developmental growth, previous reports have shown an inhibitory role of Wnt5a in CST regeneration after spinal cord injury (SCI). Interestingly, it has been subsequently demonstrated that Wnt5a also modulates the developmental growth of non‐CST axons and that different Wnt5a receptors are expressed in neurons, oligodendrocytes, NG2+ glial precursors and reactive microglia/macrophages and astrocytes after SCI. However, the role of Wnt5a in the response of these cell types, in the regeneration of non‐CST axons and in functional recovery after SCI is currently unknown. To evaluate this, rats were subjected to spinal cord contusion and injected with a lentiviral vector generated to overexpress Wnt5a. Histological analyses were performed in spinal cord sections processed for the visualization of myelin, oligodendrocytes, neurons, microglia/macrophages, astrocytes, NG2+ glial precursors and serotonergic axons. Motor and bladder function recovery were also assessed. Further advancing our knowledge on the role of Wnt5a in SCI, we found that, besides its previously reported functions, Wnt5a overexpression elicits a reduction on neuronal cell density, the accumulation of NG2+ glial precursors and the descending serotonergic innervation in the affected areas, along with impairment of motor and bladder function recovery after SCI.
Collapse
Affiliation(s)
- Pau González
- Laboratory of Molecular Neurology, Hospital Nacional de Parapléjicos, Toledo, Spain
| | | | | |
Collapse
|
70
|
McIntyre WB, Pieczonka K, Khazaei M, Fehlings MG. Regenerative replacement of neural cells for treatment of spinal cord injury. Expert Opin Biol Ther 2021; 21:1411-1427. [PMID: 33830863 DOI: 10.1080/14712598.2021.1914582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Introduction: Traumatic Spinal Cord Injury (SCI) results from primary physical injury to the spinal cord, which initiates a secondary cascade of neural cell death. Current therapeutic approaches can attenuate the consequences of the primary and secondary events, but do not address the degenerative aspects of SCI. Transplantation of neural stem/progenitor cells (NPCs) for the replacement of the lost/damaged neural cells is suggested here as a regenerative approach that is complementary to current therapeutics.Areas Covered: This review addresses how neurons, oligodendrocytes, and astrocytes are impacted by traumatic SCI, and how current research in regenerative-NPC therapeutics aims to restore their functionality. Methods used to enhance graft survival, as well as bias progenitor cells towards neuronal, oligodendrogenic, and astroglia lineages are discussed.Expert Opinion: Despite an NPC's ability to differentiate into neurons, oligodendrocytes, and astrocytes in the transplant environment, their potential therapeutic efficacy requires further optimization prior to translation into the clinic. Considering the temporospatial identity of NPCs could promote neural repair in region specific injuries throughout the spinal cord. Moreover, understanding which cells are targeted by NPC-derived myelinating cells can help restore physiologically-relevant myelin patterns. Finally, the duality of astrocytes is discussed, outlining their context-dependent importance in the treatment of SCI.
Collapse
Affiliation(s)
- William Brett McIntyre
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Katarzyna Pieczonka
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada
| | - Mohamad Khazaei
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada
| | - Michael G Fehlings
- Division of Genetics and Development, Krembil Research Institute, University Health Network, Toronto, ON, Canada.,Institute of Medical Sciences, University of Toronto, Toronto, ON, Canada.,Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
71
|
Kim S, Lee DW, Schachner M, Park HC. Small compounds mimicking the adhesion molecule L1 improve recovery in a zebrafish demyelination model. Sci Rep 2021; 11:5878. [PMID: 33723325 PMCID: PMC7960995 DOI: 10.1038/s41598-021-85412-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 03/01/2021] [Indexed: 02/05/2023] Open
Abstract
Demyelination leads to a loss of neurons, which results in, among other consequences, a severe reduction in locomotor function, and underlies several diseases in humans including multiple sclerosis and polyneuropathies. Considerable clinical progress has been made in counteracting demyelination. However, there remains a need for novel methods that reduce demyelination while concomitantly achieving remyelination, thus complementing the currently available tools to ameliorate demyelinating diseases. In this study, we used an established zebrafish demyelination model to test selected compounds, following a screening in cell culture experiments and in a mouse model of spinal cord injury that was aimed at identifying beneficial functions of the neural cell adhesion molecule L1. In comparison to mammalian nervous system disease models, the zebrafish allows testing of potentially promotive compounds more easily than what is possible in mammals. We found that our selected compounds tacrine and duloxetine significantly improved remyelination in the peripheral and central nervous system of transgenic zebrafish following pharmacologically induced demyelination. Given that both molecules are known to positively affect functions other than those related to L1 and in other disease contexts, we propose that their combined beneficial function raises hope for the use of these compounds in clinical settings.
Collapse
Affiliation(s)
- Suhyun Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, 15335, Republic of Korea
| | - Dong-Won Lee
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, 15335, Republic of Korea
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, NJ, 08554, USA.
- Center for Neuroscience, Shantou University Medical College, Shantou, 515041, Guangdong, China.
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan, 15335, Republic of Korea.
| |
Collapse
|
72
|
Eckert MA, Harris KC, Lang H, Lewis MA, Schmiedt RA, Schulte BA, Steel KP, Vaden KI, Dubno JR. Translational and interdisciplinary insights into presbyacusis: A multidimensional disease. Hear Res 2021; 402:108109. [PMID: 33189490 PMCID: PMC7927149 DOI: 10.1016/j.heares.2020.108109] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 10/19/2020] [Accepted: 10/25/2020] [Indexed: 12/18/2022]
Abstract
There are multiple etiologies and phenotypes of age-related hearing loss or presbyacusis. In this review we summarize findings from animal and human studies of presbyacusis, including those that provide the theoretical framework for distinct metabolic, sensory, and neural presbyacusis phenotypes. A key finding in quiet-aged animals is a decline in the endocochlear potential (EP) that results in elevated pure-tone thresholds across frequencies with greater losses at higher frequencies. In contrast, sensory presbyacusis appears to derive, in part, from acute and cumulative effects on hair cells of a lifetime of environmental exposures (e.g., noise), which often result in pronounced high frequency hearing loss. These patterns of hearing loss in animals are recognizable in the human audiogram and can be classified into metabolic and sensory presbyacusis phenotypes, as well as a mixed metabolic+sensory phenotype. However, the audiogram does not fully characterize age-related changes in auditory function. Along with the effects of peripheral auditory system declines on the auditory nerve, primary degeneration in the spiral ganglion also appears to contribute to central auditory system aging. These inner ear alterations often correlate with structural and functional changes throughout the central nervous system and may explain suprathreshold speech communication difficulties in older adults with hearing loss. Throughout this review we highlight potential methods and research directions, with the goal of advancing our understanding, prevention, diagnosis, and treatment of presbyacusis.
Collapse
Affiliation(s)
- Mark A Eckert
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA.
| | - Kelly C Harris
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Hainan Lang
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA
| | - Morag A Lewis
- King's College London, Wolfson Centre for Age-Related Diseases, London SE1 1UL, United Kingdom
| | - Richard A Schmiedt
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Bradley A Schulte
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA; Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Karen P Steel
- King's College London, Wolfson Centre for Age-Related Diseases, London SE1 1UL, United Kingdom
| | - Kenneth I Vaden
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA
| | - Judy R Dubno
- Medical University of South Carolina, Department of Otolaryngology - Head and Neck Surgery, Charleston, SC 29425, USA; Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC 29425, USA
| |
Collapse
|
73
|
Go V, Sarikaya D, Zhou Y, Bowley BGE, Pessina MA, Rosene DL, Zhang ZG, Chopp M, Finklestein SP, Medalla M, Buller B, Moore TL. Extracellular vesicles derived from bone marrow mesenchymal stem cells enhance myelin maintenance after cortical injury in aged rhesus monkeys. Exp Neurol 2021; 337:113540. [PMID: 33264634 PMCID: PMC7946396 DOI: 10.1016/j.expneurol.2020.113540] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2020] [Revised: 11/05/2020] [Accepted: 11/24/2020] [Indexed: 12/20/2022]
Abstract
Cortical injury, such as stroke, causes neurotoxic cascades that lead to rapid death and/or damage to neurons and glia. Axonal and myelin damage in particular, are critical factors that lead to neuronal dysfunction and impair recovery of function after injury. These factors can be exacerbated in the aged brain where white matter damage is prevalent. Therapies that can ameliorate myelin damage and promote repair by targeting oligodendroglia, the cells that produce and maintain myelin, may facilitate recovery after injury, especially in the aged brain where these processes are already compromised. We previously reported that a novel therapeutic, Mesenchymal Stem Cell derived extracellular vesicles (MSC-EVs), administered intravenously at both 24 h and 14 days after cortical injury, reduced microgliosis (Go et al. 2019), reduced neuronal pathology (Medalla et al. 2020), and improved motor recovery (Moore et al. 2019) in aged female rhesus monkeys. Here, we evaluated the effect of MSC-EV treatment on changes in oligodendrocyte maturation and associated myelin markers in the sublesional white matter using immunohistochemistry, confocal microscopy, stereology, qRT-PCR, and ELISA. Compared to vehicle control monkeys, EV-treated monkeys showed a reduction in the density of damaged oligodendrocytes. Further, EV-treatment was associated with enhanced myelin maintenance, evidenced by upregulation of myelin-related genes and increases in actively myelinating oligodendrocytes in sublesional white matter. These changes in myelination correlate with the rate of motor recovery, suggesting that improved myelin maintenance facilitates this recovery. Overall, our results suggest that EVs act on oligodendrocytes to support myelination and improves functional recovery after injury in the aged brain. SIGNIFICANCE: We previously reported that EVs facilitate recovery of function after cortical injury in the aged monkey brain, while also reducing neuronal pathology (Medalla et al. 2020) and microgliosis (Go et al. 2019). However, the effect of injury and EVs on oligodendrocytes and myelination has not been characterized in the primate brain (Dewar et al. 1999; Sozmen et al. 2012; Zhang et al. 2013). In the present study, we assessed changes in myelination after cortical injury in aged monkeys. Our results show, for the first time, that MSC-EVs support recovery of function after cortical injury by enhancing myelin maintenance in the aged primate brain.
Collapse
Affiliation(s)
- Veronica Go
- Department of Pharmacology & Experimental Therapeutics, Boston University School of Medicine, United States.
| | - Deniz Sarikaya
- Research Center for Translational Medicine, Koç University School of Medicine, Turkey
| | - Yuxin Zhou
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States
| | - Bethany G E Bowley
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States
| | - Monica A Pessina
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States
| | - Douglas L Rosene
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States; Yerkes National Primate Research Center, Emory University, United States; Center for Systems Neuroscience, Boston University, United States
| | - Zheng Gang Zhang
- Department of Neurology, Henry Ford Health Systems, United States
| | - Michael Chopp
- Department of Neurology, Henry Ford Health Systems, United States; Department of Physics, Oakland University, United States
| | - Seth P Finklestein
- Department of Neurology, Massachusetts General Hospital, United States; Stemetix, Inc., United States
| | - Maria Medalla
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States; Center for Systems Neuroscience, Boston University, United States
| | - Benjamin Buller
- Department of Neurology, Henry Ford Health Systems, United States
| | - Tara L Moore
- Department of Anatomy & Neurobiology, Boston University School of Medicine, United States; Center for Systems Neuroscience, Boston University, United States
| |
Collapse
|
74
|
Fedullo AL, Ciccotti M, Giannotta P, Alviti F, Bernardi M, Raguzzini A, Toti E, Sciarra T, Peluso I. Hormetic Effects of Bioactive Compounds from Foods, Beverages, and Food Dressing: The Potential Role in Spinal Cord Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6615752. [PMID: 33747346 PMCID: PMC7943269 DOI: 10.1155/2021/6615752] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 02/13/2021] [Accepted: 02/20/2021] [Indexed: 01/18/2023]
Abstract
Spinal cord injury (SCI) is a damage or trauma to the spinal cord resulting in a total or partial loss of motor and sensory function. SCI is characterized by a disequilibrium between the production of reactive oxygen species and the levels of antioxidant defences, causing oxidative stress and neuroinflammation. This review is aimed at highlighting the hormetic effects of some compounds from foods, beverages, and food dressing that are able to reduce oxidative stress in patients with SCI. Although curcumin, ginseng, and green tea have been proposed for SCI management, low levels of antioxidant vitamins have been reported in individuals with SCI. Mediterranean diet includes food rich in vitamins and antioxidants. Moreover, food dressing, including spices, herbs, and extra virgin olive oil (EVOO), contains multiple components with hormetic effects. The latter involves the activation of the nuclear factor erythroid-derived 2, consequently increasing the antioxidant enzymes and decreasing inflammation. Furthermore, EVOO improves the bioavailability of carotenoids and could be a delivery system for bioactive compounds. In conclusion, Mediterranean dressing in addition to plant foods can have an important effect on redox balance in individuals with SCI.
Collapse
Affiliation(s)
- Anna Lucia Fedullo
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Rome, Italy
| | | | | | - Federica Alviti
- Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Board of Physical Medicine and Rehabilitation, Sapienza University of Rome, Rome, Italy
| | - Marco Bernardi
- Department of Physiology and Pharmacology “V. Erspamer”, Sapienza University of Rome, Rome 00185, Italy
| | - Anna Raguzzini
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Rome, Italy
| | - Elisabetta Toti
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Rome, Italy
| | - Tommaso Sciarra
- Joint Veteran Center, Scientific Department, Army Medical Center, Rome, Italy
| | - Ilaria Peluso
- Research Centre for Food and Nutrition, Council for Agricultural Research and Economics (CREA-AN), Rome, Italy
| |
Collapse
|
75
|
Breton JM, Long KLP, Barraza MK, Perloff OS, Kaufer D. Hormonal Regulation of Oligodendrogenesis II: Implications for Myelin Repair. Biomolecules 2021; 11:290. [PMID: 33669242 PMCID: PMC7919830 DOI: 10.3390/biom11020290] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 02/10/2021] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Alterations in myelin, the protective and insulating sheath surrounding axons, affect brain function, as is evident in demyelinating diseases where the loss of myelin leads to cognitive and motor dysfunction. Recent evidence suggests that changes in myelination, including both hyper- and hypo-myelination, may also play a role in numerous neurological and psychiatric diseases. Protecting myelin and promoting remyelination is thus crucial for a wide range of disorders. Oligodendrocytes (OLs) are the cells that generate myelin, and oligodendrogenesis, the creation of new OLs, continues throughout life and is necessary for myelin plasticity and remyelination. Understanding the regulation of oligodendrogenesis and myelin plasticity within disease contexts is, therefore, critical for the development of novel therapeutic targets. In our companion manuscript, we review literature demonstrating that multiple hormone classes are involved in the regulation of oligodendrogenesis under physiological conditions. The majority of hormones enhance oligodendrogenesis, increasing oligodendrocyte precursor cell differentiation and inducing maturation and myelin production in OLs. Thus, hormonal treatments present a promising route to promote remyelination. Here, we review the literature on hormonal regulation of oligodendrogenesis within the context of disorders. We focus on steroid hormones, including glucocorticoids and sex hormones, peptide hormones such as insulin-like growth factor 1, and thyroid hormones. For each hormone, we describe whether they aid in OL survival, differentiation, or remyelination, and we discuss their mechanisms of action, if known. Several of these hormones have yielded promising results in both animal models and in human conditions; however, a better understanding of hormonal effects, interactions, and their mechanisms will ultimately lead to more targeted therapeutics for myelin repair.
Collapse
Affiliation(s)
- Jocelyn M Breton
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Kimberly L P Long
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
| | - Matthew K Barraza
- Molecular and Cellular Biology, University of California Berkeley, Berkeley, CA 94720, USA
| | - Olga S Perloff
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Daniela Kaufer
- Helen Wills Neuroscience Institute, University of California Berkeley, Berkeley, CA 94720, USA
- Integrative Biology, University of California Berkeley, Berkeley, CA 94720, USA
- Canadian Institute for Advanced Research, Toronto, ON M5G1M1, Canada
| |
Collapse
|
76
|
Kapoor T, Mehan S. Neuroprotective Methodologies in the Treatment of Multiple Sclerosis Current Status of Clinical and Pre-clinical Findings. Curr Drug Discov Technol 2021; 18:31-46. [PMID: 32031075 DOI: 10.2174/1570163817666200207100903] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/02/2019] [Accepted: 11/26/2019] [Indexed: 11/22/2022]
Abstract
Multiple sclerosis is an idiopathic and autoimmune associated motor neuron disorder that affects myelinated neurons in specific brain regions of young people, especially females. MS is characterized by oligodendrocytes destruction further responsible for demyelination, neuroinflammation, mitochondrial abnormalities, oxidative stress and neurotransmitter deficits associated with motor and cognitive dysfunctions, vertigo and muscle weakness. The limited intervention of pharmacologically active compounds like interferon-β, mitoxantrone, fingolimod and monoclonal antibodies used clinically are majorly associated with adverse drug reactions. Pre-clinically, gliotoxin ethidium bromide mimics the behavioral and neurochemical alterations in multiple sclerosis- like in experimental animals associated with the down-regulation of adenyl cyclase/cAMP/CREB, which is further responsible for a variety of neuropathogenic factors. Despite the considerable investigation of neuroprotection in curing multiple sclerosis, some complications still remain. The available medications only provide symptomatic relief but do not stop the disease progression. In this way, the development of unused beneficial methods tends to be ignored. The limitations of the current steady treatment may be because of their activity at one of the many neurotransmitters included or their failure to up direct signaling flag bearers detailed to have a vital part in neuronal sensitivity, biosynthesis of neurotransmitters and its discharge, development, and separation of the neuron, synaptic versatility and cognitive working. Therefore, the current review strictly focused on the exploration of various clinical and pre-clinical features available for multiple sclerosis to understand the pathogenic mechanisms and to introduce pharmacological interventions associated with the upregulation of intracellular adenyl cyclase/cAMP/CREB activation to ameliorate multiple sclerosis-like features.
Collapse
Affiliation(s)
- Tarun Kapoor
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Neuropharmacology Division, ISF College of Pharmacy, Moga, Punjab, India
| |
Collapse
|
77
|
Kaul D, Schwab SG, Mechawar N, Matosin N. How stress physically re-shapes the brain: Impact on brain cell shapes, numbers and connections in psychiatric disorders. Neurosci Biobehav Rev 2021; 124:193-215. [PMID: 33556389 DOI: 10.1016/j.neubiorev.2021.01.025] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 01/20/2021] [Accepted: 01/31/2021] [Indexed: 12/16/2022]
Abstract
Severe stress is among the most robust risk factors for the development of psychiatric disorders. Imaging studies indicate that life stress is integral to shaping the human brain, especially regions involved in processing the stress response. Although this is likely underpinned by changes to the cytoarchitecture of cellular networks in the brain, we are yet to clearly understand how these define a role for stress in human psychopathology. In this review, we consolidate evidence of macro-structural morphometric changes and the cellular mechanisms that likely underlie them. Focusing on stress-sensitive regions of the brain, we illustrate how stress throughout life may lead to persistent remodelling of the both neurons and glia in cellular networks and how these may lead to psychopathology. We support that greater translation of cellular alterations to human cohorts will support parsing the psychological sequalae of severe stress and improve our understanding of how stress shapes the human brain. This will remain a critical step for improving treatment interventions and prevention outcomes.
Collapse
Affiliation(s)
- Dominic Kaul
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia
| | - Sibylle G Schwab
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia
| | - Naguib Mechawar
- Douglas Mental Health University Institute, 6875 LaSalle blvd, Verdun, Qc, H4H 1R3, Canada
| | - Natalie Matosin
- Illawarra Health and Medical Research Institute, Northfields Ave, Wollongong 2522, Australia; Molecular Horizons, School of Chemistry and Molecular Biosciences, University of Wollongong, Northfields Ave, Wollongong 2522, Australia; Max Planck Institute of Psychiatry, Kraepelinstrasse 2-10, 80804 Munich, Germany.
| |
Collapse
|
78
|
de Curtis M, Garbelli R, Uva L. A hypothesis for the role of axon demyelination in seizure generation. Epilepsia 2021; 62:583-595. [PMID: 33493363 DOI: 10.1111/epi.16824] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/04/2021] [Accepted: 01/05/2021] [Indexed: 01/06/2023]
Abstract
Loss of myelin and altered oligodendrocyte distribution in the cerebral cortex are commonly observed both in postsurgical tissue derived from different focal epilepsies (such as focal cortical dysplasias and tuberous sclerosis) and in animal models of focal epilepsy. Moreover, seizures are a frequent symptom in demyelinating diseases, such as multiple sclerosis, and in animal models of demyelination and oligodendrocyte dysfunction. Finally, the excessive activity reported in demyelinated axons may promote hyperexcitability. We hypothesize that the extracellular potassium rise generated during epileptiform activity may be amplified by the presence of axons without appropriate myelin coating and by alterations in oligodendrocyte function. This process could facilitate the triggering of recurrent spontaneous seizures in areas of altered myelination and could result in further demyelination, thus promoting epileptogenesis.
Collapse
Affiliation(s)
- Marco de Curtis
- Epilepsy Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Rita Garbelli
- Epilepsy Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy
| | - Laura Uva
- Epilepsy Unit, IRCCS Foundation Carlo Besta Neurological Institute, Milan, Italy
| |
Collapse
|
79
|
Mu J, Wang T, Li M, Guan T, Guo Y, Zhang X, Zhang G, Kong J. Ketogenic diet protects myelin and axons in diffuse axonal injury. Nutr Neurosci 2021; 25:1534-1547. [PMID: 33487123 DOI: 10.1080/1028415x.2021.1875300] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
BACKGROUND Ketogenic diet (KD) has been identified as a potential therapy to enhance recovery after traumatic brain injury (TBI). Diffuse axonal injury (DAI) is a common type of traumatic brain injury that is characterized by delayed axonal disconnection. Previous studies showed that demyelination resulting from oligodendrocyte damage contributes to axonal degeneration in DAI. AIM The present study tests a hypothesis that ketone bodies from the ketogenic diet confers protection for myelin and attenuates degeneration of demyelinated axon in DAI. METHODS A modified Marmarou's model of DAI was induced in adult rats. The DAI rats were fed with KD and analyzed with western blot, transmission electron microscope, ELISA test and immunohistochemistry. Meanwhile, a co-culture of primary oligodendrocytes and neurons was treated with ketone body β-hydroxybutryate (βHB) to test for its effects on the myelin-axon unit. RESULTS Here we report that rats fed with KD showed an increased fatty acid metabolism and ketonemia. This dietary intervention significantly reduced demyelination and attenuated axonal damage in rats following DAI, likely through inhibition of DAI-induced excessive mitochondrial fission and promoting mitochondrial fusion. In an in vitro model of myelination, the ketone body βHB increased myelination significantly and reduced axonal degeneration induced by glucose deprivation (GD). βHB robustly increased cell viability, inhibited GD-induced collapse of mitochondrial membrane potential and attenuated death of oligodendrocytes. CONCLUSION Ketone bodies protect myelin-forming oligodendrocytes and reduce axonal damage. Ketogenic diet maybe a promising therapy for DAI.
Collapse
Affiliation(s)
- Jiao Mu
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China.,Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China.,Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Tingting Wang
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China
| | - Meiyu Li
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China
| | - Teng Guan
- Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China
| | - Ying Guo
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China.,Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, People's Republic of China
| | - Xiaoli Zhang
- Department of Life Science Research Center, Hebei North University, Zhangjiakou, People's Republic of China
| | - Guohui Zhang
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China
| | - Jiming Kong
- Department of Forensic Medicine, Hebei North University, Zhangjiakou, People's Republic of China.,Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
80
|
Zangari Del Balzo G. Statistical field theory of the transmission of nerve impulses. Theor Biol Med Model 2021; 18:1. [PMID: 33407639 PMCID: PMC7787247 DOI: 10.1186/s12976-020-00132-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 10/27/2020] [Indexed: 11/23/2022] Open
Abstract
Background Stochastic processes leading voltage-gated ion channel dynamics on the nerve cell membrane are a sufficient condition to describe membrane conductance through statistical mechanics of disordered and complex systems. Results Voltage-gated ion channels in the nerve cell membrane are described by the Ising model. Stochastic circuit elements called “Ising Neural Machines” are introduced. Action potentials are described as quasi-particles of a statistical field theory for the Ising system. Conclusions The particle description of action potentials is a new point of view and a powerful tool to describe the generation and propagation of nerve impulses, especially when classical electrophysiological models break down. The particle description of action potentials allows us to develop a new generation of devices to study neurodegenerative and demyelinating diseases as Multiple Sclerosis and Alzheimer’s disease, even integrated by connectomes. It is also suitable for the study of complex networks, quantum computing, artificial intelligence, machine and deep learning, cryptography, ultra-fast lines for entanglement experiments and many other applications of medical, physical and engineering interest.
Collapse
|
81
|
Al-Gholam MA, Issa NM. Effect of cypermethrin on the postnatal development of the medulla oblongata and the possible protective role of melatonin in albino rats. Anat Cell Biol 2020; 53:460-470. [PMID: 33361544 PMCID: PMC7769100 DOI: 10.5115/acb.20.193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 09/01/2020] [Accepted: 09/16/2020] [Indexed: 11/27/2022] Open
Abstract
Previous studies have shown that cypermethrin (CYP), a broad spectrum pesticide has a teratogenic effect on rat offspring born to an exposed dam with no information on its effect on the development of the brain. To the best of our knowledge, this research is the first attempt to study the postnatal development medulla oblongata of rat offspring exposed to CYP during the perinatal period and the possible neuroprotective role of melatonin. The offspring of treated female rats were organized into control, melatonin (1 mg/kg/day orally); CYP (12 mg/kg/day orally); and CYP/melatonin groups. The mothers received treatments from day 6 of gestation until day 21 after birth. At Postnatal days 7 and 21, the animals were sacrificed and their medulla oblongata was removed and subjected to histological, immunohistochemical, and electron microscopic studies. CYP induced neuronal degeneration by chromatolysis and pyknosis. Nuclear changes, cytoplasmic vacuolation, damage mitochondria, and breakdown of RER were also detected. Reduction of microtubule-associated protein-2 (MAP-2), myelin basic protein (MBP), and oligodendrocyte transcription factor expressions and increment of glial fibrillary acidic protein expression in the medulla oblongata of the developing rats were observed. On the other hand, melatonin led to an obvious improvement of the injured medulla oblongata tissues and ameliorating the damaging effects of CYP. In conclusion, melatonin has protected rats against CYP-induced histopathological and immunohistochemical changes. This may be due to the protection of MAP-2, conservation of MBP, an increment of oligodendrocytes, and alleviation of astrogliosis.
Collapse
Affiliation(s)
- Marwa A Al-Gholam
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Shebin El-Kom, Egypt
| | - Noha M Issa
- Department of Anatomy and Embryology, Faculty of Medicine, Menoufia University, Shebin El-Kom, Egypt
| |
Collapse
|
82
|
Kim HK, Lee DW, Kim E, Jeong I, Kim S, Kim BJ, Park HC. Notch Signaling Controls Oligodendrocyte Regeneration in the Injured Telencephalon of Adult Zebrafish. Exp Neurobiol 2020; 29:417-424. [PMID: 33281119 PMCID: PMC7788307 DOI: 10.5607/en20050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 12/19/2022] Open
Abstract
The myelination of axons in the vertebrate nervous system through oligodendrocytes promotes efficient axonal conduction, which is required for the normal function of neurons. The central nervous system (CNS) can regenerate damaged myelin sheaths through the process of remyelination, but the failure of remyelination causes neurological disorders such as multiple sclerosis. In mammals, parenchymal oligodendrocyte progenitor cells (OPCs) are known to be the principal cell type responsible for remyelination in demyelinating diseases and traumatic injuries to the adult CNS. However, growing evidence suggests that neural stem cells (NSCs) are implicated in remyelination in animal models of demyelination. We have previously shown that olig2+ radial glia (RG) have the potential to function as NSCs to produce oligodendrocytes in adult zebrafish. In this study, we developed a zebrafish model of adult telencephalic injury to investigate cellular and molecular mechanisms underlying the regeneration of oligodendrocytes. Using this model, we showed that telencephalic injury induced the proliferation of olig2+ RG and parenchymal OPCs shortly after injury, which was followed by the regeneration of new oligodendrocytes in the adult zebrafish. We also showed that blocking Notch signaling promoted the proliferation of olig2+ RG and OPCs in the normal and injured telencephalon of adult zebrafish. Taken together, our data suggest that Notch-regulated proliferation of olig2+ RG and parenchymal OPCs is responsible for the regeneration of oligodendrocytes in the injured telencephalon of adult zebrafish.
Collapse
Affiliation(s)
- Hwan-Ki Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan 15355, Korea
| | - Dong-Won Lee
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan 15355, Korea
| | - Eunmi Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan 15355, Korea
| | - Inyoung Jeong
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan 15355, Korea
| | - Suhyun Kim
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan 15355, Korea
| | - Bum-Joon Kim
- Department of Neurosurgery, Korea University Ansan Hospital, Ansan 15355, Korea
| | - Hae-Chul Park
- Department of Biomedical Sciences, College of Medicine, Korea University, Ansan 15355, Korea
| |
Collapse
|
83
|
Feng T, Sheng RR, Solé-Domènech S, Ullah M, Zhou X, Mendoza CS, Enriquez LCM, Katz II, Paushter DH, Sullivan PM, Wu X, Maxfield FR, Hu F. A role of the frontotemporal lobar degeneration risk factor TMEM106B in myelination. Brain 2020; 143:2255-2271. [PMID: 32572497 DOI: 10.1093/brain/awaa154] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 03/04/2020] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
TMEM106B encodes a lysosomal membrane protein and was initially identified as a risk factor for frontotemporal lobar degeneration. Recently, a dominant D252N mutation in TMEM106B was shown to cause hypomyelinating leukodystrophy. However, how TMEM106B regulates myelination is still unclear. Here we show that TMEM106B is expressed and localized to the lysosome compartment in oligodendrocytes. TMEM106B deficiency in mice results in myelination defects with a significant reduction of protein levels of proteolipid protein (PLP) and myelin oligodendrocyte glycoprotein (MOG), the membrane proteins found in the myelin sheath. The levels of many lysosome proteins are significantly decreased in the TMEM106B-deficient Oli-neu oligodendroglial precursor cell line. TMEM106B physically interacts with the lysosomal protease cathepsin D and is required to maintain proper cathepsin D levels in oligodendrocytes. Furthermore, we found that TMEM106B deficiency results in lysosome clustering in the perinuclear region and a decrease in lysosome exocytosis and cell surface PLP levels. Moreover, we found that the D252N mutation abolished lysosome enlargement and lysosome acidification induced by wild-type TMEM106B overexpression. Instead, it stimulates lysosome clustering near the nucleus as seen in TMEM106B-deficient cells. Our results support that TMEM106B regulates myelination through modulation of lysosome function in oligodendrocytes.
Collapse
Affiliation(s)
- Tuancheng Feng
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University Ithaca, NY 14853, USA
| | - Rory R Sheng
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University Ithaca, NY 14853, USA
| | | | - Mohammed Ullah
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University Ithaca, NY 14853, USA
| | - Xiaolai Zhou
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University Ithaca, NY 14853, USA
| | - Christina S Mendoza
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University Ithaca, NY 14853, USA
| | - Laura Camila Martinez Enriquez
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University Ithaca, NY 14853, USA
| | - Isabel Iscol Katz
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University Ithaca, NY 14853, USA
| | - Daniel H Paushter
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University Ithaca, NY 14853, USA
| | - Peter M Sullivan
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University Ithaca, NY 14853, USA
| | - Xiaochun Wu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University Ithaca, NY 14853, USA
| | - Frederick R Maxfield
- Department of Biochemistry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Fenghua Hu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University Ithaca, NY 14853, USA
| |
Collapse
|
84
|
Cellular senescence and failure of myelin repair in multiple sclerosis. Mech Ageing Dev 2020; 192:111366. [DOI: 10.1016/j.mad.2020.111366] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 08/10/2020] [Accepted: 09/23/2020] [Indexed: 01/10/2023]
|
85
|
The Role of Extracellular Vesicles in Demyelination of the Central Nervous System. Int J Mol Sci 2020; 21:ijms21239111. [PMID: 33266211 PMCID: PMC7729475 DOI: 10.3390/ijms21239111] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 11/27/2020] [Accepted: 11/28/2020] [Indexed: 12/21/2022] Open
Abstract
It is being increasingly demonstrated that extracellular vesicles (EVs) are deeply involved in the physiology of the central nervous system (CNS). Processes such as synaptic activity, neuron-glia communication, myelination and immune response are modulated by EVs. Likewise, these vesicles may participate in many pathological processes, both as triggers of disease or, on the contrary, as mechanisms of repair. EVs play relevant roles in neurodegenerative disorders such as Alzheimer’s or Parkinson’s diseases, in viral infections of the CNS and in demyelinating pathologies such as multiple sclerosis (MS). This review describes the involvement of these membrane vesicles in major demyelinating diseases, including MS, neuromyelitis optica, progressive multifocal leukoencephalopathy and demyelination associated to herpesviruses.
Collapse
|
86
|
van der Weijden CWJ, García DV, Borra RJH, Thurner P, Meilof JF, van Laar PJ, Dierckx RAJO, Gutmann IW, de Vries EFJ. Myelin quantification with MRI: A systematic review of accuracy and reproducibility. Neuroimage 2020; 226:117561. [PMID: 33189927 DOI: 10.1016/j.neuroimage.2020.117561] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/27/2020] [Accepted: 11/07/2020] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVES Currently, multiple sclerosis is treated with anti-inflammatory therapies, but these treatments lack efficacy in progressive disease. New treatment strategies aim to repair myelin damage and efficacy evaluation of such new therapies would benefit from validated myelin imaging techniques. Several MRI methods for quantification of myelin density are available now. This systematic review aims to analyse the performance of these MRI methods. METHODS Studies comparing myelin quantification by MRI with histology, the current gold standard, or assessing reproducibility were retrieved from PubMed/MEDLINE and Embase (until December 2019). Included studies assessed both myelin histology and MRI quantitatively. Correlation or variance measurements were extracted from the studies. Non-parametric tests were used to analyse differences in study methodologies. RESULTS The search yielded 1348 unique articles. Twenty-two animal studies and 13 human studies correlated myelin MRI with histology. Eighteen clinical studies analysed the reproducibility. Overall bias risk was low or unclear. All MRI methods performed comparably, with a mean correlation between MRI and histology of R2=0.54 (SD=0.30) for animal studies, and R2=0.54 (SD=0.18) for human studies. Reproducibility for the MRI methods was good (ICC=0.75-0.93, R2=0.90-0.98, COV=1.3-27%), except for MTR (ICC=0.05-0.51). CONCLUSIONS Overall, MRI-based myelin imaging methods show a fairly good correlation with histology and a good reproducibility. However, the amount of validation data is too limited and the variability in performance between studies is too large to select the optimal MRI method for myelin quantification yet.
Collapse
Affiliation(s)
- Chris W J van der Weijden
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| | - David Vállez García
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands; Department of Radiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| | - Ronald J H Borra
- Department of Radiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| | - Patrick Thurner
- Universitätsklinik für Radiologie und Nuklearmedizin, Medizinische Universität Wien, Währinger Gürtel 18-20, 1090 Wien, Austria.
| | - Jan F Meilof
- Multiple Sclerosis Center Noord Nederland, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| | - Peter-Jan van Laar
- Department of Radiology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands; Department of Radiology, Zorggroep Twente, Zilvermeeuw 1, 7609 PP Almelo, the Netherlands.
| | - Rudi A J O Dierckx
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| | - Ingomar W Gutmann
- Physics of Functional Material, Faculty of Physics, University of Vienna, Boltzmanngasse 5, 1090 Vienna, Austria.
| | - Erik F J de Vries
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, the Netherlands.
| |
Collapse
|
87
|
Koudriavtseva T, Stefanile A, Fiorelli M, Lapucci C, Lorenzano S, Zannino S, Conti L, D'Agosto G, Pimpinelli F, Di Domenico EG, Mandoj C, Giannarelli D, Donzelli S, Blandino G, Salvetti M, Inglese M. Coagulation/Complement Activation and Cerebral Hypoperfusion in Relapsing-Remitting Multiple Sclerosis. Front Immunol 2020; 11:548604. [PMID: 33193314 PMCID: PMC7655134 DOI: 10.3389/fimmu.2020.548604] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
Introduction Multiple sclerosis (MS) is a demyelinating disease of the central nervous system with an underlying immune-mediated and inflammatory pathogenesis. Innate immunity, in addition to the adaptive immune system, plays a relevant role in MS pathogenesis. It represents the immediate non-specific defense against infections through the intrinsic effector mechanism “immunothrombosis” linking inflammation and coagulation. Moreover, decreased cerebral blood volume (CBV), cerebral blood flow (CBF), and prolonged mean transit time (MTT) have been widely demonstrated by MRI in MS patients. We hypothesized that coagulation/complement and platelet activation during MS relapse, likely during viral infections, could be related to CBF decrease. Our specific aims are to evaluate whether there are differences in serum/plasma levels of coagulation/complement factors between relapsing-remitting (RR) MS patients (RRMS) in relapse and those in remission and healthy controls as well as to assess whether brain hemodynamic changes detected by MRI occur in relapse compared with remission. This will allow us to correlate coagulation status with perfusion and demographic/clinical features in MS patients. Materials and Methods This is a multi-center, prospective, controlled study. RRMS patients (1° group: 30 patients in relapse; 2° group: 30 patients in remission) and age/sex-matched controls (3° group: 30 subjects) will be enrolled in the study. Patients and controls will be tested for either coagulation/complement (C3, C4, C4a, C9, PT, aPTT, fibrinogen, factor II, VIII, and X, D-dimer, antithrombin, protein C, protein S, von-Willebrand factor), soluble markers of endothelial damage (thrombomodulin, Endothelial Protein C Receptor), antiphospholipid antibodies, lupus anticoagulant, complete blood count, viral serological assays, or microRNA microarray. Patients will undergo dynamic susceptibility contrast-enhanced MRI using a 3.0-T scanner to evaluate CBF, CBV, MTT, lesion number, and volume. Statistical Analysis ANOVA and unpaired t-tests will be used. The level of significance was set at p ≤ 0.05. Discussion Identifying a link between activation of coagulation/complement system and cerebral hypoperfusion could improve the identification of novel molecular and/or imaging biomarkers and targets, leading to the development of new effective therapeutic strategies in MS. Clinical Trial Registration Clinicaltrials.gov, identifier NCT04380220.
Collapse
Affiliation(s)
- Tatiana Koudriavtseva
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Annunziata Stefanile
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marco Fiorelli
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Caterina Lapucci
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Svetlana Lorenzano
- Department of Human Neurosciences, Sapienza University of Rome, Rome, Italy
| | - Silvana Zannino
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Laura Conti
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanna D'Agosto
- Clinical Pathology and Microbiology Unit, IRCC San Gallicano Institute, Rome, Italy
| | - Fulvia Pimpinelli
- Clinical Pathology and Microbiology Unit, IRCC San Gallicano Institute, Rome, Italy
| | | | - Chiara Mandoj
- Department of Clinical Experimental Oncology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Diana Giannarelli
- Biostatistics, Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Sara Donzelli
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Giovanni Blandino
- Oncogenomic and Epigenetic Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marco Salvetti
- Department of Neuroscience Mental Health and Sensory Organs (NEMOS), Sapienza University, Sant'Andrea Hospital, Rome, Italy
| | - Matilde Inglese
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy.,Department of Neurology, Radiology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| |
Collapse
|
88
|
Dcf1 deficiency induces hypomyelination by activating Wnt signaling. Exp Neurol 2020; 335:113486. [PMID: 32991932 DOI: 10.1016/j.expneurol.2020.113486] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/06/2020] [Accepted: 09/25/2020] [Indexed: 02/01/2023]
Abstract
Myelination is extremely important in achieving neural function. Hypomyelination causes a variety of neurological diseases. However, little is known about how hypomyelination occurs. Here we investigated the effect of dendritic cell factor 1(Dcf1) on myelination, using in vitro and in vivo models and found that Dcf1 is essential for normal myelination, motor coordination and balance. Lack of Dcf1 downregulated myelin-associated proteins, such as myelin basic protein (MBP), myelin associated glycoprotein (MAG), and 2'3'-cyclic nucleotide 3'-phosphodiesterase (CNPase) in the hippocampus and corpus callosum of Dcf1-null mice, as a result, the myelin sheath of these mice became thinner. Transmission electron microscopy revealed hypomyelination in Dcf1-deficient mice. Motor coordination and balance tests confirmed impaired neurological function in Dcf1-null mice. Gain-of-function analysis via In utero electroporation showed that hypomyelination could be rescued by re-expression of Dcf1 in Dcf1-null mouse brain. Dcf1-null mice exhibited a phenotype similar to that of cuprizone-induced demyelinated mice, thereby supporting the finding of hypomyelination caused by Dcf1 knockout. Mechanistically, we further revealed that insufficient Dcf1 leads to hyperactivation of the Wnt/β-catenin signaling pathway. Our work describes the role of Dcf1 in maintaining normal myelination, and this could help improve the current understanding of hypomyelination and its pathogenesis.
Collapse
|
89
|
Spontaneous Local Calcium Transients Regulate Oligodendrocyte Development in Culture through Store-Operated Ca 2+ Entry and Release. eNeuro 2020; 7:ENEURO.0347-19.2020. [PMID: 32409508 PMCID: PMC7438061 DOI: 10.1523/eneuro.0347-19.2020] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 04/30/2020] [Accepted: 05/06/2020] [Indexed: 12/14/2022] Open
Abstract
Oligodendrocytes (OLs) insulate axonal fibers for fast conduction of nerve impulses by wrapping axons of the CNS with compact myelin membranes. Differentiating OLs undergo drastic chances in cell morphology. Bipolar oligodendroglial precursor cells (OPCs) transform into highly ramified multipolar OLs, which then expand myelin membranes that enwrap axons. While significant progress has been made in understanding the molecular and genetic mechanisms underlying CNS myelination and its disruption in diseases, the cellular mechanisms that regulate OL differentiation are not fully understood. Here, we report that developing rat OLs in culture exhibit spontaneous Ca2+ local transients (sCaLTs) in their process arbors in the absence of neurons. Importantly, we find that the frequency of sCaLTs markedly increases as OLs undergo extensive process outgrowth and branching. We further show that sCaLTs are primarily generated through a combination of Ca2+ influx through store-operated Ca2+ entry (SOCE) and Ca2+ release from internal Ca2+ stores. Inhibition of sCaLTs impairs the elaboration and branching of OL processes, as well as substantially reduces the formation of large myelin sheets in culture. Together, our findings identify an important role for spontaneous local Ca2+ signaling in OL development.
Collapse
|
90
|
Vacca V, Madaro L, De Angelis F, Proietti D, Cobianchi S, Orsini T, Puri PL, Luvisetto S, Pavone F, Marinelli S. Revealing the Therapeutic Potential of Botulinum Neurotoxin Type A in Counteracting Paralysis and Neuropathic Pain in Spinally Injured Mice. Toxins (Basel) 2020; 12:E491. [PMID: 32751937 PMCID: PMC7472120 DOI: 10.3390/toxins12080491] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/21/2022] Open
Abstract
Botulinum neurotoxin type A (BoNT/A) is a major therapeutic agent that has been proven to be a successful treatment for different neurological disorders, with emerging novel therapeutic indications each year. BoNT/A exerts its action by blocking SNARE complex formation and vesicle release through the specific cleavage of SNAP-25 protein; the toxin is able to block the release of pro-inflammatory molecules for months after its administration. Here we demonstrate the extraordinary capacity of BoNT/A to neutralize the complete paralysis and pain insensitivity induced in a murine model of severe spinal cord injury (SCI). We show that the toxin, spinally administered within one hour from spinal trauma, exerts a long-lasting proteolytic action, up to 60 days after its administration, and induces a complete recovery of muscle and motor function. BoNT/A modulates SCI-induced neuroglia hyperreactivity, facilitating axonal restoration, and preventing secondary cells death and damage. Moreover, we demonstrate that BoNT/A affects SCI-induced neuropathic pain after moderate spinal contusion, confirming its anti-nociceptive action in this kind of pain, as well. Our results provide the intriguing and real possibility to identify in BoNT/A a therapeutic tool in counteracting SCI-induced detrimental effects. Because of the well-documented BoNT/A pharmacology, safety, and toxicity, these findings strongly encourage clinical translation.
Collapse
Affiliation(s)
- Valentina Vacca
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, 00015 Monterotondo Scalo (RM), Italy; (V.V.); (T.O.); (S.L.)
- IRCCS Santa Lucia Foundation, 00143 Roma, Italy; (L.M.); (F.D.A.); (D.P.)
| | - Luca Madaro
- IRCCS Santa Lucia Foundation, 00143 Roma, Italy; (L.M.); (F.D.A.); (D.P.)
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | | | - Daisy Proietti
- IRCCS Santa Lucia Foundation, 00143 Roma, Italy; (L.M.); (F.D.A.); (D.P.)
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Stefano Cobianchi
- Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autonoma de Barcelona, E-08193 Bellaterra, Spain;
| | - Tiziana Orsini
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, 00015 Monterotondo Scalo (RM), Italy; (V.V.); (T.O.); (S.L.)
| | - Pier Lorenzo Puri
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA;
| | - Siro Luvisetto
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, 00015 Monterotondo Scalo (RM), Italy; (V.V.); (T.O.); (S.L.)
| | - Flaminia Pavone
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, 00015 Monterotondo Scalo (RM), Italy; (V.V.); (T.O.); (S.L.)
- IRCCS Santa Lucia Foundation, 00143 Roma, Italy; (L.M.); (F.D.A.); (D.P.)
| | - Sara Marinelli
- CNR—National Research Council, Institute of Biochemistry and Cell Biology, 00015 Monterotondo Scalo (RM), Italy; (V.V.); (T.O.); (S.L.)
- IRCCS Santa Lucia Foundation, 00143 Roma, Italy; (L.M.); (F.D.A.); (D.P.)
| |
Collapse
|
91
|
The ameliorative effects of a phenolic derivative of Moringa oleifera leave against vanadium-induced neurotoxicity in mice. IBRO Rep 2020; 9:164-182. [PMID: 32803016 PMCID: PMC7417907 DOI: 10.1016/j.ibror.2020.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 07/03/2020] [Indexed: 12/29/2022] Open
Abstract
Vanadium, a transition series metal released during some industrial activities, induces oxidative stress and lipid peroxidation. Ameliorative effect of a pure compound from the methanolic extract of Moringa oleifera leaves, code-named MIMO2, in 14-day old mice administered with vanadium (as sodium metavanadate 3 mg/kg) for 2 weeks was assessed. Results from body weight monitoring, muscular strength, and open field showed slight reduction in body weight and locomotion deficit in vanadium-exposed mice, ameliorated with MIMO2 co-administration. Degeneration of the Purkinje cell layer and neuronal death in the hippocampal CA1 region were observed in vanadium-exposed mice and both appeared significantly reduced with MIMO2 co-administration. Demyelination involving the midline of the corpus callosum, somatosensory and retrosplenial cortices was also reduced with MIMO2. Microglia activation and astrogliosis observed through immunohistochemistry were also alleviated. Immunohistochemistry for myelin, axons and oligodendrocyte lineage cells were also carried out and showed that in vanadium-treated mice brains, oligodendrocyte progenitor cells increased NG2 immunolabelling with hypertrophy and bushy, ramified appearance of their processes. MIMO2 displayed ameliorative and antioxidative effects in vanadium-induced neurotoxicity in experimental murine species. This is likely the first time MIMO2 is being used in vivo in an animal model.
Collapse
|
92
|
Wellman SM, Guzman K, Stieger KC, Brink LE, Sridhar S, Dubaniewicz MT, Li L, Cambi F, Kozai TDY. Cuprizone-induced oligodendrocyte loss and demyelination impairs recording performance of chronically implanted neural interfaces. Biomaterials 2020; 239:119842. [PMID: 32065972 PMCID: PMC7540937 DOI: 10.1016/j.biomaterials.2020.119842] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Revised: 01/29/2020] [Accepted: 02/03/2020] [Indexed: 12/16/2022]
Abstract
Biological inflammation induced during penetrating cortical injury can disrupt functional neuronal and glial activity within the cortex, resulting in potential recording failure of chronically implanted neural interfaces. Oligodendrocytes provide critical support for neuronal health and function through direct contact with neuronal soma and axons within the cortex. Given their fundamental role to regulate neuronal activity via myelin, coupled with their heightened vulnerability to metabolic brain injury due to high energetic demands, oligodendrocytes are hypothesized as a possible source of biological failure in declining recording performances of intracortical microelectrode devices. To determine the extent of their contribution to neuronal activity and function, a cuprizone-inducible model of oligodendrocyte depletion and demyelination in mice was performed prior to microelectrode implantation. At 5 weeks of cuprizone exposure, mice demonstrated significantly reduced cortical oligodendrocyte density and myelin expression. Mice were then implanted with functional recording microelectrodes in the visual cortex and neuronal activity was evaluated up to 7 weeks alongside continued cuprizone administration. Cuprizone-induced oligodendrocyte loss and demyelination was associated with significantly reduced recording performances at the onset of implantation, which remained relatively stable over time. In contast, recording performances for mice on a normal diet were intially elevated before decreasing over time to the recording level of tcuprizone-treated mice. Further electrophysiological analysis revealed deficits in multi-unit firing rates, frequency-dependent disruptions in neuronal oscillations, and altered laminar communication within the cortex of cuprizone-treated mice. Post-mortem immunohistochemistry revealed robust depletion of oligodendrocytes around implanted microelectrode arrays alongside comparable neuronal densities to control mice, suggesting that oligodendrocyte loss was a possible contributor to chronically impaired device performances. This study highlights potentially significant contributions from the oligodendrocyte lineage population concerning the biological integration and long-term functional performance of neural interfacing technology.
Collapse
Affiliation(s)
- Steven M Wellman
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | - Kelly Guzman
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA
| | - Kevin C Stieger
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA
| | | | - Sadhana Sridhar
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | | | - Lehong Li
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Franca Cambi
- Veterans Administration Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurology, University of Kentucky, Lexington, KY, USA
| | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA; Center for Neural Basis of Cognition, Pittsburgh, PA, USA; Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, USA; McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA; NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, USA.
| |
Collapse
|
93
|
Kang W, Nguyen KCQ, Hébert JM. Transient Redirection of SVZ Stem Cells to Oligodendrogenesis by FGFR3 Activation Promotes Remyelination. Stem Cell Reports 2020; 12:1223-1231. [PMID: 31189094 PMCID: PMC6565886 DOI: 10.1016/j.stemcr.2019.05.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 05/06/2019] [Accepted: 05/07/2019] [Indexed: 01/02/2023] Open
Abstract
Stimulating oligodendrocyte (OL) production from endogenous progenitor cells is an important strategy for myelin repair and functional restoration after disease or injury-induced demyelination. Subventricular zone (SVZ) stem cells are multipotential, generating neurons and oligodendroglia. The factors that regulate the fate of these stem cells are poorly defined. In this study, we show that genetically increasing fibroblast growth factor receptor-3 (FGFR3) activity in adult SVZ stem cells transiently and dramatically redirects their differentiation from the neuronal to the oligodendroglial lineage after pathological demyelination. The increased SVZ-derived oligodendrogenesis leads to improved OL regeneration and myelin repair, not only in the corpus callosum (a normal destination for SVZ-derived oligodendroglial cells), but also in the lower cortical layers. This study identifies FGF signaling as a potent target for improving endogenous SVZ-derived OL regeneration and remyelination. Adult neuronal progenitors with increased FGFR activity switch to gliogenesis in vivo FGFR-induced increase in OPCs (30-fold) and oligodendrocytes (10-fold) is reversible FGFR-induced increase in oligodendrocytes results in remyelination after chronic insult
Collapse
Affiliation(s)
- Wenfei Kang
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Ken C Q Nguyen
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jean M Hébert
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Genetics, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
94
|
de Jong CGHM, Gabius HJ, Baron W. The emerging role of galectins in (re)myelination and its potential for developing new approaches to treat multiple sclerosis. Cell Mol Life Sci 2020; 77:1289-1317. [PMID: 31628495 PMCID: PMC7113233 DOI: 10.1007/s00018-019-03327-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory, demyelinating and neurodegenerative disease of the central nervous system with unknown etiology. Currently approved disease-modifying treatment modalities are immunomodulatory or immunosuppressive. While the applied drugs reduce the frequency and severity of the attacks, their efficacy to regenerate myelin membranes and to halt disease progression is limited. To achieve such therapeutic aims, understanding biological mechanisms of remyelination and identifying factors that interfere with remyelination in MS can give respective directions. Such a perspective is given by the emerging functional profile of galectins. They form a family of tissue lectins, which are potent effectors in processes as diverse as adhesion, apoptosis, immune mediator release or migration. This review focuses on endogenous and exogenous roles of galectins in glial cells such as oligodendrocytes, astrocytes and microglia in the context of de- and (re)myelination and its dysregulation in MS. Evidence is arising for a cooperation among family members so that timed expression and/or secretion of galectins-1, -3 and -4 result in modifying developmental myelination, (neuro)inflammatory processes, de- and remyelination. Dissecting the mechanisms that underlie the distinct activities of galectins and identifying galectins as target or tool to modulate remyelination have the potential to contribute to the development of novel therapeutic strategies for MS.
Collapse
Affiliation(s)
- Charlotte G H M de Jong
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands
| | - Hans-Joachim Gabius
- Institute of Physiological Chemistry, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Wia Baron
- Department of Biomedical Sciences of Cells & Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, A. Deusinglaan 1, 9713 AV, Groningen, The Netherlands.
| |
Collapse
|
95
|
Arpan I, Fino PC, Fling BW, Horak F. Local dynamic stability during long-fatiguing walks in people with multiple sclerosis. Gait Posture 2020; 76:122-127. [PMID: 31760315 DOI: 10.1016/j.gaitpost.2019.10.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 09/18/2019] [Accepted: 10/23/2019] [Indexed: 02/02/2023]
Abstract
BACKGROUND Altered balance/stability during walking is common in people with multiple sclerosis (PwMS). While dynamic gait stability has been related to falling and localized muscle fatigue, it has rarely been studied in MS. Specifically, the effects of walking-related fatigue on dynamic stability are unclear in PwMS. RESEARCH QUESTIONS 1) Are temporal changes in dynamic stability during long-walks different among PwMS and healthy controls (HC)? 2) Is there a relationship between stability and walking performance changes in PwMS? METHODS Twenty-five PwMS and ten HC participated in the six-minute walk test (6MWT) wearing six-wireless inertial sensors. Local dynamic stability (LDS) during gait was quantified by maximum-finite-time Lyapunov exponents (λS), where larger λS indicates less stable dynamics. Linear mixed models were fit to compare changes in LDS and walking performance over time among two groups. Additionally, the percent changes in λS and distance from minute 1 to 6 were recorded as Dynamic Stability Index (DSI6-1) and Distance-Walked Index (DWI6-1) respectively. Finally, Pearson correlation compared the association between DSI6-1 and DWI6-1. RESULTS A significant group*time interaction was found for LDS. PwMS did not have different LDS than HC until minute-4 of walking, and differences persisted at minute-6. Further, PwMS walked significantly shorter distances and demonstrated a greater decline in walking performance (DWI6-1) during the 6MWT. Finally, DSI6-1 and DWI6-1 were significantly correlated in PwMS. Significance The dynamic stability differences among PwMS and HC were only apparent after 3-minutes of walking and ∼60% of PwMS became less stable over time, supporting the use of long walks in MS to capture stability changes during the motor task performance. A significant relationship between the decline in stability and poor walking performance over time during the 6MWT suggested a possible role of walking-related fatigue in the worsening of balance during long walks in PwMS.
Collapse
Affiliation(s)
- I Arpan
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States; Advanced Imaging Center, Oregon Health & Science University, Portland, OR, United States.
| | - P C Fino
- Department of Health, Kinesiology, & Recreation, University of Utah, Salt Lake City, UT, United States
| | - B W Fling
- Department of Health and Exercise Science, Colorado State University, Fort Collins, CO, United States
| | - F Horak
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
96
|
Abstract
Schwann cells as glial cells in the peripheral nervous system can participate in neurons protection and forming myelin. Additionally, they are important for nerve pulse conduction supporting along axons. On the other hand, it was demonstrated that they are promising cells for the treatment of demyelinating disorders and also central nervous system damages. Herein, for therapeutic application, Schwann cells should be manufactured based on good manufacturing practice standards to achieve safe and effective clinical products. In this respect, the current chapter tries to introduce a standard protocol for manufacturing of human GMP-compliant Schwann cells for clinical application.
Collapse
|
97
|
Nguyen LH, Ong W, Wang K, Wang M, Nizetic D, Chew SY. Effects of miR-219/miR-338 on microglia and astrocyte behaviors and astrocyte-oligodendrocyte precursor cell interactions. Neural Regen Res 2020; 15:739-747. [PMID: 31638099 PMCID: PMC6975139 DOI: 10.4103/1673-5374.266922] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
MiR-219 and miR-338 (miR-219/miR-338) are oligodendrocyte-specific microRNAs. The overexpression of these miRs in oligodendrocyte precursor cells promotes their differentiation and maturation into oligodendrocytes, which may enhance axonal remyelination after nerve injuries in the central nervous system (CNS). As such, the delivery of miR-219/miR-338 to the CNS to promote oligodendrocyte precursor cell differentiation, maturation and myelination could be a promising approach for nerve repair. However, nerve injuries in the CNS also involve other cell types, such as microglia and astrocytes. Herein, we investigated the effects of miR-219/miR-338 treatment on microglia and astrocytes in vitro and in vivo. We found that miR-219/miR-338 diminished microglial expression of pro-inflammatory cytokines and suppressed astrocyte activation. In addition, we showed that miR-219/miR-338 enhanced oligodendrocyte precursor cell differentiation and maturation in a scratch assay paradigm that re-created a nerve injury condition in vitro. Collectively, our results suggest miR-219/miR-338 as a promising treatment for axonal remyelination in the CNS following nerve injuries. All experimental procedures were approved by the Institutional Animal Care and Use Committee (IACUC), Nanyang Technological University (approval No. A0309 and A0333) on April 27, 2016 and October 8, 2016.
Collapse
Affiliation(s)
- Lan Huong Nguyen
- School of Chemical and Biomedical Engineering, Nanyang Technological University; Current address: NUS Synthetic Biology for Clinical and Technological Innovation (SynCTI), Life Sciences Institute, National University of Singapore, Singapore
| | - William Ong
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Kai Wang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Mingfeng Wang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore
| | - Dean Nizetic
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore; Blizard Institute, Barts & The London School of Medicine, Queen Mary University of London, London, UK
| | - Sing Yian Chew
- School of Chemical and Biomedical Engineering; Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| |
Collapse
|
98
|
Vancamp P, Demeneix BA, Remaud S. Monocarboxylate Transporter 8 Deficiency: Delayed or Permanent Hypomyelination? Front Endocrinol (Lausanne) 2020; 11:283. [PMID: 32477268 PMCID: PMC7237703 DOI: 10.3389/fendo.2020.00283] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/15/2020] [Indexed: 12/11/2022] Open
Abstract
Monocarboxylate transporter 8 (MCT8) deficiency or the Allan-Herndon-Dudley Syndrome (AHDS) is an X-linked psychomotor disability syndrome with around 320 clinical cases described worldwide. SLC16A2 gene mutations, encoding the thyroid hormone (TH) transporter MCT8, result in intellectual disability due to impaired TH uptake in the developing brain. MCT8 deficiency is a multi-organ affecting disease with a predominant neuronal cell-based pathology, with the glial component inadequately investigated. However, deficiency in myelin, a key component of white matter (WM) enabling fast nerve conduction, is a TH-dependent hallmark of the disease. Nevertheless, analysis of the myelin status in AHDS patients has led to conflicting interpretations. The majority of individual case studies reported delayed myelination, that was restored later in life. In contrast, post-mortem studies and high-resolution MRIs detected WM (micro-) abnormalities throughout adolescence, suggesting permanent hypomyelination. Thus, interpretations vary depending on methodology to investigate WM microstructure. Further, it is unknown whether the mutation within the MCT8 is linked to the severity of the myelin deficiency. Consequently, terminology is inconsistent among reports, and AHDS is occasionally misdiagnosed as another WM disorder. The evolutionary conserved TH signaling pathway that promotes the generation of myelinating oligodendrocytes enabled deciphering how the lack of MCT8 might affect myelinogenesis. Linking patient findings on myelination to those obtained from models of MCT8 deficiency revealed underlying pathophysiological mechanisms, but knowledge gaps remain, notably how myelination progresses both spatially and temporally in MCT8 deficiency. This limits predicting how myelin integrity might benefit therapeutically, and when to initiate. A recurrent observation in clinical trials is the absence of neurological improvement. Testing MCT8-independent thyromimetics in models, and evaluating treatments used in other demyelinating diseases, despite different etiologies, is crucial to propose new therapeutic strategies combatting this devastating disease.
Collapse
Affiliation(s)
- Pieter Vancamp
- UMR 7221 Molecular Physiology and Adaptation, Centre National de le Recherche Scientifique-Muséum National d'Histoire Naturelle, Paris, France
| | - Barbara A Demeneix
- UMR 7221 Molecular Physiology and Adaptation, Centre National de le Recherche Scientifique-Muséum National d'Histoire Naturelle, Paris, France
| | - Sylvie Remaud
- UMR 7221 Molecular Physiology and Adaptation, Centre National de le Recherche Scientifique-Muséum National d'Histoire Naturelle, Paris, France
| |
Collapse
|
99
|
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
|
100
|
Imamura O, Arai M, Dateki M, Oishi K, Takishima K. Donepezil-induced oligodendrocyte differentiation is mediated through estrogen receptors. J Neurochem 2019; 155:494-507. [PMID: 31778582 DOI: 10.1111/jnc.14927] [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: 04/11/2019] [Revised: 11/21/2019] [Accepted: 11/21/2019] [Indexed: 12/24/2022]
Abstract
Loss of oligodendrocytes, the myelin-forming cells of the central nervous system, and subsequent failure of myelin development result in serious neurological disorders such as multiple sclerosis. Using primary mouse embryonic neural stem cells (NSCs), we previously demonstrated that donepezil, an acetylcholinesterase inhibitor developed for the treatment of Alzheimer's disease, stimulates the differentiation of NSCs into oligodendrocytes and neurons, albeit at the expense of astrogenesis. However, the precise mechanisms underlying donepezil-induced differentiation remain unclear. In this study, we aimed at elucidating the molecular pathways contributing to donepezil-induced differentiation of mouse-induced pluripotent stem cell-derived neural stem cells (miPSC-NSCs). We used cell-based reporter gene arrays to investigate effects of donepezil on differentiation of miPSC-NSCs. Subsequently, we assessed the molecular pathway underlying donepezil action on differentiation of miPSC-NSCs into mature oligodendrocytes. Donepezil increased the transcriptional activity of estrogen response element under differentiating conditions. Moreover, estrogen receptors α (ERα) and β (ERβ) were highly expressed in MBP-positive mature oligodendrocytes. The ER antagonist ICI 182,780 abrogated the number of MBP-positive oligodendrocytes induced by donepezil, but showed no effect on the differentiation of miPSC-NSCs into Tuj1-positive neurons and GFAP-positive astrocytes. Furthermore, the donepezil-induced generation of mature oligodendrocytes from miPSC-NSC was significantly attenuated by antagonists and siRNA targeting ERα and ERβ. In conclusion, we demonstrated, for the first time, that donepezil-induced oligodendrogenesis is mediated through both ER subtypes, ERα and ERβ. Cover Image for this issue: https://doi.org/10.1111/jnc.14771.
Collapse
Affiliation(s)
- Osamu Imamura
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Masaaki Arai
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Minori Dateki
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Saitama, Japan
| | - Kazuhiko Oishi
- Department of Pharmacology, Meiji Pharmaceutical University, Kiyose, Tokyo, Japan
| | - Kunio Takishima
- Department of Biochemistry, National Defense Medical College, Tokorozawa, Saitama, Japan
| |
Collapse
|