1
|
Serneels PJ, De Schutter JD, De Groef L, Moons L, Bergmans S. Oligodendroglial heterogeneity in health, disease, and recovery: deeper insights into myelin dynamics. Neural Regen Res 2025; 20:3179-3192. [PMID: 39665821 PMCID: PMC11881716 DOI: 10.4103/nrr.nrr-d-24-00694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 09/19/2024] [Accepted: 10/17/2024] [Indexed: 12/13/2024] Open
Abstract
Decades of research asserted that the oligodendroglial lineage comprises two cell types: oligodendrocyte precursor cells and oligodendrocytes. However, recent studies employing single-cell RNA sequencing techniques have uncovered novel cell states, prompting a revision of the existing terminology. Going forward, the oligodendroglial lineage should be delineated into five distinct cell states: oligodendrocyte precursor cells, committed oligodendrocyte precursor cells, newly formed oligodendrocytes, myelin-forming oligodendrocytes, and mature oligodendrocytes. This new classification system enables a deeper understanding of the oligodendroglia in both physiological and pathological contexts. Adopting this uniform terminology will facilitate comparison and integration of data across studies. This, including the consolidation of findings from various demyelinating models, is essential to better understand the pathogenesis of demyelinating diseases. Additionally, comparing injury models across species with varying regenerative capacities can provide insights that may lead to new therapeutic strategies to overcome remyelination failure. Thus, by standardizing terminology and synthesizing data from diverse studies across different animal models, we can enhance our understanding of myelin pathology in central nervous system disorders such as multiple sclerosis, Alzheimer's disease, and amyotrophic lateral sclerosis, all of which involve oligodendroglial and myelin dysfunction.
Collapse
Affiliation(s)
- Pieter-Jan Serneels
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Julie D. De Schutter
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Lies De Groef
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Cellular Communication & Neurodegeneration Research Group, Leuven, Belgium
| | - Lieve Moons
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| | - Steven Bergmans
- KU Leuven, Leuven Brain Institute, Department of Biology, Animal Physiology and Neurobiology Division, Neural Circuit Development & Regeneration Research Group, Leuven, Belgium
| |
Collapse
|
2
|
Liu L, Zhao Y, Bu J, Peng S, Li Y, Su P, Li Y. Baicalin and kaempferol alleviates cuprizone-induced demyelination and microglial activation by inhibiting the STAT3 and NF-κB signaling pathways. Int Immunopharmacol 2025; 154:114592. [PMID: 40174341 DOI: 10.1016/j.intimp.2025.114592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/04/2025]
Abstract
BACKGROUND Microglia-mediated neuroinflammation plays a critical role in myelin loss in multiple sclerosis (MS). Baicalin (BAI) and kaempferol (KAE) are known for their potent anti-inflammatory properties; however, their potential to alleviate demyelination and treat MS has rarely been rarely investigated. METHODS Demyelination was induced in mice using cuprizone (CPZ), and behavioral assessments were conducted to evaluate motor function recovery following treatment with BAI and KAE. Myelin loss was assessed using transmission electron microscopy. Immunofluorescence and Western blot analyses were employed to assess microglial activation and neuroinflammatory responses. Moreover, network pharmacology, molecular docking, and Drug Affinity Responsive Target Stability (DARTS) were utilized to identify molecular targets involved in the therapeutic effects of BAI and KAE. RESULTS Both BAI and KAE significantly improved motor function and mitigated myelin loss in CPZ-treated mice. These compounds effectively inhibited CPZ-induced microglial activation and attenuated secretion of pro-inflammatory cytokines. Network pharmacology identified the STAT3 and NF-κB as potential targets of BAI and KAE. In vivo and in vitro validation further confirmed that both compounds significantly suppressed the phosphorylation of STAT3 and NF-κB p65 proteins. CONCLUSION Our findings suggest that BAI and KAE alleviate CPZ-induced demyelination by inhibiting STAT3 and NF-κB signaling pathways, thereby reduced microglial activation and alleviation of neuroinflammation.
Collapse
Affiliation(s)
- Liming Liu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain for Yu-Yao of Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, PR China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, PR China
| | - Yufang Zhao
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain for Yu-Yao of Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, PR China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, PR China
| | - Jingjing Bu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain for Yu-Yao of Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, PR China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, PR China
| | - Shuaijun Peng
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain for Yu-Yao of Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, PR China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, PR China
| | - Yuyao Li
- College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, PR China
| | - Pan Su
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain for Yu-Yao of Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, PR China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| | - Yucheng Li
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain for Yu-Yao of Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, PR China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, PR China.
| |
Collapse
|
3
|
Tahmasebi F, Asl ER, Faghihi F, Bolandi N, Barati S. Synergistic Effects of Olfactory Ecto-Mesenchymal Stem Cell Supernatant and Ellagic Acid on Demyelination and Glial Modulation in a Chronic Multiple Sclerosis Model. Cell Mol Neurobiol 2025; 45:40. [PMID: 40314717 DOI: 10.1007/s10571-025-01558-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 04/22/2025] [Indexed: 05/03/2025]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system. Stem cells as a source of neurotrophic factors and ellagic acid (EA) as an antioxidant reduce the progression of neurodegenerative diseases. In this study, we evaluated the effect of olfactory ecto-mesenchymal stem cell (EMSC) supernatant and EA on A1 astrocytes, M1 microglia, and demyelination in cuprizone model. To induce the chronic demyelination model, mice received a diet containing 0.2% cuprizone/kg of food for 12 weeks. EMSC supernatant was injected into the lateral ventricle of mice. EA was administered intraperitoneally daily at a dose of 80 mg/kg body weight for two weeks. Two weeks after injection, immunohistochemistry was performed to detect the presence of astrocytes (GFAP), microglia/macrophages (Iba-1), and oligodendrocytes (Olig2). The level of gene expression of EMSC (TGF-β and BDNF), astrocytes (C3 and GBP2) and microglia (iNOS, TNF-α and IL-6) was evaluated by qRT-PCR method. The results showed that injection of EMSC and EA increased the expression of TGF-β and BDNF genes as trophic factors. LFB images showed that supernatant and EA significantly improved remyelination, which was accompanied by an increase in oligodendrocyte population. The astrocyte population increased in the cuprizone group, while it decreased after supernatant and EA administration. The supernatant and EA decreased microglia after cuprizone induction. The qRT-PCR showed neurotoxic genes of A1 and M1 decreased after supernatant and EA administration. Here, we demonstrate that EMSC supernatant and EA could improve demyelination in neurodegenerative diseases such as MS by reducing microgliosis and astrocytosis in addition to increasing myelination.
Collapse
Affiliation(s)
- Fatemeh Tahmasebi
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran
| | - Elmira Roshani Asl
- Department of Biochemistry, Saveh University of Medical Sciences, Saveh, Iran
| | - Faezeh Faghihi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Immunogenetics Research Center, Mazandaran University of Medical Sciences, Sari, Iran
| | - Nadia Bolandi
- Student Research Committee, Department of Clinical Biochemistry, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shirin Barati
- Department of Anatomy, Saveh University of Medical Sciences, Saveh, Iran.
| |
Collapse
|
4
|
van Veggel L, Schepers M, Tiane A, Kumar V, Willems E, Rombaut B, Noordijk J, Vangansewinkel T, Li A, Wolfs E, Ozcan B, Nouboers E, Moya PR, Sauer DB, Diliën H, Hellings N, Schreiber R, Vanmierlo T. EAAT3 modulation: A potential novel avenue towards remyelination in multiple sclerosis. Biomed Pharmacother 2025; 186:117960. [PMID: 40138922 DOI: 10.1016/j.biopha.2025.117960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/24/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Modulating the excitatory amino acid transporter 3 (EAAT3) can be considered a novel approach for the treatment of multiple sclerosis (MS). EAAT3 plays a crucial role in regulating oxidative stress and oligodendrocyte function through its ability to transport cysteine, the rate-limiting building block in the synthesis of the antioxidant glutathione. Therefore, EAAT3 activation is hypothesised to improve oligodendrocyte health and relieve its differentiation block in MS, improving remyelination capacity. Using a cuprizone-induced demyelination model, the effects of EAAT3 overexpression by viral transduction of oligodendrocytes and pharmacological inhibition of EAAT3 were examined. Surprisingly, EAAT3 overexpression significantly hampered remyelination, while EAAT3 inhibition prevented demyelination and improved functional remyelination as assessed by visual evoked potentials and post mortem myelin basic protein fluorescent staining. Next, cellular mechanisms underlying these results were investigated. Consistent with the in vivo findings, post mortem gene expression analysis of the corpus callosum of cuprizone treated animals revealed a trend towards upregulation of oligodendrocyte lineage genes in response to EAAT3 inhibition, supporting its role in oligodendrocyte health and myelination processes. In vitro studies using the human oligodendroglioma (HOG) cell line demonstrated the beneficial effects of EAAT3 inhibition on cellular morphology, indicating potential roles in promoting oligodendrocyte maturation and myelination. In contrast, EAAT3 overexpression appears to hamper these processes. These findings suggest that, contrary to our initial hypothesis, EAAT3 inhibition could improve oligodendrocyte function and myelination processes, highlighting its potential as a therapeutic target for demyelinating disorders. Future studies should address the exact molecular mechanism through which this effect is obtained.
Collapse
Affiliation(s)
- Lieve van Veggel
- Department of Neuroscience, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands; University MS Center (UMSC), Hasselt-Pelt, Belgium
| | - Melissa Schepers
- Department of Neuroscience, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands; University MS Center (UMSC), Hasselt-Pelt, Belgium
| | - Assia Tiane
- Department of Neuroscience, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands; University MS Center (UMSC), Hasselt-Pelt, Belgium
| | - Vijay Kumar
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Emily Willems
- Department of Neuroscience, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands; University MS Center (UMSC), Hasselt-Pelt, Belgium
| | - Ben Rombaut
- Department of Neuroscience, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands; University MS Center (UMSC), Hasselt-Pelt, Belgium
| | - Jurrie Noordijk
- Sensor Engineering Department, Faculty of Science and Engineering, Maastricht University, Maastricht, the Netherlands
| | - Tim Vangansewinkel
- Department of Neuroscience, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Anna Li
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Esther Wolfs
- Department of Neuroscience, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Berra Ozcan
- Department of Neuroscience, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Evelien Nouboers
- Department of Neuroscience, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium
| | - Pablo R Moya
- Facultad de Ciencias, Instituto de Fisiología, Centro Interdisciplinario de Neurociencia de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
| | - David B Sauer
- Centre for Medicines Discovery, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Hanne Diliën
- Sensor Engineering Department, Faculty of Science and Engineering, Maastricht University, Maastricht, the Netherlands
| | - Niels Hellings
- Department of Neuroscience, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands
| | - Rudy Schreiber
- Section of Psychopharmacology, Neuropsychology and Psychopharmacology, Faculty of Psychology and Neuroscience, Maastricht University, Maastricht, the Netherlands
| | - Tim Vanmierlo
- Department of Neuroscience, BIOMED Biomedical Research Institute, Faculty of Medicine and Life Sciences, Hasselt University, Hasselt, Belgium; Department of Psychiatry and Neuropsychology, Division of Translational Neuroscience, European Graduate School of Neuroscience, Mental Health and Neuroscience Research Institute, Maastricht University, Maastricht, the Netherlands; University MS Center (UMSC), Hasselt-Pelt, Belgium.
| |
Collapse
|
5
|
Rahamathulla M, Kumar R, AlRashdan Y, Bose MM, Muthukumar A, Salahuddin M, Ahmed MM, Shivanandappa TB, Pasha I. Neuroprotective Effects of Cucurbita pepo Extract in Cuprizone-Induced Multiple Sclerosis Animal Model. Mol Neurobiol 2025:10.1007/s12035-025-04934-1. [PMID: 40254703 DOI: 10.1007/s12035-025-04934-1] [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: 10/22/2024] [Accepted: 04/08/2025] [Indexed: 04/22/2025]
Abstract
Cuprizone is a neurotoxin with copper-chelating ability, and it produces symptoms that are similar to those caused by multiple sclerosis lesions. Multiple sclerosis induced by cuprizone in animal models depicts oxidative stress as one of the prime factors in pathogenesis. Neurodegeneration has become an extremely serious issue on a global scale. It is preferable to use natural therapy to stop the disease's progression and future occurrences. Cucurbita pepo is a plant with neuroprotective properties and antioxidant properties. The purpose of this study was to determine the ameliorative capability of C. pepo in cuprizone-induced behavioral parameters and histopathological alterations in Wistar rats. The study included four groups of rats: a control group (normal saline), a cuprizone-induced group, and two treatment groups receiving C. pepo at low (200 mg/kg) and high (400 mg/kg) doses for 5 weeks. We exposed the rats to a rotarod test, a forced swimming test, and an elevated plus maze test to assess their behavioral parameters, including motor coordination and anxiety level. Following the study, animals were sacrificed, and brain tissues were extracted to resolve biochemical analysis for oxidative stress along with histopathological changes to determine and compare the oxidative stress between the inducer and treated group and also to analyze the remyelination ability of C. pepo in the disease-induced rat. Results revealed that the rats treated with C. pepo demonstrated significant improvements in motor coordination (P < 0.001) and reduced anxiety-like behavior (P < 0.01) and oxidative stress markers, such as superoxide dismutase and catalase activity. Histopathological analysis showed reduced demyelination and enhanced neuronal integrity in treated groups. These findings indicate that C. pepo attenuates cuprizone-induced oxidative stress and demyelination, supporting its neuroprotective potential in multiple sclerosis models.
Collapse
Affiliation(s)
- Mohamed Rahamathulla
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Al Faraa, 62223, Abha, Saudi Arabia
| | - Rupesh Kumar
- Department of Pharmacology, Alameen College of Pharmacy, Bangalore, India.
| | - Yazan AlRashdan
- Department of Biopharmaceutics and Clinical Pharmacy, School of Pharmacy, University of Jordan, Amman, 11180, Jordan
| | - Madhura M Bose
- Department of Pharmacology, Alameen College of Pharmacy, Bangalore, India
| | | | - Md Salahuddin
- Department of Pharmaceutical Chemistry, Al-Ameen College of Pharmacy, Bengaluru, Karnataka, 560027, India
| | - Mohammed Muqtader Ahmed
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdul Aziz University, Al Kharj, 11942, Saudi Arabia
| | | | - Ismail Pasha
- Department of Pharmacology, Orotta College of Medicine and Health Science, Asmara, Eritrea.
| |
Collapse
|
6
|
Chadarevian JP, Davtyan H, Chadarevian AL, Nguyen J, Capocchi JK, Le L, Escobar A, Chadarevian T, Mansour K, Deynega E, Mgerian M, Tu C, Kiani Shabestari S, Carlen-Jones W, Eskandari-Sedighi G, Hasselmann J, Spitale RC, Blurton-Jones M. Harnessing human iPSC-microglia for CNS-wide delivery of disease-modifying proteins. Cell Stem Cell 2025:S1934-5909(25)00099-2. [PMID: 40233761 DOI: 10.1016/j.stem.2025.03.009] [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: 07/10/2024] [Revised: 12/03/2024] [Accepted: 03/17/2025] [Indexed: 04/17/2025]
Abstract
Widespread delivery of therapeutic proteins to the brain remains challenging. To determine whether human induced pluripotent stem cell (iPSC)-microglia (iMG) could enable brain-wide and pathology-responsive delivery of therapeutic cargo, we utilized CRISPR gene editing to engineer iMG to express the Aβ-degrading enzyme neprilysin under control of the plaque-responsive promoter, CD9. To further determine whether increased engraftment enhances efficacy, we utilized a CSF1R-inhibitor resistance approach. Interestingly, both localized and brain-wide engraftment in Alzheimer's disease (AD) mice reduced multiple biochemical measures of pathology. However, within the plaque-dense subiculum, reductions in plaque load, dystrophic neurites, and astrogliosis and preservation of neuronal density were only achieved following widespread microglial engraftment. Lastly, we examined chimeric models of breast cancer brain metastases and demyelination, demonstrating that iMG adopt diverse transcriptional responses to differing neuropathologies, which could be harnessed to enable widespread and pathology-responsive delivery of therapeutics to the CNS.
Collapse
Affiliation(s)
- Jean Paul Chadarevian
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Hayk Davtyan
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Alina L Chadarevian
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Jasmine Nguyen
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA
| | - Joia K Capocchi
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA
| | - Lauren Le
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA
| | - Adrian Escobar
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Talar Chadarevian
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Kimiya Mansour
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Ekaterina Deynega
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA
| | - Michael Mgerian
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Christina Tu
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Sepideh Kiani Shabestari
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - William Carlen-Jones
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA
| | - Ghazaleh Eskandari-Sedighi
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA
| | - Jonathan Hasselmann
- Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA
| | - Robert C Spitale
- Department of Pharmaceutical Sciences, University of California, Irvine, Irvine, CA 92697, USA.
| | - Mathew Blurton-Jones
- Department of Neurobiology & Behavior, University of California, Irvine, Irvine, CA 92697, USA; Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, Irvine, CA 92697, USA; Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, CA 92697, USA.
| |
Collapse
|
7
|
SoRelle ED, Luftig MA. Multiple sclerosis and infection: history, EBV, and the search for mechanism. Microbiol Mol Biol Rev 2025; 89:e0011923. [PMID: 39817754 PMCID: PMC11948499 DOI: 10.1128/mmbr.00119-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025] Open
Abstract
SUMMARYInfection has long been hypothesized as the cause of multiple sclerosis (MS), and recent evidence for Epstein-Barr virus (EBV) as the trigger of MS is clear and compelling. This clarity contrasts with yet uncertain viral mechanisms and their relation to MS neuroinflammation and demyelination. As long as this disparity persists, it will invigorate virologists, molecular biologists, immunologists, and clinicians to ascertain how EBV potentiates MS onset, and possibly the disease's chronic activity and progression. Such efforts should take advantage of the diverse body of basic and clinical research conducted over nearly two centuries since the first clinical descriptions of MS plaques. Defining the contribution of EBV to the complex and multifactorial pathology of MS will also require suitable experimental models and techniques. Such efforts will broaden our understanding of virus-driven neuroinflammation and specifically inform the development of EBV-targeted therapies for MS management and, ultimately, prevention.
Collapse
Affiliation(s)
- Elliott D. SoRelle
- Department of Molecular Genetics & Microbiology, Center for Virology, Duke University, Durham, North Carolina, USA
| | - Micah A. Luftig
- Department of Molecular Genetics & Microbiology, Center for Virology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
8
|
Alidadi M, Omidi N, Abdi M, Mohammadi M, Shabani M, Kashani IR. Melatonin ameliorates astrogliosis and microgliosis in a cuprizone demyelinating mouse model. Biochem Biophys Rep 2025; 41:101929. [PMID: 39926210 PMCID: PMC11803163 DOI: 10.1016/j.bbrep.2025.101929] [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: 09/14/2024] [Revised: 01/16/2025] [Accepted: 01/21/2025] [Indexed: 02/11/2025] Open
Abstract
Background and purpose Several investigations have reported that melatonin is involved in the amelioration of the inflammatory process, improvement of myelin function, and regeneration in the central nervous system (CNS). The current study aimed to evaluate the protective effect of melatonin in cuprizone (CPZ)-induced myelin damage in the corpus callosum (CC) and explore the plausible underlying mechanisms of remyelination capacity and/or neuroprotection. Method We administered cuprizone in chow either alone daily for 6 weeks or combined with simultaneously applied melatonin intra-peritoneal injections. we studied demyelination by LFB staining, oligodendrocyte staining using anti-Olig2 or anti-APC antibodies. In addition, we visualized microgliosis and astrocytosis by staining with anti-Iba-1 and anti-GFAP antibodies. Furthermore, we study the effect of melatonin on mRNA expression of Musashi-1, Hes-1 and Notch-1 genes. Results Our data showed that cuprizone intoxication caused a significant oligodendrocyte loss, demyelination, and reactive gliosis in CC. Administration of melatonin prevented the demyelination in CC as determined by Luxol fast blue staining. Furthermore, we found that the melatonin significantly suppressed the cuprizone-induced microgliosis and astrocytosis. while the frequency of oligodendrocytes (Olig2+) was significantly enhanced in the CC after melatonin administration. In addition, melatonin significantly modulated Musashi1, Hes1, and Notch1 mRNA expression in the CC of mice. Conclusion These results provide evidence that melatonin abolishes destructive cuprizone effects in the mouse corpus callosum by restoring oligodendrocyte generation, remyelination, and decreasing astrogliosis and microgliosis.
Collapse
Affiliation(s)
- Mehdi Alidadi
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Negar Omidi
- Cardiac Primary Prevention Research Center, Tehran Heart Center AND Department of Cardiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahdad Abdi
- Department of Anatomy, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Maryam Mohammadi
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
9
|
El-Sayed SAM, Fouad GI, Rizk MZ, Beherei HH, Mabrouk M. Comparative Neuroprotective Potential of Nanoformulated and Free Resveratrol Against Cuprizone-Induced Demyelination in Rats. Mol Neurobiol 2025; 62:2710-2725. [PMID: 39152208 PMCID: PMC11790707 DOI: 10.1007/s12035-024-04415-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 07/30/2024] [Indexed: 08/19/2024]
Abstract
Demyelination is a frequent yet crippling neurological disease associated with multiple sclerosis (MS). The cuprizone (CZ) model, which causes demyelination through oxidative stress and neuroinflammation, is a popular tool used by researchers to examine this process. The polyphenol resveratrol (RESV) has become a promising neuroprotective agent in seeking for efficient therapies. In a rat model given CZ, we created and examined iron oxide nanoparticles (IONPs) loaded with RESV (IONP-RESV) to see how effective they were as a therapeutic agent against free RESV. According to molecular mechanisms, exposure to CZ resulted in a marked downregulation of myelin proteolipid protein (PLP) expression and an overexpression of the inflammatory markers tumor necrosis factor-α (TNF-α) and S100β, which are indicators of demyelination and neuroinflammation. It is remarkable that these CZ-induced alterations could be reversed by therapy with either RESV or IONP-RESV. Interestingly, IONP-RESV showed even stronger anti-inflammatory activity, as shown by a more noticeable downregulation of TNF-α and S100β expression. These results were confirmed by histopathological examination of the cerebral cortices. Our findings support the better neuroprotective benefits of RESV-loaded IONPs over free RESV in reducing demyelination and neuroinflammation brought on by CZ. Owing to their pro-remyelinating, anti-inflammatory, and antioxidant properties, RESV-loaded IONPs show promise as a neurotherapeutic intervention in the future for neurological diseases such as multiple sclerosis.
Collapse
Affiliation(s)
- Sara A M El-Sayed
- Refractories, Ceramics and Building Materials Department, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt.
| | - Ghadha Ibrahim Fouad
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Maha Z Rizk
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Hanan H Beherei
- Refractories, Ceramics and Building Materials Department, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| |
Collapse
|
10
|
Lohrberg M, Mortensen LS, Thomas C, Fries F, van der Meer F, Götz A, Landt C, Rhee HJ, Rhee J, Gómez-Varela D, Schmidt M, Möbius W, Ruhwedel T, Pardo LA, Remling L, Kramann N, Wrzos C, Bahn E, Stadelmann C, Barrantes-Freer A. Astroglial modulation of synaptic function in the non-demyelinated cerebellar cortex is dependent on MyD88 signaling in a model of toxic demyelination. J Neuroinflammation 2025; 22:47. [PMID: 39988657 PMCID: PMC11849172 DOI: 10.1186/s12974-025-03368-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/03/2025] [Indexed: 02/25/2025] Open
Abstract
Progressive neurological decline in multiple sclerosis is associated with axonal loss and synaptic dysfunction in the non-demyelinated normal appearing gray matter (NAGM) and prominently in the cerebellum. In contrast to early disease stages, where synaptic and neuro-axonal pathology correlates with the extent of T cell infiltration, a prominent role of the innate immune system has been proposed for progressive MS. However, the specific contribution of microglia and astrocytes to synaptic cerebellar pathology in the NAGM- independent of an adaptive T cell response - remains largely unexplored. In the present study, we quantified synaptic changes in the cerebellar NAGM distant from demyelinated lesions in a mouse model of toxic demyelination. Proteomic analysis of the cerebellar cortex revealed differential regulation of synaptic and glutamate transport proteins in the absence of evident structural synaptic pathology or local gray matter demyelination. At the functional level, synaptic changes manifested as a reduction in frequency-dependent facilitation at the parallel fiber- Purkinje cell synapse. Further, deficiency of MyD88, an adaptor protein of the innate immune response, associated with a functional recovery in facilitation, reduced changes in the differential expression of synaptic and glutamate transport proteins, and reduced transcription levels of inflammatory cytokines. Nevertheless, the characteristics of demyelinating lesions and their associated cellular response were similar to wild type animals. Our work brings forward an experimental paradigm mimicking the diffuse synaptic pathology independent of demyelination in late stage MS and highlights the complex regulation of synaptic pathology in the cerebellar NAGM. Moreover, our findings suggest a role of astrocytes, in particular Bergmann glia, as key cellular determinants of cerebellar synaptic dysfunction.
Collapse
Affiliation(s)
- Melanie Lohrberg
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Campus Institute Data Science, Göttingen, Germany
| | - Lena Sünke Mortensen
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, Leipzig, Germany
- Paul-Flechsig-Institute of Neuropathology, University Medical Center Leipzig, Leipzig, Germany
| | - Carolina Thomas
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
- Paul-Flechsig-Institute of Neuropathology, University Medical Center Leipzig, Leipzig, Germany
| | - Franziska Fries
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | | | - Alexander Götz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Carolin Landt
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Hong Jun Rhee
- Department of Molecular Neurobiology, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - JeongSeop Rhee
- Department of Molecular Neurobiology, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - David Gómez-Varela
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Manuela Schmidt
- Division of Pharmacology and Toxicology, Department of Pharmaceutical Sciences, University of Vienna, Vienna, Austria
| | - Wiebke Möbius
- Department of Neurogenetics, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Network of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Luis A Pardo
- Oncophysiology Group, Max-Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Linus Remling
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Nadine Kramann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Claudia Wrzos
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Erik Bahn
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines to Network of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany.
| | - Alonso Barrantes-Freer
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany.
- Paul-Flechsig-Institute of Neuropathology, University Medical Center Leipzig, Leipzig, Germany.
| |
Collapse
|
11
|
Rida Zainab S, Zeb Khan J, Khalid Tipu M, Jahan F, Irshad N. A review on multiple sclerosis: Unravelling the complexities of pathogenesis, progression, mechanisms and therapeutic innovations. Neuroscience 2025; 567:133-149. [PMID: 39709058 DOI: 10.1016/j.neuroscience.2024.12.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/25/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Multiple sclerosis (MS) is a chronic, inflammatory demyelinating disorder of the central nervous system (CNS) targeting myelinated axons. Pathogenesis of MS entails an intricate genetic, environmental, and immunological interaction. Dysregulation of immune response i.e. autoreactive T & B-Cells and macrophage infiltration into the CNS leads to inflammation, demyelination, and neurodegeneration. Disease progression of MS varies among individuals transitioning from one form of relapsing-remitting to secondary progressive MS (SPMS). Research advances have unfolded various molecular targets involved in MS from oxidative stress to blood-brain barrier (BBB) disruption. Different pathways are being targeted so far such as inflammatory and cytokine signaling pathways to overcome disease progression. Therapeutic innovations have significantly transformed the management of MS, especially the use of disease-modifying therapies (DMTs) to reduce relapse rates and control disease progression. Advancements in research, neuroprotective strategies, and remyelination strategies hold promising results in reversing CNS damage. Various mice models are being adopted for testing new entities in MS research.
Collapse
Affiliation(s)
- Syeda Rida Zainab
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Jehan Zeb Khan
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Muhammad Khalid Tipu
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| | - Faryal Jahan
- Shifa College of Pharmaceutical Sciences, STMU, Islamabad, Pakistan.
| | - Nadeem Irshad
- Department of Pharmacy, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, Pakistan.
| |
Collapse
|
12
|
Jing F, Wang Q, Xu Y, Dong J, Huang H, Li Y. MAZ regulates ferroptosis, apoptosis and differentiation of oligodendrocyte precursor cells. Brain Res 2025; 1849:149349. [PMID: 39581526 DOI: 10.1016/j.brainres.2024.149349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/08/2024] [Accepted: 11/21/2024] [Indexed: 11/26/2024]
Abstract
Oligodendrocyte precursor cells (OPCs) respond rapidly to demyelination injury. However, the rescuing effects may be hindered by cell death of OPCs, leading to incomplete remyelination. This study aimed to explore the expression of MYC-associated zinc finger protein (MAZ) in demyelinated mice and the effects of MAZ on cell death form and differentiation of OPCs. Mice received demyelinating agent (cuprizone, CZ) for 5 weeks. Histological staining demonstrated that CZ feeding triggered demyelination in the corpus callosum (CC). Detection of iron content and ferroptosis markers indicated that ferroptosis was inducted in the CC of CZ-treated mice. Notably, we found CZ feeding resulted in a reduction of MAZ expression within the CC. Using CCK-8 assay, detection of iron content, MDA level, GSH level, and ferroptosis markers, lipid ROS detection, and immunofluorescence staining for 4-HNE, we found that knockdown of MAZ facilitated OPC ferroptosis. We also evaluated the susceptibility of MAZ-overexpressing OPCs to ferroptosis inducer, erastin, and demonstrated that MAZ-overexpressing OPCs were resistant to erastin-induced ferroptosis. TUNEL staining and western blot analysis indicated that MAZ knockdown promoted apoptosis of OPCs by inhibiting PI3K/Akt activation. Immunofluorescence staining of MBP indicated that knockdown of MAZ inhibited OPC differentiation. Moreover, we elucidate the mechanism responsible for MAZ's protective effects on OPC death and differentiation, which may be achieved through transcriptional activation of SOX2. Our findings introduced MAZ as a beneficial modulator of OPC survival and differentiation, and it could serve as a potential therapeutic target for demyelination diseases.
Collapse
Affiliation(s)
- Fangkun Jing
- Department of Neurosurgery, The People's Hospital of China Medical University, 110067 Liaoning, China
| | - Quancai Wang
- Department of Neurosurgery, The People's Hospital of China Medical University, 110067 Liaoning, China
| | - Yangxi Xu
- Department of Neurosurgery, The People's Hospital of China Medical University, 110067 Liaoning, China
| | - Jingyu Dong
- Department of Neurosurgery, The People's Hospital of China Medical University, 110067 Liaoning, China
| | - Haitao Huang
- Department of Neurosurgery, The People's Hospital of China Medical University, 110067 Liaoning, China.
| | - Yanfeng Li
- Department of Neurosurgery, The People's Hospital of China Medical University, 110067 Liaoning, China.
| |
Collapse
|
13
|
Ibrahim Fouad G, Mabrouk M, El-Sayed SAM, Abdelhameed MF, Rizk MZ, Beherei HH. Berberine-loaded iron oxide nanoparticles alleviate cuprizone-induced astrocytic reactivity in a rat model of multiple sclerosis. Biometals 2025; 38:203-229. [PMID: 39543075 PMCID: PMC11754386 DOI: 10.1007/s10534-024-00648-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 10/20/2024] [Indexed: 11/17/2024]
Abstract
Berberine (BBN) is a naturally occurring alkaloid as a secondary metabolite in many plants and exhibits several benefits including neuroprotective activities. However, data on the neuromodulating potential of nanoformulated BBN are still lacking. In the present study, BBN loaded within iron oxide nanoparticles (BBN-IONP) were prepared and characterized by transmission electron microscopy FTIR, X-ray photoelectron spectroscopy particle-size distribution, zeta potential, and HPLC. The remyelinating neuroprotective potential of BBN-IONP relative to free BBN was evaluated against cuprizone (CPZ)-induced neurotoxicity (rats administered 0.2% CPZ powder (w/w) for five weeks). CPZ rats were treated with either free BBN or IONP-BBN (50 mg/kg/day, orally) for 14 days. Cognitive function was estimated using Y-maze. Biochemically, total antioxidant capacity lipid peroxides and reduced glutathione in the brain tissue, as well as, serum interferon-gamma levels were estimated. Moreover, the genetic expression contents of myelin basic protein Matrix metallopeptidase-9 Tumor necrosis factor-α (TNF-α), and S100β were measured. The histopathological patterns and immunohistochemical assessment of Glial Fibrillary Acidic Protein in both cerebral cortex and hippocampus CA1 regions were investigated. CPZ-rats treated with either free BBN or IONP-BBN demonstrated memory restoring, anti-oxidative, anti-inflammatory, anti-astrocytic, and remyelinating activities. Comparing free BBN with IONP-BBN revealed that the latter altered the neuromodulating activities of BBN, showing superior neuroprotective activities of IONP-BBN relative to BBN. In conclusion, both forms of BBN possess neuroprotective potential. However, the use of IONPs for brain delivery and the safety of these nano-based forms need further investigation.
Collapse
Affiliation(s)
- Ghadha Ibrahim Fouad
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, 33 El-Bohouth St., Dokki, Cairo, 12622, Egypt.
| | - Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Sara A M El-Sayed
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Mohamed F Abdelhameed
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Bohouth St., Dokki, Cairo, 12622, Egypt
| | - Maha Z Rizk
- Department of Therapeutic Chemistry, Pharmaceutical and Drug Industries Research Institute, National Research Centre, 33 El-Bohouth St., Dokki, Cairo, 12622, Egypt
| | - Hanan H Beherei
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| |
Collapse
|
14
|
Liu SS, Zha Z, Li C, Li CY, Wang L. The mechanism of exosomes of BMSCs modified with Bu Shen Yi Sui capsule in promoting remyelination via regulating miR-15b/Wnt signaling pathway-mediated differentiation of oligodendrocytes. JOURNAL OF ETHNOPHARMACOLOGY 2025; 340:119283. [PMID: 39733800 DOI: 10.1016/j.jep.2024.119283] [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: 10/16/2024] [Revised: 12/11/2024] [Accepted: 12/21/2024] [Indexed: 12/31/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Bu Shen Yi Sui capsule (BSYS), a modified version of the classical Chinese medicine formula Liu Wei Di Huang pill, has demonstrated therapeutic efficacy in the treatment of multiple sclerosis (MS). Nevertheless, the precise mechanism through which BSYS facilitates remyelination remains to be elucidated. AIM OF THE STUDY This research investigates the role and potential mechanisms of BSYS-modified exosomes (exos) derived from bone marrow mesenchymal stem cells (BMSCs) in promoting remyelination in a cuprizone (CPZ)-induced demyelination model in mice. MATERIALS AND METHODS C57BL/6J mice were administered a 0.2% CPZ-containing diet for 5 weeks to induce demyelination, followed by treatment with exosomes derived from BMSC (BMSC-exos) and BSYS-modified BMSC exosomes (BSYS-BMSC-exos) twice weekly for 2 weeks. Body weight measurements were recorded, and motor function was evaluated using the rotarod test. Pathological changes in myelin and axons were assessed via Luxol fast blue (LFB) staining, transmission electron microscopy (TEM), and immunofluorescence (IF) staining. Oligodendrocyte proliferation, differentiation, and maturation were analyzed using IF double-staining, Western blot (WB), and real-time quantitative reverse transcription PCR (qRT-PCR). Additionally, microRNA (miRNA) sequencing and a luciferase reporter assay were conducted to verify miRNA binding to its target gene. Key markers of the Wnt/β-catenin signaling pathway were examined using WB and qRT-PCR. RESULTS BSYS-BMSC-exos treatment significantly increased both body weight and rotarod performance in CPZ mice. Moreover, BMSC-exos and BSYS-BMSC-exos reversed myelin loss and axonal damage. These treatments enhanced oligodendrocytes proliferation, differentiation, and maturation, with BSYS-BMSC-exos exhibiting a particularly pronounced effect on the expression of adenomatous polyposis coli clone CC1 (CC1), 2',3'-cyclic nucleotide 3'-phosphodiesterase (CNPase), proteolipid protein (PLP), myelin oligodendrocyte glycoprotein (MOG), and myelin basic protein (MBP). Sequencing and luciferase assays revealed that miR-15b-5p, enriched in BSYS-BMSC-exos, directly targets Wnt3a. Furthermore, BSYS-BMSC-exos elevated axis inhibition protein 2 (Axin2) expression while markedly reducing Wnt family member 3A (Wnt3a), phospho-glycogen synthase kinase-3β (p-GSK3β), β-catenin, and T-cell specific transcription factor 4/transcription factor 7-like 2 (TCF4/TCF7L2) levels. CONCLUSIONS The findings suggest that BSYS-BMSC-exos alleviate neurological deficits, enhance oligodendrocytes differentiation and maturation, and promote remyelination in CPZ mice. miR-15b-5p, enriched in BSYS-BMSC-exos, targets and downregulates Wnt3a, thereby inhibiting the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Si-Si Liu
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Zheng Zha
- Department of Neurology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Chen Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Chun-Yu Li
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Lei Wang
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| |
Collapse
|
15
|
Polyák H, Galla Z, Rajda C, Monostori P, Klivényi P, Vécsei L. Plasma and Visceral Organ Kynurenine Metabolites Correlate in the Multiple Sclerosis Cuprizone Animal Model. Int J Mol Sci 2025; 26:976. [PMID: 39940744 PMCID: PMC11817772 DOI: 10.3390/ijms26030976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/19/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
The cuprizone (CPZ) model of multiple sclerosis (MS) is excellent for studying the molecular differences behind the damage caused by poisoning. Metabolic differences in the kynurenine pathway (KP) of tryptophan (TRP) degradation are observed in both MS and a CPZ mouse model. Our goal was to analyze the kynurenine, serotonin, and indole pathways of TRP degradation on the periphery, in the neurodegenerative processes of inflammation. In our study, mice were fed with 0.2% CPZ toxin for 5 weeks. We examined the metabolites in the three pathways of TRP breakdown in urine, plasma, and relevant visceral organs with bioanalytical measurements. In our analyses, we found a significant increase in plasma TRP, 5-hydroxytryptophan (5-HTP), and indole-3-acetic acid (IAA) levels, while a decrease in the concentrations of 3-hydroxy-L-kynurenine (3-HK), xanthurenic acid (XA), kynurenic acid (KYNA), and quinaldic acid in the plasma of toxin-treated group was found. A marked decrease in the levels of 3-HK, XA, KYNA, quinaldic acid, and indole-3-lactic acid was also observed in the visceral organs by the end of the poisoning. Furthermore, we noticed a decrease in the urinary levels of the TRP, KYNA, and XA metabolites, while an increase in serotonin and 5-hydroxyindoleacetic acid in the CPZ group was noticed. The toxin treatment resulted in elevated tryptamine and indoxyl sulfate levels and reduced IAA concentration. Moreover, the urinary para-cresyl sulfate concentration also increased in the treated group. In the present study, we showed the differences in the three main metabolic pathways of TRP degradation in the CPZ model. We confirmed the relationship and correlation between the content of the kynurenine metabolites in the plasma and the tissues of the visceral organs. We emphasized the suppression of the KP and the activity of the serotonin and indole pathways with a particular regard to the involvement of the microbiome by the indole pathway. Consequently, this is the first study to analyze in detail the distribution of the kynurenine, serotonin, and indole pathways of TRP degradation in the periphery.
Collapse
Affiliation(s)
- Helga Polyák
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (H.P.); (C.R.); (P.K.)
- Preventive Health Sciences Research Group, Incubation Competence Centre of the Centre of Excellence for Interdisciplinary Research, Development and Innovation of the University of Szeged, H-6720 Szeged, Hungary
| | - Zsolt Galla
- Department of Pediatrics, Albert Szent-Györgyi Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary; (Z.G.); (P.M.)
| | - Cecilia Rajda
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (H.P.); (C.R.); (P.K.)
| | - Péter Monostori
- Department of Pediatrics, Albert Szent-Györgyi Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary; (Z.G.); (P.M.)
| | - Péter Klivényi
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (H.P.); (C.R.); (P.K.)
- HUN-REN-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| | - László Vécsei
- Department of Neurology, Albert Szent-Györgyi Medical School, University of Szeged, Semmelweis u. 6, H-6725 Szeged, Hungary; (H.P.); (C.R.); (P.K.)
- HUN-REN-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos krt. 113, H-6725 Szeged, Hungary
| |
Collapse
|
16
|
Alderbi RM, Alam MZ, Alghamdi BS, Alsufiani HM, Abd El-Aziz GS, Omar UM, Al-Ghamdi MA. Neurotherapeutic impact of vanillic acid and ibudilast on the cuprizone model of multiple sclerosis. Front Mol Neurosci 2025; 17:1503396. [PMID: 39866908 PMCID: PMC11760597 DOI: 10.3389/fnmol.2024.1503396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/23/2024] [Indexed: 01/28/2025] Open
Abstract
Multiple sclerosis (MS) affects 2.8 million people worldwide. Although the cause is unknown, various risk factors might be involved. MS involves the immune system attacking the central nervous system's myelin sheath, leading to neuron damage. This study used a cuprizone (CPZ)-intoxicated mouse model to simulate MS's demyelination/remyelination process. It evaluated the molecular, histological, and behavioral effects of vanillic acid (VA), a natural phenolic acid, alone and with Ibudilast (IBD), a clinically tested MS medication. Mice were divided into a control group (regular chow) and a CPZ group (0.3% cuprizone chow for 5 consecutive weeks). During remyelination, the CPZ group was split into four groups: no therapy, 10 mg/kg of IBD, 30 mg/kg of VA, and combined, each treated for 4 weeks. Behavioral, biochemical, molecular, and histopathological tests occurred in the 5th week (demyelination), 7th (early remyelination), and 9th (late remyelination). Cognitive assessments were at weeks 5 and 9. VA enhanced motor, coordination, and cognitive impairments in CPZ-intoxicated mice and improved histopathological, molecular, and biochemical features during early remyelination. IBD improved behavioral abnormalities across all tests, but combined therapy showed no significant difference from single therapies. Further investigations are necessary to understand VA's mechanisms and potential as an MS treatment.
Collapse
Affiliation(s)
- Rasha M. Alderbi
- Research Centre, King Faisal Specialist Hospital and Research Centre, Jeddah, Saudi Arabia
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Neuroscience and Geroscience Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Z. Alam
- Neuroscience and Geroscience Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Badrah S. Alghamdi
- Neuroscience and Geroscience Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Physiology, Neuroscience Unit, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hadeil M. Alsufiani
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Gamal S. Abd El-Aziz
- Department of Clinical Anatomy, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ulfat M. Omar
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Princess Dr. Najlaa Bint Saud Al-Saud Center of Excellence Research in Biotechnology, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maryam A. Al-Ghamdi
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Vitamin D Pharmacogenomics Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
- Experimental Biochemistry Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
17
|
Sajrawi C, Odeh M, Tiwari AK, Agranovich B, Abramovich I, Zubedat S, Saar G, Shaulov L, Avital A, Reznik D, Benhar M, Radzishevsky I, Engelender S, Wolosker H. Endogenous histidine peptides are physiological antioxidants that prevent oligodendrocyte cell death and myelin loss in vivo. Glia 2025; 73:122-139. [PMID: 39360557 DOI: 10.1002/glia.24624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 09/19/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
Histidine dipeptides (HDs) are synthesized in brain oligodendrocytes by carnosine synthase (carns1), but their role is unknown. Using metabolomics and in vivo experiments with both constitutive and oligodendrocyte-selective carns1-KO mouse models, we found that HDs are critical for oligodendrocyte survival and protect against oxidative stress. Carns1-KO mouse models had lower numbers of mature oligodendrocytes, increased lipid peroxidation, and behavioral changes. Cuprizone administration, which increases reactive oxygen species in vivo, resulted in higher oligodendrocyte death, demyelination, axonal alterations, and oxidative damage in the corpus callosum of carns1-KO mice. Gliosis and oxidative damage by cuprizone were prevented by pretreatment with the antioxidant N-acetylcysteine. NADPH levels were increased threefold in the brains of carns1-KO mice as an antioxidant response to oxidative stress through acceleration of the pentose phosphate pathway (PPP). This was due to overexpression of glucose-6-phosphate dehydrogenase, the rate-limiting enzyme of the PPP. Likewise, expression of NAD kinase, the biosynthetic enzyme for NADP+, and NAMPT, which replenishes the NAD+ pool, was higher in carns1-KO mice brains than in controls. Our observations suggest that HDs cell-autonomously protect oligodendrocytes from oxidative stress, with implications for demyelinating diseases.
Collapse
Affiliation(s)
- Clara Sajrawi
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Maali Odeh
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Akshay K Tiwari
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Bella Agranovich
- Laura and Isaac Perlmutter Metabolomics Center, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ifat Abramovich
- Laura and Isaac Perlmutter Metabolomics Center, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Salman Zubedat
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| | - Galit Saar
- In vivo Imaging Unit, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Lihi Shaulov
- Electron Microscopy Unit, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Avi Avital
- Department of Occupational Therapy, Faculty of Social Welfare and Health Sciences, University of Haifa, Haifa, Israel
| | - Dan Reznik
- Data Science Consulting, Rio de Janeiro, RJ, Brazil
| | - Moran Benhar
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Inna Radzishevsky
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Simone Engelender
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Herman Wolosker
- Department of Biochemistry. Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Laura and Isaac Perlmutter Metabolomics Center, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
18
|
Maupin EA, Adams KL. Cellular Senescence in Glial Cells: Implications for Multiple Sclerosis. J Neurochem 2025; 169:e16301. [PMID: 39831743 PMCID: PMC11745082 DOI: 10.1111/jnc.16301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 12/16/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Aging is the most common risk factor for Multiple Sclerosis (MS) disease progression. Cellular senescence, the irreversible state of cell cycle arrest, is the main driver of aging and has been found to accumulate prematurely in neurodegenerative diseases, including Alzheimer's and Parkinson's disease. Cellular senescence in the central nervous system of MS patients has recently gained attention, with several studies providing evidence that demyelination induces cellular senescence, with common hallmarks of p16INK4A and p21 expression, oxidative stress, and senescence-associated secreted factors. Here we discuss the current evidence of cellular senescence in animal models of MS and different glial populations in the central nervous system, highlighting the major gaps in the field that still remain. As premature senescence in MS may exacerbate demyelination and inflammation, resulting in inhibition of myelin repair, it is critical to increase understanding of cellular senescence in vivo, the functional effects of senescence on glial cells, and the impact of removing senescent cells on remyelination and MS. This emerging field holds promise for opening new avenues of treatment for MS patients.
Collapse
Affiliation(s)
- Elizabeth A. Maupin
- Department of Biological SciencesUniversity of Notre DameNotre DameIndianaUSA
| | - Katrina L. Adams
- Department of Biological SciencesUniversity of Notre DameNotre DameIndianaUSA
- The Center for Stem Cells and Regenerative MedicineUniversity of Notre DameNotre DameIndianaUSA
| |
Collapse
|
19
|
Sun R, Ma T, Zhao Z, Gao Y, Feng J, Yang X. Phospholipase D Family Member 4 Regulates Microglial Phagocytosis and Remyelination via the AKT Pathway in a Cuprizone-Induced Multiple Sclerosis Mouse Model. CNS Neurosci Ther 2024; 30:e70111. [PMID: 39548665 PMCID: PMC11567942 DOI: 10.1111/cns.70111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/30/2024] [Accepted: 10/23/2024] [Indexed: 11/18/2024] Open
Abstract
AIMS Remyelination is an endogenous repair process that is often deficient in multiple sclerosis (MS). Stimulation of remyelination is thought to help limit the progression of MS. This study aimed to investigate the expression pattern and function of a microglial phagocytosis-related gene, phospholipase D family member 4 (PLD4), in a cuprizone (CPZ)-induced MS mouse model. METHODS The extent of remyelination was assessed using LFB staining. Myelin phagocytosis assay was used to investigate the effect of Pld4 on microglial phagocytic activity. RESULTS Pld4 was upregulated in the corpus callosum during demyelination and remyelination. AAV9-mediated Pld4 deficiency impaired remyelination and reduced the number of Olig2-positive cells. In the corpus callosum of Pld4-deficient mice, the microglial phagocytosis marker MAC2 was reduced, accompanied by inhibition of TrkA/AKT signaling. Similarly, the phagocytosis assay showed that Pld4 knockdown significantly inhibited myelin debris phagocytosis by BV2 cells. The AKT activator SC79 reversed the Pld4 deficiency-induced inhibition of microglial phagocytic activity and rescued the impaired remyelination in Pld4-deficient mice. CONCLUSION PLD4 is upregulated in CPZ-induced MS and modulates microglial phagocytosis and remyelination via the AKT pathway. Our findings provide experimental evidence for a better understanding of the molecular mechanism of MS.
Collapse
Affiliation(s)
- Ran Sun
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Tengyun Ma
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Zheng Zhao
- Department of Emergency MedicineShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Yan Gao
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Juan Feng
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| | - Xue Yang
- Department of NeurologyShengjing Hospital of China Medical UniversityShenyangPeople's Republic of China
| |
Collapse
|
20
|
Mwema A, Gratpain V, Ucakar B, Vanvarenberg K, Perdaens O, van Pesch V, Muccioli GG, des Rieux A. Impact of calcitriol and PGD 2-G-loaded lipid nanocapsules on oligodendrocyte progenitor cell differentiation and remyelination. Drug Deliv Transl Res 2024; 14:3128-3146. [PMID: 38366115 DOI: 10.1007/s13346-024-01535-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/26/2024] [Indexed: 02/18/2024]
Abstract
Multiple sclerosis (MS) is a demyelinating and inflammatory disease of the central nervous system (CNS) in need of a curative treatment. MS research has recently focused on the development of pro-remyelinating treatments and neuroprotective therapies. Here, we aimed at favoring remyelination and reducing neuro-inflammation in a cuprizone mouse model of brain demyelination using nanomedicines. We have selected lipid nanocapsules (LNC) coated with the cell-penetrating peptide transactivator of translation (TAT), loaded with either a pro-remyelinating compound, calcitriol (Cal-LNC TAT), or an anti-inflammatory bioactive lipid, prostaglandin D2-glycerol ester (PGD2-G) (PGD2-G-LNC TAT). Following the characterization of these formulations, we showed that Cal-LNC TAT in combination with PGD2-G-LNC TAT increased the mRNA expression of oligodendrocyte differentiation markers both in the CG-4 cell line and in primary mixed glial cell (MGC) cultures. However, while the combination of Cal-LNC TAT and PGD2-G-LNC TAT showed promising results in vitro, no significant impact, in terms of remyelination, astrogliosis, and microgliosis, was observed in vivo in the corpus callosum of cuprizone-treated mice following intranasal administration. Thus, although calcitriol's beneficial effects have been abundantly described in the literature in the context of MS, here, we show that the different doses of calcitriol tested had a negative impact on the mice well-being and showed no beneficial effect in the cuprizone model in terms of remyelination and neuro-inflammation, alone and when combined with PGD2-G-LNC TAT.
Collapse
Affiliation(s)
- Ariane Mwema
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Viridiane Gratpain
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Bernard Ucakar
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Kevin Vanvarenberg
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium
| | - Océane Perdaens
- Cellular and Molecular Division, Institute of Neuroscience, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 53, 1200, Brussels, Belgium
| | - Vincent van Pesch
- Cellular and Molecular Division, Institute of Neuroscience, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 53, 1200, Brussels, Belgium
| | - Giulio G Muccioli
- Bioanalysis and Pharmacology of Bioactive Lipids, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium.
| | - Anne des Rieux
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, UCLouvain, Avenue E. Mounier 73, 1200, Brussels, Belgium.
| |
Collapse
|
21
|
Maktabi B, Collins A, Safee R, Bouyer J, Wisner AS, Williams FE, Schiefer IT. Zebrafish as a Model for Multiple Sclerosis. Biomedicines 2024; 12:2354. [PMID: 39457666 PMCID: PMC11504653 DOI: 10.3390/biomedicines12102354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Zebrafish have become a key model organism in neuroscience research because of their unique advantages. Their genetic, anatomical, and physiological similarities to humans, coupled with their rapid development and transparent embryos, make them an excellent tool for investigating various aspects of neurobiology. They have specifically emerged as a valuable and versatile model organism in biomedical research, including the study of neurological disorders such as multiple sclerosis. Multiple sclerosis is a chronic autoimmune disease known to cause damage to the myelin sheath that protects the nerves in the brain and spinal cord. Objective: This review emphasizes the importance of continued research in both in vitro and in vivo models to advance our understanding of MS and develop effective treatments, ultimately improving the quality of life for those affected by this debilitating disease. Conclusions: Recent studies show the significance of zebrafish as a model organism for investigating demyelination and remyelination processes, providing new insights into MS pathology and potential therapies.
Collapse
Affiliation(s)
- Briana Maktabi
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Abigail Collins
- Center for Drug Design and Development 3, University of Toledo, Toledo, OH 43614, USA
| | - Raihaanah Safee
- Department of Pharmacy Practice, University of Toledo, Toledo, OH 43614, USA
| | - Jada Bouyer
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Alexander S. Wisner
- Center for Drug Design and Development 3, University of Toledo, Toledo, OH 43614, USA
| | - Frederick E. Williams
- Department of Pharmacology and Experimental Therapeutics, University of Toledo, Toledo, OH 43614, USA
| | - Isaac T. Schiefer
- Department of Pharmacy Practice, University of Toledo, Toledo, OH 43614, USA
- Department of Medicinal and Biological Chemistry, University of Toledo, Toledo, OH 43614, USA
| |
Collapse
|
22
|
Abdelalim LR, Elnaggar YSR, Abdallah OY. Lactoferrin, chitosan double-coated oleosomes loaded with clobetasol propionate for remyelination in multiple sclerosis: Physicochemical characterization and in-vivo assessment in a cuprizone-induced demyelination model. Int J Biol Macromol 2024; 277:134144. [PMID: 39053824 DOI: 10.1016/j.ijbiomac.2024.134144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 07/10/2024] [Accepted: 07/23/2024] [Indexed: 07/27/2024]
Abstract
Multiple sclerosis is a chronic inflammatory demyelinating disorder of the CNS characterized by continuous myelin damage accompanied by deterioration in functions. Clobetasol propionate (CP) is the most potent topical corticosteroid with serious side effects related to systemic absorption. Previous studies introduced CP for remyelination without considering systemic toxicity. This work aimed at fabrication and optimization of double coated nano-oleosomes loaded with CP to achieve brain targeting through intranasal administration. The optimized formulation was coated with lactoferrin and chitosan for the first time. The obtained double-coated oleosomes had particle size (220.07 ± 0.77 nm), zeta potential (+30.23 ± 0.41 mV) along with antioxidant capacity 9.8 μM ascorbic acid equivalents. Double coating was well visualized by TEM and significantly decreased drug release. Three different doses of CP were assessed in-vivo using cuprizone-induced demyelination in C57Bl/6 mice. Neurobehavioral tests revealed improvement in motor and cognitive functions of mice in a dose-dependent manner. Histopathological examination of the brain showed about 2.3 folds increase in corpus callosum thickness in 0.3 mg/kg CP dose. Moreover, the measured biomarkers highlighted the significant antioxidant and anti-inflammatory capacity of the formulation. In conclusion, the elaborated biopolymer-integrating nanocarrier succeeded in remyelination with 6.6 folds reduction in CP dose compared to previous studies.
Collapse
Affiliation(s)
- Lamiaa R Abdelalim
- Department of Pharmaceutics and pharmaceutical technology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt
| | - Yosra S R Elnaggar
- Department of Pharmaceutics and pharmaceutical technology, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, Egypt; Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt.
| | - Ossama Y Abdallah
- Department of Pharmaceutics, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| |
Collapse
|
23
|
van de Wetering R, Bibi R, Biggerstaff A, Hong S, Pengelly B, Prisinzano TE, La Flamme AC, Kivell BM. Nalfurafine promotes myelination in vitro and facilitates recovery from cuprizone + rapamycin-induced demyelination in mice. Glia 2024; 72:1801-1820. [PMID: 38899723 DOI: 10.1002/glia.24583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 06/05/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024]
Abstract
The kappa opioid receptor has been identified as a promising therapeutic target for promoting remyelination. In the current study, we evaluated the ability of nalfurafine to promote oligodendrocyte progenitor cell (OPC) differentiation and myelination in vitro, and its efficacy in an extended, cuprizone-induced demyelination model. Primary mouse (C57BL/6J) OPC-containing cultures were treated with nalfurafine (0.6-200 nM), clemastine (0.01-100 μM), T3 (30 ng/mL), or vehicle for 5 days. Using immunocytochemistry and confocal microscopy, we found that nalfurafine treatment increased OPC differentiation, oligodendrocyte (OL) morphological complexity, and myelination of nanofibers in vitro. Adult male mice (C57BL/6J) were given a diet containing 0.2% cuprizone and administered rapamycin (10 mg/kg) once daily for 12 weeks followed by 6 weeks of treatment with nalfurafine (0.01 or 0.1 mg/kg), clemastine (10 mg/kg), or vehicle. We quantified the number of OLs using immunofluorescence, gross myelination using black gold staining, and myelin thickness using electron microscopy. Cuprizone + rapamycin treatment produced extensive demyelination and was accompanied by a loss of mature OLs, which was partially reversed by therapeutic administration of nalfurafine. We also assessed these mice for functional behavioral changes in open-field, horizontal bar, and mouse motor skill sequence tests (complex wheel running). Cuprizone + rapamycin treatment resulted in hyperlocomotion, poorer horizontal bar scores, and less distance traveled on the running wheels. Partial recovery was observed on both the horizontal bar and complex running wheel tests over time, which was facilitated by nalfurafine treatment. Taken together, these data highlight the potential of nalfurafine as a remyelination-promoting therapeutic.
Collapse
Affiliation(s)
- Ross van de Wetering
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Rabia Bibi
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Andy Biggerstaff
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Sheein Hong
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Bria Pengelly
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| | - Thomas E Prisinzano
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky, USA
| | - Anne C La Flamme
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Bronwyn M Kivell
- School of Biological Sciences, Centre for Biodiscovery, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
24
|
Hilton JBW, Kysenius K, Liddell JR, Mercer SW, Rautengarten C, Hare DJ, Buncic G, Paul B, Murray SS, McLean CA, Kilpatrick TJ, Beckman JS, Ayton S, Bush AI, White AR, Roberts BR, Donnelly PS, Crouch PJ. Integrated elemental analysis supports targeting copper perturbations as a therapeutic strategy in multiple sclerosis. Neurotherapeutics 2024; 21:e00432. [PMID: 39164165 PMCID: PMC11579877 DOI: 10.1016/j.neurot.2024.e00432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/23/2024] [Accepted: 08/07/2024] [Indexed: 08/22/2024] Open
Abstract
Multiple sclerosis (MS) is a debilitating affliction of the central nervous system (CNS) that involves demyelination of neuronal axons and neurodegeneration resulting in disability that becomes more pronounced in progressive forms of the disease. The involvement of neurodegeneration in MS underscores the need for effective neuroprotective approaches necessitating identification of new therapeutic targets. Herein, we applied an integrated elemental analysis workflow to human MS-affected spinal cord tissue utilising multiple inductively coupled plasma-mass spectrometry methodologies. These analyses revealed shifts in atomic copper as a notable aspect of disease. Complementary gene expression and biochemical analyses demonstrated that changes in copper levels coincided with altered expression of copper handling genes and downstream functionality of cuproenzymes. Copper-related problems observed in the human MS spinal cord were largely reproduced in the experimental autoimmune encephalomyelitis (EAE) mouse model during the acute phase of disease characterised by axonal demyelination, lesion formation, and motor neuron loss. Treatment of EAE mice with the CNS-permeant copper modulating compound CuII(atsm) resulted in recovery of cuproenzyme function, improved myelination and lesion volume, and neuroprotection. These findings support targeting copper perturbations as a therapeutic strategy for MS with CuII(atsm) showing initial promise.
Collapse
Affiliation(s)
- James B W Hilton
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
| | - Kai Kysenius
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia; Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Jeffrey R Liddell
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
| | - Stephen W Mercer
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
| | | | - Dominic J Hare
- Atomic Medicine Initiative, University of Technology Sydney, Australia
| | - Gojko Buncic
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| | - Bence Paul
- School of Geography, Earth and Atmospheric Sciences, The University of Melbourne, Victoria 3010, Australia; Elemental Scientific Lasers, LLC, 685 Old Buffalo Trail, Bozeman, MT 59715, United States
| | - Simon S Murray
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
| | | | - Trevor J Kilpatrick
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Joseph S Beckman
- Linus Pauling Institute, Department of Biochemistry and Biophysics, Oregon State University, 97331, United States
| | - Scott Ayton
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia; Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Ashley I Bush
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia; Melbourne Dementia Research Centre, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Anthony R White
- Queensland Institute of Medical Research Berghofer, Herston, Queensland 4006, Australia
| | - Blaine R Roberts
- Department of Biochemistry, Emory University, Atlanta, GA 30322, United States
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| | - Peter J Crouch
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia.
| |
Collapse
|
25
|
Kushwaha S, Saji J, Verma R, Singh V, Ansari JA, Mishra SK, Roy O, Patnaik S, Ghosh D. Microglial Neuroinflammation-Independent Reversal of Demyelination of Corpus Callosum by Arsenic in a Cuprizone-Induced Demyelinating Mouse Model. Mol Neurobiol 2024; 61:6822-6841. [PMID: 38353925 DOI: 10.1007/s12035-024-03978-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 01/19/2024] [Indexed: 08/22/2024]
Abstract
Demyelination is the loss of myelin in CNS, resulting in damaged myelin sheath. Oxidative stress and neuroinflammation play a key role in inducing demyelinating diseases like MS; hence, controlling oxidative stress and neuroinflammation is important. Cuprizone (CPZ), a copper chelator, generates oxidative stress and neuroinflammation, thereby inducing demyelination. Therefore, the CPZ-induced demyelinating mouse model (CPZ model) is widely used in research. The present study was intended to unravel a mechanism of inhibition of demyelination by arsenic in a CPZ model, which is otherwise known for its toxicity. We investigated an alternative mechanism of inhibition of demyelination by arsenic through the reversal of SOD1 activity employing in silico analysis, analytical chemistry techniques, and in vitro and in vivo experiments. In vivo experiments showed protection of body weight, survivability, and myelination of the corpus callosum in CPZ and arsenic-co-exposed animals, where neuroinflammation was apparently not involved. In vitro experiments revealed that arsenic-mediated reversal of impaired SOD1 activity leads to reduced cellular ROS levels and better viability of primary oligodendrocytes. Reversal of SOD1 activity was also observed in the corpus callosum tissue isolated from experimental animals. In silico and analytical chemistry studies revealed that similar to copper, arsenic can potentially bind to CPZ and thereby make the copper freely available for SOD1 activity. Suitable neurobehavior tests further validated the protective effect of arsenic. Taken together, the present study revealed that arsenic protects oligodendrocytes and demyelination of corpus callosum by reversing CPZ-induced impaired SOD1 activity.
Collapse
Affiliation(s)
- Shaivya Kushwaha
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Joel Saji
- Water Analysis Laboratory, Drug and Chemical Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, 226001, India
| | - Rahul Verma
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- Water Analysis Laboratory, Drug and Chemical Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, 226001, India
| | - Vikas Singh
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, 44195, USA
| | - Jamal Ahmad Ansari
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
- Genome Instability and Chromatin Remodeling Section, NIH-National Institute of Aging, Baltimore, USA
| | - Shubhendra Kumar Mishra
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
- Department of Pediatrics, Division of Neonatology, McGill University Health Centre-Research Institute (RI-MUHC), Montreal, QC, Canada
| | - Opalina Roy
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India
| | - Satyakam Patnaik
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
- Water Analysis Laboratory, Drug and Chemical Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research, Lucknow, Uttar Pradesh, 226001, India.
| | - Debabrata Ghosh
- Immunotoxicology Laboratory, System Toxicology Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhawan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
26
|
Jansen MI, Mahmood Y, Lee J, Broome ST, Waschek JA, Castorina A. Targeting the PAC1 receptor mitigates degradation of myelin and synaptic markers and diminishes locomotor deficits in the cuprizone demyelination model. J Neurochem 2024; 168:3250-3267. [PMID: 39115025 DOI: 10.1111/jnc.16199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 10/04/2024]
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system with a strong neuroinflammatory component. Current treatments principally target the immune system but fail to preserve long-term myelin health and do not prevent neurological decline. Studies over the past two decades have shown that the structurally related neuropeptides VIP and PACAP (vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide, respectively) exhibit pronounced anti-inflammatory activities and reduce clinical symptoms in MS disease models, largely via actions on their bivalent VIP receptor type 1 and 2. Here, using the cuprizone demyelination model, we demonstrate that PACAP and VIP, and strikingly the PACAP-selective receptor PAC1 agonist maxadilan, prevented locomotor deficits in the horizontal ladder and open field tests. Moreover, only PACAP and maxadilan were able to prevent myelin deterioration, as assessed by a reduction in the expression of the myelin markers proteolipid protein 1, oligodendrocyte transcription factor 2, quaking-7 (APC) and Luxol Fast Blue staining. Furthermore, PACAP and maxadilan (but not VIP), prevented striatal synaptic loss and diminished astrocyte and microglial activation in the corpus callosum of cuprizone-fed mice. In vitro, PACAP or maxadilan prevented lipopolysaccharide (LPS)-induced polarisation of primary astrocytes at 12-24 h, an effect that was not seen with maxadilan in LPS-stimulated microglia. Taken together, our data demonstrates for the first time that PAC1 agonists provide distinctive protective effects against white matter deterioration, neuroinflammation and consequent locomotor dysfunctions in the cuprizone model. The results indicate that targeting the PAC1 receptor may provide a path to treat myelin-related diseases in humans.
Collapse
Affiliation(s)
- Margo I Jansen
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Yasir Mahmood
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Jordan Lee
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Sarah Thomas Broome
- Semel Institute for Neuroscience and Human Behavior/Neuropsychiatric Institute, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, California, USA
| | - James A Waschek
- Semel Institute for Neuroscience and Human Behavior/Neuropsychiatric Institute, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, California, USA
| | - Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
27
|
Wang LB, Liao BY, Li YJ, Wang ZH, Yu Y, Li X, Zhang QH. Engineered PDGFA-ligand-modified exosomes delivery T3 for demyelinating disease targeted therapy. Exp Neurol 2024; 375:114730. [PMID: 38401853 DOI: 10.1016/j.expneurol.2024.114730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 02/06/2024] [Accepted: 02/14/2024] [Indexed: 02/26/2024]
Abstract
Demyelination is a proper syndrome in plenty of central nervous system (CNS) diseases, which is the main obstacle to recovery and still lacks an effective treatment. To overcome the limitations of the brain-blood barrier on drug permeability, we modified an exosome secreted by neural stem cells (NSCs), which had transfected with lentivirus armed with platelet-derived growth factors A (PDGFA)-ligand. Through the in vivo and in vitro exosomes targeting test, the migration ability to the lesion areas and OPCs significantly improved after ligand modification. Furthermore, the targeted exosomes loaded with 3,5, 30-L-triiodothyronine (T3) have a critical myelination ability in CNS development, administrated to the cuprizone animal model treatment. The data shows that the novel drug vector loaded with T3 significantly promotes remyelination compared with T3 alone. At the same time, it improved the CNS microenvironment by reducing astrogliosis, inhibiting pro-inflammatory microglia, and alleviating axon damage. This investigation provides a straightforward strategy to produce a targeting exosome and indicates a possible therapeutic manner for demyelinating disease.
Collapse
Affiliation(s)
- Li-Bin Wang
- Neurosurgery department of Huazhong University of Science and Technology Unions Shenzhen Hospital, Shenzhen Nanshan Hospital; Shenzhen 518052, China; The General Hospital of Ningxia Medical University, Ningxia Nervous System Disease Diagnosis & Treatment Engineering Technology Research Center, Yinchuan 750004, China
| | - Bao-Ying Liao
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China
| | - Yong-Jun Li
- The General Hospital of Ningxia Medical University, Ningxia Nervous System Disease Diagnosis & Treatment Engineering Technology Research Center, Yinchuan 750004, China
| | - Zhen-Hai Wang
- The General Hospital of Ningxia Medical University, Ningxia Nervous System Disease Diagnosis & Treatment Engineering Technology Research Center, Yinchuan 750004, China
| | - Yang Yu
- Neurosurgery department of Huazhong University of Science and Technology Unions Shenzhen Hospital, Shenzhen Nanshan Hospital; Shenzhen 518052, China
| | - Xing Li
- National Engineering Laboratory for Resource Development of Endangered Crude Drugs in Northwest China, The Key Laboratory of Medicinal Resources and Natural Pharmaceutical Chemistry, The Ministry of Education, College of Life Sciences, Shaanxi Normal University, Xi'an 710119, China.
| | - Qing-Hua Zhang
- Neurosurgery department of Huazhong University of Science and Technology Unions Shenzhen Hospital, Shenzhen Nanshan Hospital; Shenzhen 518052, China.
| |
Collapse
|
28
|
Khan MB, Alam H, Siddiqui S, Shaikh MF, Sharma A, Rehman A, Baban B, Arbab AS, Hess DC. Exercise Improves Cerebral Blood Flow and Functional Outcomes in an Experimental Mouse Model of Vascular Cognitive Impairment and Dementia (VCID). Transl Stroke Res 2024; 15:446-461. [PMID: 36689081 PMCID: PMC10363247 DOI: 10.1007/s12975-023-01124-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 12/14/2022] [Accepted: 01/04/2023] [Indexed: 01/24/2023]
Abstract
Vascular cognitive impairment and dementia (VCID) are a growing threat to public health without any known treatment. The bilateral common carotid artery stenosis (BCAS) mouse model is valid for VCID. Previously, we have reported that remote ischemic postconditioning (RIPostC) during chronic cerebral hypoperfusion (CCH) induced by BCAS increases cerebral blood flow (CBF), improves cognitive function, and reduces white matter damage. We hypothesized that physical exercise (EXR) would augment CBF during CCH and prevent cognitive impairment in the BCAS model. BCAS was performed in C57/B6 mice of both sexes to establish CCH. One week after the BCAS surgery, mice were randomized to treadmill exercise once daily or no EXR for four weeks. CBF was monitored with an LSCI pre-, post, and 4 weeks post-BCAS. Cognitive testing was performed for post-BCAS after exercise training, and brain tissue was harvested for histopathology and biochemical test. BCAS led to chronic hypoperfusion resulting in impaired cognitive function and other functional outcomes. Histological examination revealed that BCAS caused changes in neuronal morphology and cell death in the cortex and hippocampus. Immunoblotting showed that BCAS was associated with a significant downregulate of AMPK and pAMPK and NOS3 and pNOS3. BCAS also decreased red blood cell (RBC) deformability. EXR therapy increased and sustained improved CBF and cognitive function, muscular strength, reduced cell death, and loss of white matter. EXR is effective in the BCAS model, improving CBF and cognitive function, reducing white matter damage, improving RBC deformability, and increasing RBC NOS3 and AMPK. The mechanisms by which EXR improves CBF and attenuates tissue damage need further investigation.
Collapse
Affiliation(s)
- Mohammad Badruzzaman Khan
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15thStreet, CA 1053, Augusta, GA, 30912, USA.
| | - Haroon Alam
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15thStreet, CA 1053, Augusta, GA, 30912, USA
| | - Shahneela Siddiqui
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15thStreet, CA 1053, Augusta, GA, 30912, USA
| | - Muhammad Fasih Shaikh
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15thStreet, CA 1053, Augusta, GA, 30912, USA
| | - Abhinav Sharma
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15thStreet, CA 1053, Augusta, GA, 30912, USA
| | - Amna Rehman
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15thStreet, CA 1053, Augusta, GA, 30912, USA
| | - Babak Baban
- Department of Oral Biology, Dental College of Georgia, Augusta University, Augusta, GA, 30912, USA
| | - Ali S Arbab
- Georgia Cancer Center, Augusta University, Augusta, GA, 30912, USA
| | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15thStreet, CA 1053, Augusta, GA, 30912, USA
| |
Collapse
|
29
|
Nicola MA, Attaai AH, Abdel-Raheem MH, Mohammed AF, Abu-Elhassan YF. Neuroprotective effects of rutin against cuprizone-induced multiple sclerosis in mice. Inflammopharmacology 2024; 32:1295-1315. [PMID: 38512652 PMCID: PMC11006763 DOI: 10.1007/s10787-024-01442-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2023] [Accepted: 01/24/2024] [Indexed: 03/23/2024]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory neurodegenerative disease of the central nervous system that injures the myelin sheath, provoking progressive axonal degeneration and functional impairments. No efficient therapy is available at present to combat such insults, and hence, novel safe and effective alternatives for MS therapy are extremely required. Rutin (RUT) is a flavonoid that exhibits antioxidant, anti-inflammatory, and neuroprotective effects in several brain injuries. The present study evaluated the potential beneficial effects of two doses of RUT in a model of pattern-III lesion of MS, in comparison to the conventional standard drug; dimethyl fumarate (DMF). Demyelination was induced in in male adult C57BL/6 mice by dietary 0.2% (w/w) cuprizone (CPZ) feeding for 6 consecutive weeks. Treated groups received either oral RUT (50 or 100 mg/kg) or DMF (15 mg/kg), along with CPZ feeding, for 6 consecutive weeks. Mice were then tested for behavioral changes, followed by biochemical analyses and histological examinations of the corpus callosum (CC). Results revealed that CPZ caused motor dysfunction, demyelination, and glial activation in demyelinated lesions, as well as significant oxidative stress, and proinflammatory cytokine elevation. Six weeks of RUT treatment significantly improved locomotor activity and motor coordination. Moreover, RUT considerably improved remyelination in the CC of CPZ + RUT-treated mice, as revealed by luxol fast blue staining and transmission electron microscopy. Rutin also significantly attenuated CPZ-induced oxidative stress and inflammation in the CC of tested animals. The effect of RUT100 was obviously more marked than either that of DMF, regarding most of the tested parameters, or even its smaller tested dose. In silico docking revealed that RUT binds tightly within NF-κB at the binding site of the protein-DNA complex, with a good negative score of -6.79 kcal/mol. Also, RUT-Kelch-like ECH-associated protein 1 (Keap1) model clarifies the possible inhibition of Keap1-Nrf2 protein-protein interaction. Findings of the current study provide evidence for the protective effect of RUT in CPZ-induced demyelination and behavioral dysfunction in mice, possibly by modulating NF-κB and Nrf2 signaling pathways. The present study may be one of the first to indicate a pro-remyelinating effect for RUT, which might represent a potential additive benefit in treating MS.
Collapse
Affiliation(s)
- Mariam A Nicola
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Asyût, 71526, Egypt.
| | - Abdelraheim H Attaai
- Department of Anatomy and Histology, School of Veterinary Medicine, Badr University in Assiut, New Nasser City, West of Assiut, Asyût, Egypt
- Department of Anatomy and Embryology, Faculty of Veterinary Medicine, Assiut University, Asyût, 71526, Egypt
| | | | - Anber F Mohammed
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Assiut University, Asyût, 71526, Egypt
| | - Yasmin F Abu-Elhassan
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Assiut University, Asyût, 71526, Egypt
| |
Collapse
|
30
|
Chen S, Liu S, Huang Y, Huang S, Zhang W, Xie H, Lu L. 5Z-7-Oxozaenol attenuates cuprizone-induced demyelination in mice through microglia polarization regulation. Brain Behav 2024; 14:e3487. [PMID: 38648385 PMCID: PMC11034864 DOI: 10.1002/brb3.3487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/06/2023] [Accepted: 02/21/2024] [Indexed: 04/25/2024] Open
Abstract
INTRODUCTION Demyelination is a key factor in axonal degeneration and neural loss, leading to disability in multiple sclerosis (MS) patients. Transforming growth factor beta activated kinase 1 (TAK1) is a critical molecule involved in immune and inflammatory signaling pathways. Knockout of microglia TAK1 can inhibit autoimmune inflammation of the brain and spinal cord and improve the outcome of MS. However, it is unclear whether inhibiting TAK1 can alleviate demyelination. METHODS Eight-week-old male c57bl/6j mice were randomly divided into five groups: (a) the control group, (b) the group treated with cuprizone (CPZ) only, (c) the group treated with 5Z-7-Oxozaenol (OZ) only, and (d) the group treated with both cuprizone and 15 μg/30 μg OZ. Demyelination in the mice of this study was induced by administration of CPZ (ig) at a daily dose of 400 mg/kg for consecutive 5 weeks. OZ was intraperitoneally administered at mentioned doses twice a week, starting from week 3 after beginning cuprizone treatment. Histology, rotarod test, grasping test, pole test, Western blot, RT-PCR, and ELISA were used to evaluate corpus callosum demyelination, behavioral impairment, oligodendrocyte differentiation, TAK1 signaling pathway expression, microglia, and related cytokines. RESULTS Our results demonstrated that OZ protected against myelin loss and behavior impairment caused by CPZ. Additionally, OZ rescued the loss of oligodendrocytes in CPZ-induced mice. OZ inhibited the activation of JNK, p65, and p38 pathways, transformed M1 polarized microglia into M2 phenotype, and increased brain-derived neurotrophic factor (BDNF) expression to attenuate demyelination in CPZ-treated mice. Furthermore, OZ reduced the expression of proinflammatory cytokines and increases anti-inflammatory cytokines in CPZ-treated mice. CONCLUSION These findings suggest that inhibiting TAK1 may be an effective approach for treating demyelinating diseases.
Collapse
Affiliation(s)
- Shiyu Chen
- Department of NeurologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
- Department of General PracticeZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Siyao Liu
- Department of General PracticeZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Yalun Huang
- Department of NeurologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Shiwen Huang
- Department of NeurologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Wanzhou Zhang
- Department of NeurologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Huifang Xie
- Department of NeurologyZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| | - Lingli Lu
- Department of General PracticeZhujiang Hospital, Southern Medical UniversityGuangzhouChina
| |
Collapse
|
31
|
Fu JT, Yang CJ, Lee LY, Chen WP, Chen YW, Chen CC, Sun YT, Yang CS, Tzeng SF. Erinacine S, a small active component derived from Hericium erinaceus, protects oligodendrocytes and alleviates mood abnormalities in cuprizone-exposed rodents. Biomed Pharmacother 2024; 173:116297. [PMID: 38394854 DOI: 10.1016/j.biopha.2024.116297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Revised: 02/08/2024] [Accepted: 02/17/2024] [Indexed: 02/25/2024] Open
Abstract
Hericium erinaceus mycelium extract (HEM), containing erinacine A (HeA) and erinacine S (HeS), has shown promise in promoting the differentiation of oligodendrocyte precursor cells (OPCs) into mature oligodendrocytes (OLs), crucial for myelin production in the central nervous system (CNS). The main aim of this study was to characterize the protective effects of HEM and its components on OLs and myelin in demyelinating rodents by exposure to cuprizone (CPZ), a copper chelating agent commonly used to induce demyelination in the corpus callosum of the brain. Rats were fed by CPZ-containing diet and simultaneously orally administered HEM, HeA, or HeS on a daily basis for three weeks. We found that HEM and HeS preserved myelin and OLs in the corpus callosum of CPZ-fed rats, along with reduced microglia and astrocyte activation, and downregulated IL-1β expression. Furthermore, post-treatment with HeS, in mouse models with acute (6 weeks) or chronic (12 weeks) CPZ-induced demyelination demonstrated oral administration during the final 4 weeks (HeS4/6 or HeS4/12) effectively preserved myelin in the corpus callosum. Additionally, HeS4/6 and HeS4/12 inhibited anxious and depressive-like behaviors in CPZ-fed mice. In summary, simultaneous administration of HEM and HeS in rats during short-term CPZ intoxication preserved OLs and myelin. Furthermore, post-administration of HeS not only inhibited demyelination and gliosis but also alleviated anxiety and depression in both acute and chronic CPZ-fed mice. This study presents compelling evidence supporting the potential of HeS as a promising small active compound for protecting OLs and preserving myelin in demyelinating diseases associated with emotional disorders.
Collapse
Affiliation(s)
- Jing-Ting Fu
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Jou Yang
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Li-Ya Lee
- Biotech Research Institute, Grape King Biotechnology Inc, Taoyuan, Taiwan
| | - Wan-Ping Chen
- Biotech Research Institute, Grape King Biotechnology Inc, Taoyuan, Taiwan
| | - Yu-Wen Chen
- Biotech Research Institute, Grape King Biotechnology Inc, Taoyuan, Taiwan
| | - Chin-Chu Chen
- Biotech Research Institute, Grape King Biotechnology Inc, Taoyuan, Taiwan
| | - Yuan-Ting Sun
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chung-Shi Yang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan Town, Miaoli County, Taiwan
| | - Shun-Fen Tzeng
- Department of Life Sciences, College of Bioscience and Biotechnology, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
32
|
Chapman TW, Kamen Y, Piedra ET, Hill RA. Oligodendrocyte Maturation Alters the Cell Death Mechanisms That Cause Demyelination. J Neurosci 2024; 44:e1794232024. [PMID: 38395617 PMCID: PMC10977033 DOI: 10.1523/jneurosci.1794-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Myelinating oligodendrocytes die in human disease and early in aging. Despite this, the mechanisms that underly oligodendrocyte death are not resolved and it is also not clear whether these mechanisms change as oligodendrocyte lineage cells are undergoing differentiation and maturation. Here, we used a combination of intravital imaging, single-cell ablation, and cuprizone-mediated demyelination, in both female and male mice, to discover that oligodendrocyte maturation dictates the dynamics and mechanisms of cell death. After single-cell phototoxic damage, oligodendrocyte precursor cells underwent programmed cell death within hours, differentiating oligodendrocytes died over several days, while mature oligodendrocytes took weeks to die. Importantly cells at each maturation stage all eventually died but did so with drastically different temporal dynamics and morphological features. Consistent with this, cuprizone treatment initiated a caspase-3-dependent form of rapid cell death in differentiating oligodendrocytes, while mature oligodendrocytes never activated this executioner caspase. Instead, mature oligodendrocytes exhibited delayed cell death which was marked by DNA damage and disruption in poly-ADP-ribose subcellular localization. Thus, oligodendrocyte maturation plays a key role in determining the mechanism of death a cell undergoes in response to the same insult. This means that oligodendrocyte maturation is important to consider when designing strategies for preventing cell death and preserving myelin while also enhancing the survival of new oligodendrocytes in demyelinating conditions.
Collapse
Affiliation(s)
- Timothy W Chapman
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Yasmine Kamen
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Enrique T Piedra
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| | - Robert A Hill
- Department of Biological Sciences, Dartmouth College, Hanover, New Hampshire 03755
| |
Collapse
|
33
|
Zarini D, Pasbakhsh P, Mojaverrostami S, Amirizadeh S, Hashemi M, Shabani M, Noshadian M, Kashani IR. Microglia/macrophage polarization regulates spontaneous remyelination in intermittent cuprizone model of demyelination. Biochem Biophys Rep 2024; 37:101630. [PMID: 38234370 PMCID: PMC10793082 DOI: 10.1016/j.bbrep.2023.101630] [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: 10/28/2023] [Revised: 12/20/2023] [Accepted: 12/26/2023] [Indexed: 01/19/2024] Open
Abstract
Central nervous system (CNS) lesions can repeatedly be de-and remyelinated during demyelinating diseases such as multiple sclerosis (MS). Here, we designed an intermittent demyelination model by 0.3 % Cuprizone feeding in C57/BL6 mice followed by two weeks recovery. Histochemical staining of luxol fast blue (LFB) was used for study of remyelination, detection of glial and endothelial cells was performed by immunohistochemistry staining for the following antibodies: anti Olig2 for oligodendrocyte progenitor cells, anti APC for mature oligodendrocytes, anti GFAP for astrocytes, and anti Iba-1 for microglia/macrophages, anti iNOS for M1 microglia/macrophage phenotype, anti TREM-2 for M2 microglia/macrophage phenotype and anti CD31 for endothelial cells. Also, real-time polymerase chain reaction was performed for assessment of the expression of the targeted genes. LFB staining results showed enhanced remyelination in the intermittent cuprizone (INTRCPZ) group, which was accompanied by improved motor function, increased mature oligodendrocyte cells, and reduction of astrogliosis and microgliosis. Moreover, switching from M1 to M2 polarity increased in the INTRCPZ group that was in association with downregulation of pro-inflammatory and upregulation of anti-inflammatory genes. Finally, evaluation of microvascular changes revealed a remarkable decrease in the endothelial cells in the cuprizone (CPZ) group which recovered in the INTERCPZ group. The outcomes demonstrate enhanced myelin content during recovery in the intermittent demyelination model which is in association with reshaping macrophage polarity and modification of glial and endothelial cells.
Collapse
Affiliation(s)
- Davood Zarini
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Parichehr Pasbakhsh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sina Mojaverrostami
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Shiva Amirizadeh
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maedeh Hashemi
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Shabani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrazin Noshadian
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomy, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
34
|
Li J, Qi H, Chen Y, Zhu X. Epilepsy and demyelination: Towards a bidirectional relationship. Prog Neurobiol 2024; 234:102588. [PMID: 38378072 DOI: 10.1016/j.pneurobio.2024.102588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Demyelination stands out as a prominent feature in individuals with specific types of epilepsy. Concurrently, individuals with demyelinating diseases, such as multiple sclerosis (MS) are at a greater risk of developing epilepsy compared to non-MS individuals. These bidirectional connections raise the question of whether both pathological conditions share common pathogenic mechanisms. This review focuses on the reciprocal relationship between epilepsy and demyelination diseases. We commence with an overview of the neurological basis of epilepsy and demyelination diseases, followed by an exploration of how our comprehension of these two disorders has evolved in tandem. Additionally, we discuss the potential pathogenic mechanisms contributing to the interactive relationship between these two diseases. A more nuanced understanding of the interplay between epilepsy and demyelination diseases has the potential to unveiling the molecular intricacies of their pathological relationships, paving the way for innovative directions in future clinical management and treatment strategies for these diseases.
Collapse
Affiliation(s)
- Jiayi Li
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China; Clinical Medicine, Medical School of Southeast University, Nanjing, China
| | - Honggang Qi
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Yuzhou Chen
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China; Clinical Medicine, Medical School of Southeast University, Nanjing, China
| | - Xinjian Zhu
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China.
| |
Collapse
|
35
|
Kipp M. How to Use the Cuprizone Model to Study De- and Remyelination. Int J Mol Sci 2024; 25:1445. [PMID: 38338724 PMCID: PMC10855335 DOI: 10.3390/ijms25031445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune and inflammatory disorder affecting the central nervous system whose cause is still largely unknown. Oligodendrocyte degeneration results in demyelination of axons, which can eventually be repaired by a mechanism called remyelination. Prevention of demyelination and the pharmacological support of remyelination are two promising strategies to ameliorate disease progression in MS patients. The cuprizone model is commonly employed to investigate oligodendrocyte degeneration mechanisms or to explore remyelination pathways. During the last decades, several different protocols have been applied, and all have their pros and cons. This article intends to offer guidance for conducting pre-clinical trials using the cuprizone model in mice, focusing on discovering new treatment approaches to prevent oligodendrocyte degeneration or enhance remyelination.
Collapse
Affiliation(s)
- Markus Kipp
- Rostock University Medical Center, Institute of Anatomy, 18057 Rostock, Germany
| |
Collapse
|
36
|
Lei Z, Lin W. Mechanisms Governing Oligodendrocyte Viability in Multiple Sclerosis and Its Animal Models. Cells 2024; 13:116. [PMID: 38247808 PMCID: PMC10814231 DOI: 10.3390/cells13020116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/04/2024] [Accepted: 01/05/2024] [Indexed: 01/23/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune inflammatory demyelinating disease of the central nervous system (CNS), which is triggered by an autoimmune assault targeting oligodendrocytes and myelin. Recent research indicates that the demise of oligodendrocytes due to an autoimmune attack contributes significantly to the pathogenesis of MS and its animal model experimental autoimmune encephalomyelitis (EAE). A key challenge in MS research lies in comprehending the mechanisms governing oligodendrocyte viability and devising therapeutic approaches to enhance oligodendrocyte survival. Here, we provide an overview of recent findings that highlight the contributions of oligodendrocyte death to the development of MS and EAE and summarize the current literature on the mechanisms governing oligodendrocyte viability in these diseases.
Collapse
Affiliation(s)
- Zhixin Lei
- School of Chemistry, Chemical Engineering and Life Science, Wuhan University of Technology, Wuhan 430070, China;
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
37
|
Hilton JBW, Kysenius K, Liddell JR, Mercer SW, Hare DJ, Buncic G, Paul B, Wang Y, Murray SS, Kilpatrick TJ, White AR, Donnelly PS, Crouch PJ. Evidence for decreased copper associated with demyelination in the corpus callosum of cuprizone-treated mice. Metallomics 2024; 16:mfad072. [PMID: 38178638 PMCID: PMC10797489 DOI: 10.1093/mtomcs/mfad072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 11/24/2023] [Indexed: 01/06/2024]
Abstract
Demyelination within the central nervous system (CNS) is a significant feature of debilitating neurological diseases such as multiple sclerosis and administering the copper-selective chelatorcuprizone to mice is widely used to model demyelination in vivo. Conspicuous demyelination within the corpus callosum is generally attributed to cuprizone's ability to restrict copper availability in this vulnerable brain region. However, the small number of studies that have assessed copper in brain tissue from cuprizone-treated mice have produced seemingly conflicting outcomes, leaving the role of CNS copper availability in demyelination unresolved. Herein we describe our assessment of copper concentrations in brain samples from mice treated with cuprizone for 40 d. Importantly, we applied an inductively coupled plasma mass spectrometry methodology that enabled assessment of copper partitioned into soluble and insoluble fractions within distinct brain regions, including the corpus callosum. Our results show that cuprizone-induced demyelination in the corpus callosum was associated with decreased soluble copper in this brain region. Insoluble copper in the corpus callosum was unaffected, as were pools of soluble and insoluble copper in other brain regions. Treatment with the blood-brain barrier permeant copper compound CuII(atsm) increased brain copper levels and this was most pronounced in the soluble fraction of the corpus callosum. This effect was associated with significant mitigation of cuprizone-induced demyelination. These results provide support for the involvement of decreased CNS copper availability in demyelination in the cuprizone model. Relevance to human demyelinating disease is discussed.
Collapse
Affiliation(s)
- James B W Hilton
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
| | - Kai Kysenius
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Jeffrey R Liddell
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
| | - Stephen W Mercer
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
| | - Dominic J Hare
- Atomic Medicine Initiative, University of Technology Sydney, Sydney, New South Wales 2007, Australia
| | - Gojko Buncic
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| | - Bence Paul
- School of Earth Sciences, The University of Melbourne, Victoria 3010, Australia
| | - YouJia Wang
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
| | - Simon S Murray
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Trevor J Kilpatrick
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
- Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Victoria 3010, Australia
| | - Anthony R White
- Queensland Institute of Medical Research Berghofer, Herston, Queensland 4006, Australia
| | - Paul S Donnelly
- School of Chemistry and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria 3010, Australia
| | - Peter J Crouch
- Department of Anatomy & Physiology, The University of Melbourne, Victoria 3010, Australia
| |
Collapse
|
38
|
Alavi O, Alizadeh A, Dehghani F, Alipour H, Tanideh N. Anti-inflammatory Effects of Umbilical Cord Mesenchymal Stem Cell and Autologous Conditioned Serum on Oligodendrocyte, Astrocyte, and Microglial Specific Gene in Cuprizone Animal Model. Curr Stem Cell Res Ther 2024; 19:71-82. [PMID: 36852798 DOI: 10.2174/1574888x18666230228102731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 11/23/2022] [Accepted: 12/29/2022] [Indexed: 03/01/2023]
Abstract
BACKGROUND Inflammation, myelin loss, astrocytosis, and microgliosis are pathological signs of the autoimmune and demyelinating disease known as multiple sclerosis (MS). Axonal and neuronal degenerations have basic molecular pathways. The remyelination process can be influenced by the secretome of mesenchymal stem cells due to their capacity for immunomodulation, differentiation, and neuroprotection. Microglial cells are divided into two subgroups: M1 and M2 phenotypes. A crucial component of the microglial function is the colony stimulating factor 1 receptor (CSF1R). We aimed to evaluate the immunomodulating effects of secretome and conditioned serum on the microglial phenotypes and improvement of demyelination in a cuprizone model of MS. METHODS The study used 48 male C57BL/6 mice, which were randomly distributed into 6 subgroups (n = 8), i.e., control, cuprizone, MSC (confluency 40% and 80%) secretome group, and blood derived conditioned serum (autologous and humanized). The animals were fed with 0.2% cuprizone diet for 12 weeks. Supplements were injected into the lateral tail vein using a 27-gauge needle every 3 days 500 μl per injection. RESULTS At 14 days after transplantation, animals from each group were sacrificed and analyzed by Real time PCR. The results showed that the administration of MSC secretome can efficiently reduce expression of pro-inflammatory cytokines (IL-1, IL6 and TNF-α) in the corpus callosum; also, conditioned serum downregulated IL-1. Moreover, the oligodendrocyte-specific gene was upregulated by secretome and conditioned serum treatment. Also, the expression of microglial- specific gene was reduced after treatment. CONCLUSION These findings demonstrated that the secretome isolated from MSCs used as a therapy decreased and increased the M1 and M2 levels, respectively, to control neuroinflammation in CPZ mice. In conclusion, the current study showed the viability of devising a method to prepare suitable MSCs and secreted factor to cure neurodegenerative diseases, as well as the capability of regulating MSC secretome patterns by manipulating the cell density.
Collapse
Affiliation(s)
- Omid Alavi
- Department of Tissue Engineering, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Aliakbar Alizadeh
- Department of Tissue Engineering, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Dehghani
- Department of Tissue Engineering, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Histomorphometry and Stereology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Anatomical Sciences, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Alipour
- Department of Tissue Engineering, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Nader Tanideh
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iranaz Iran
- Department of Pharmacology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
39
|
Gadhave DG, Sugandhi VV, Kokare CR. Potential biomaterials and experimental animal models for inventing new drug delivery approaches in the neurodegenerative disorder: Multiple sclerosis. Brain Res 2024; 1822:148674. [PMID: 37952871 DOI: 10.1016/j.brainres.2023.148674] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 09/14/2023] [Accepted: 11/07/2023] [Indexed: 11/14/2023]
Abstract
The tight junction of endothelial cells in the central nervous system (CNS) has an ideal characteristic, acting as a biological barrier that can securely regulate the movement of molecules in the brain. Tightly closed astrocyte cell junctions on blood capillaries are the blood-brain barrier (BBB). This biological barrier prohibits the entry of polar drugs, cells, and ions, which protect the brain from harmful toxins. However, delivering any therapeutic agent to the brain in neurodegenerative disorders (i.e., schizophrenia, multiple sclerosis, etc.) is extremely difficult. Active immune responses such as microglia, astrocytes, and lymphocytes cross the BBB and attack the nerve cells, which causes the demyelination of neurons. Therefore, there is a hindrance in transmitting electrical signals properly, resulting in blindness, paralysis, and neuropsychiatric problems. The main objective of this article is to shed light on the performance of biomaterials, which will help researchers to create nanocarriers that can cross the blood-brain barrier and achieve a therapeutic concentration of drugs in the CNS of patients with multiple sclerosis (MS). The present review focuses on the importance of biomaterials with diagnostic and therapeutic efficacy that can help enhance multiple sclerosis therapeutic potential. Currently, the development of MS in animal models is limited by immune responses, which prevent MS induction in healthy animals. Therefore, this article also showcases animal models currently used for treating MS. A future advance in developing a novel effective strategy for treating MS is now a potential area of research.
Collapse
Affiliation(s)
- Dnyandev G Gadhave
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India; Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA; Department of Pharmaceutics, Dattakala Shikshan Sanstha's, Dattakala College of Pharmacy (Affiliated to Savitribai Phule Pune University), Swami Chincholi, Daund, Pune 413130, Maharashtra, India.
| | - Vrashabh V Sugandhi
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY 11439, USA
| | - Chandrakant R Kokare
- Department of Pharmaceutics, Sinhgad Technical Education Society's, Sinhgad Institute of Pharmacy (Affiliated to Savitribai Phule Pune University), Narhe, Pune 411041, Maharashtra, India
| |
Collapse
|
40
|
Safwat SM, Aboonq MS, El Tohamy M, Mojaddidi M, Al-Qahtani SAM, Zakari MO, ElGendy AA, Hussein AM. New Insight into the Possible Roles of L-Carnitine in a Rat Model of Multiple Sclerosis. Brain Sci 2023; 14:23. [PMID: 38248238 PMCID: PMC10813446 DOI: 10.3390/brainsci14010023] [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: 10/29/2023] [Revised: 12/10/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
OBJECTIVE We investigated the effect of L-carnitine (LC) on cuprizone (Cup) demyelinating rat model and its possible underlying mechanisms. METHODS Thirty male Sprague-Dawley (SD) rats were randomly allocated to three groups: the normal control group; the Cup group, in which Cup was administrated at a dose of 450 mg/kg per day orally via gastric gavage for 5 weeks; and the Cup + LC group, which received the same dose of Cup as the Cup group, except that the rats were treated additionally with LC 100 mg/kg/day orally for 5 weeks. The nerve conduction (NCV) in isolated sciatic nerves was measured; then, the sciatic nerves were isolated for H&E staining and electron microscope examination. The expression of myelin basic protein (MBP), IL-1β, p53, iNOS, and NF-KB by immunohistochemistry was detected in the isolated nerves. A PCR assay was also performed to detect the expression of antioxidant genes Nrf2 and HO-1. In addition, the level of IL-17 was measured by ELISA. RESULTS There was a significant reduction in NCV in the Cup group compared to normal rats (p < 0.001), which was significantly improved in the LC group (p < 0.001). EM and histopathological examination revealed significant demyelination and deterioration of the sciatic nerve fibers, with significant improvement in the LC group. The level of IL-17 as well as the expression of IL-1β, p53, iNOS, and NF-KB were significantly increased, with significant reduction expression of MBP in the sciatic nerves (p < 0.01), and LC treatment significantly improved the studied parameters (p < 0.01). CONCLUSION The current study demonstrates a neuroprotective effect of LC in a Cup-induced demyelinating rat model. This effect might be due to its anti-inflammatory and antioxidant actions.
Collapse
Affiliation(s)
- Sally M. Safwat
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (S.M.S.); (M.E.T.); (A.A.E.)
| | - Moutasem Salih Aboonq
- Department of Medical Physiology, College of Medicine, Taibah University, KSA, Medina 42353, Saudi Arabia; (M.S.A.); (M.M.); (S.A.M.A.-Q.); (M.O.Z.)
| | - Mahmoud El Tohamy
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (S.M.S.); (M.E.T.); (A.A.E.)
| | - Moaz Mojaddidi
- Department of Medical Physiology, College of Medicine, Taibah University, KSA, Medina 42353, Saudi Arabia; (M.S.A.); (M.M.); (S.A.M.A.-Q.); (M.O.Z.)
| | - Saeed Awad M. Al-Qahtani
- Department of Medical Physiology, College of Medicine, Taibah University, KSA, Medina 42353, Saudi Arabia; (M.S.A.); (M.M.); (S.A.M.A.-Q.); (M.O.Z.)
| | - Madaniah Omar Zakari
- Department of Medical Physiology, College of Medicine, Taibah University, KSA, Medina 42353, Saudi Arabia; (M.S.A.); (M.M.); (S.A.M.A.-Q.); (M.O.Z.)
| | - Ahmed A. ElGendy
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (S.M.S.); (M.E.T.); (A.A.E.)
- Department of Medical Physiology, College of Medicine, Taibah University, KSA, Medina 42353, Saudi Arabia; (M.S.A.); (M.M.); (S.A.M.A.-Q.); (M.O.Z.)
| | - Abdelaziz M. Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (S.M.S.); (M.E.T.); (A.A.E.)
| |
Collapse
|
41
|
Safwat SM, El Tohamy M, Aboonq MS, Alrehaili A, Assinnari AA, Bahashwan AS, ElGendy AA, Hussein AM. Vanillic Acid Ameliorates Demyelination in a Cuprizone-Induced Multiple Sclerosis Rat Model: Possible Underlying Mechanisms. Brain Sci 2023; 14:12. [PMID: 38248227 PMCID: PMC10813517 DOI: 10.3390/brainsci14010012] [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: 10/20/2023] [Revised: 12/09/2023] [Accepted: 12/20/2023] [Indexed: 01/23/2024] Open
Abstract
OBJECTIVE To investigate the effect of vanillic acid (VA) on a Cuprizone (Cup) demyelinating rat model and the mechanisms behind such effect. METHODS Thirty adult male Sprague Dawley (SD) rats were randomly divided into three groups: control, Cuprizone, and VA groups. Cuprizone was administrated at a dose of 450 mg/kg per day orally via gastric gavage for 5 weeks. The nerve conduction velocity (NCV) was studied in an isolated sciatic nerve, and then the sciatic nerve was isolated for histopathological examination, electron microscope examination, immunohistochemical staining, and biochemical and PCR assay. The level of IL17 was detected using ELISA, while the antioxidant genes Nrf2, HO-1 expression at the level of mRNA, expression of the myelin basic protein (MBP), interferon-gamma factor (INF)-γ and tumor necrosis factor (TNF)-α, and apoptotic marker (caspase-3) were measured using immunohistochemistry in the sciatic nerve. RESULTS There was a significant reduction in NCV in Cup compared to normal rats (p < 0.001), which was markedly improved in the VA group (p < 0.001). EM and histopathological examination revealed significant demyelination and deterioration of the sciatic nerve fibers with significant improvement in the VA group. The level of IL17 as well as the expression of INF-γ and caspase-3 were significantly increased with a significant reduction in the expression of MBP, Nrf2, and HO-1 in the sciatic nerve (p < 0.01), and VA treatment significantly improved the studied parameters (p < 0.01). CONCLUSION The current study demonstrated a neuroprotective effect for VA against the Cup-induced demyelinating rat model. This effect might be precipitated by the inhibition of inflammation, oxidative stress, and apoptosis.
Collapse
Affiliation(s)
- Sally M. Safwat
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (S.M.S.); (M.E.T.); (A.A.E.)
| | - Mahmoud El Tohamy
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (S.M.S.); (M.E.T.); (A.A.E.)
| | - Moutasem Salih Aboonq
- Department of Medical Physiology, College of Medicine, Taibah University, KSA, Madinah 42353, Saudi Arabia; (M.S.A.); (A.A.); (A.A.A.); (A.S.B.)
| | - Amaal Alrehaili
- Department of Medical Physiology, College of Medicine, Taibah University, KSA, Madinah 42353, Saudi Arabia; (M.S.A.); (A.A.); (A.A.A.); (A.S.B.)
| | - Ahmad A. Assinnari
- Department of Medical Physiology, College of Medicine, Taibah University, KSA, Madinah 42353, Saudi Arabia; (M.S.A.); (A.A.); (A.A.A.); (A.S.B.)
| | - Abdulrahman S. Bahashwan
- Department of Medical Physiology, College of Medicine, Taibah University, KSA, Madinah 42353, Saudi Arabia; (M.S.A.); (A.A.); (A.A.A.); (A.S.B.)
| | - Ahmed A. ElGendy
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (S.M.S.); (M.E.T.); (A.A.E.)
- Department of Medical Physiology, College of Medicine, Taibah University, KSA, Madinah 42353, Saudi Arabia; (M.S.A.); (A.A.); (A.A.A.); (A.S.B.)
| | - Abdelaziz M. Hussein
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura 35516, Egypt; (S.M.S.); (M.E.T.); (A.A.E.)
| |
Collapse
|
42
|
Wang M, Gu C, Yang Y, Chen L, Chen K, Du J, Wu H, Li Y. Ursolic acid derivative UAOS-Na treats experimental autoimmune encephalomyelitis by immunoregulation and protecting myelin. Front Neurol 2023; 14:1269862. [PMID: 38107649 PMCID: PMC10723162 DOI: 10.3389/fneur.2023.1269862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 11/15/2023] [Indexed: 12/19/2023] Open
Abstract
Introduction Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS). Ursolic acid (UA) can be used in the MS treatment with anti-inflammatory and neuroprotective activities. However, UA is insoluble in water, which may affect its medication effectiveness. In our previous study, UAOS-Na, a water-soluble derivative of UA was obtained. In this study, we evaluated the pharmacological effects and explored its underlying mechanism of UAOS-Na on experimental autoimmune encephalomyelitis (EAE). Methods Firstly, the pharmacodynamics of UAOS-Na was investigated in EAE and Cuprizone-induced mice. And then the possible mechanisms were investigated by TMT proteomics and verified by in vitro and in vivo experiments. Results UAOS-Na (30 mg/kg/d) delayed the onset time of EAE from 11.78 days post immunization (dpi) to 14.33 dpi, reduced the incidence from 90.0% to 42.9%. UAOS-Na (60 mg/kg/d) reduced the serum levels of IFN-γ, IL-17A, TNF-α and IL-6, reduced the mononuclear cell infiltration of spinal cord, and inhibited the overexpression of key transcription factors T-bet and ROR-γt of EAE mouse spinal cord. In addition, UAOS-Na attenuated demyelination and astrogliosis in the CNS of EAE and cuprizone-induced mice. Mechanistically, proteomics showed that 96 differential expression proteins (DEPs) were enriched and 94 were upregulated in EAE mice compared with normal group. After UAOS-Na treatment, 16 DEPs were enriched and 15 were downregulated, and these DEPs were markedly enriched in antigen processing and presentation (APP) signaling pathway. Moreover, UAOS-Na downregulated the protein levels of Tapbp and H2-T23 in MHC-I antigen presentation pathway and reduced the proliferation of splenic CD8 T cells, thereby inhibiting the CNS infiltration of CD8 T cells. Conclusion Our findings demonstrated that UAOS-Na has both myelin protective and anti-inflammatory effects. And it could reduce the inflammation of MS by downregulating the expression of Tapbp and H2-T23 in the MHC-I antigen presentation pathway.
Collapse
Affiliation(s)
- Maolin Wang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chenming Gu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yifu Yang
- Experiment Center for Science and Technology, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Liang Chen
- Nutrition Science, Amway (Shanghai) Innovation and Science Co., Ltd., Shanghai, China
| | - Kaixian Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun Du
- Nutrition Science, Amway (Shanghai) Innovation and Science Co., Ltd., Shanghai, China
| | - Huali Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiming Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
43
|
Gakare SG, Bhatt JM, Narasimhan KKS, Dravid SM. Glutamate delta-1 receptor regulates oligodendrocyte progenitor cell differentiation and myelination in normal and demyelinating conditions. PLoS One 2023; 18:e0294583. [PMID: 37983226 PMCID: PMC10659214 DOI: 10.1371/journal.pone.0294583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/05/2023] [Indexed: 11/22/2023] Open
Abstract
In this study, we investigated the role of glutamate delta 1 receptor (GluD1) in oligodendrocyte progenitor cell (OPC)-mediated myelination during basal (development) and pathophysiological (cuprizone-induced demyelination) conditions. Initially, we sought to determine the expression pattern of GluD1 in OPCs and found a significant colocalization of GluD1 puncta with neuron-glial antigen 2 (NG2, OPC marker) in the motor cortex and dorsal striatum. Importantly, we found that the ablation of GluD1 led to an increase in the number of myelin-associated glycoprotein (MAG+) cells in the corpus callosum and motor cortex at P40 without affecting the number of NG2+ OPCs, suggesting that GluD1 loss selectively facilitates OPC differentiation rather than proliferation. Further, deletion of GluD1 enhanced myelination in the corpus callosum and motor cortex, as indicated by increased myelin basic protein (MBP) staining at P40, suggesting that GluD1 may play an essential role in the developmental regulation of myelination during the critical window period. In contrast, in cuprizone-induced demyelination, we observed reduced MBP staining in the corpus callosum of GluD1 KO mice. Furthermore, cuprizone-fed GluD1 KO mice showed more robust motor deficits. Collectively, our results demonstrate that GluD1 plays a critical role in OPC regulation and myelination in normal and demyelinating conditions.
Collapse
Affiliation(s)
- Sukanya G. Gakare
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Jay M. Bhatt
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Kishore Kumar S. Narasimhan
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| | - Shashank M. Dravid
- Department of Pharmacology and Neuroscience, Creighton University School of Medicine, Omaha, NE, United States of America
| |
Collapse
|
44
|
Zhang X, Weickenmeier J. Brain Stiffness Follows Cuprizone-Induced Variations in Local Myelin Content. Acta Biomater 2023; 170:507-518. [PMID: 37660962 DOI: 10.1016/j.actbio.2023.08.033] [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: 01/25/2023] [Revised: 08/08/2023] [Accepted: 08/17/2023] [Indexed: 09/05/2023]
Abstract
Brain maturation and neurological diseases are intricately linked to microstructural changes that inherently affect the brain's mechanical behavior. Animal models are frequently used to explore relative brain stiffness changes as a function of underlying microstructure. Here, we are using the cuprizone mouse model to study indentation-derived stiffness changes resulting from acute and chronic demyelination during a 15-week observation period. We focus on the corpus callosum, cingulum, and cortex which undergo different degrees of de- and remyelination and, therefore, result in region-specific stiffness changes. Mean stiffness of the corpus callosum starts at 1.1 ± 0.3 kPa in untreated mice, then cuprizone treatment causes stiffness to drop to 0.6 ± 0.1 kPa by week 3, temporarily increase to 0.9 ± 0.3 kPa by week 6, and ultimately stabilize around 0.7 ± 0.1 kPa by week 9 for the rest of the observation period. The cingulum starts at 3.2 ± 0.9 kPa, then drops to 1.6 ± 0.4 kPa by week 3, and then gradually stabilizes around 1.4 ± 0.3 kPa by week 9. Cortical stiffness exhibits less stiffness variations overall; it starts at 4.2 ± 1.3 kPa, drops to 2.4 ± 0.6 kPa by week 3, and stabilizes around 2.7 ± 0.9 kPa by week 6. We also assess the impact of tissue fixation on indentation-based mechanical tissue characterization. On the one hand, fixation drastically increases untreated mean tissue stiffness by a factor of 3.3 for the corpus callosum, 2.9 for the cingulum, and 3.6 for the cortex; on the other hand, fixation influences interregional stiffness ratios during demyelination, thus suggesting that fixation affects individual brain tissues differently. Lastly, we determine the spatial correlation between stiffness measurements and myelin density and observe a region-specific proportionality between myelin content and tissue stiffness. STATEMENT OF SIGNIFICANCE: Despite extensive work, the relationship between microstructure and mechanical behavior in the brain remains mostly unknown. Additionally, the existing variation of measurement results reported in literature requires in depth investigation of the impact of individual cell and protein populations on tissue stiffness and interregional stiffness ratios. Here, we used microindentation measurements to show that brain stiffness changes with myelin density in the cuprizone-based demyelination mouse model. Moreover, we explored the impact of tissue fixation prior to mechanical characterization because of conflicting results reported in literature. We observe that fixation has a distinctly different impact on our three regions of interest, thus causing region-specific tissue stiffening and, more importantly, changing interregional stiffness ratios.
Collapse
Affiliation(s)
- Xuesong Zhang
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030 United States
| | - Johannes Weickenmeier
- Department of Mechanical Engineering, Stevens Institute of Technology, Hoboken, NJ 07030 United States.
| |
Collapse
|
45
|
Adebiyi OE, Bynoe MS. Roles of Adenosine Receptor (subtypes A 1 and A 2A) in Cuprizone-Induced Hippocampal Demyelination. Mol Neurobiol 2023; 60:5878-5890. [PMID: 37358743 DOI: 10.1007/s12035-023-03440-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 06/10/2023] [Indexed: 06/27/2023]
Abstract
Hippocampal demyelination in multiple sclerosis (MS) has been linked with cognitive deficits, however, patients could benefit from treatment that induces oligodendroglial cell function and promotes remyelination. We investigated the role of A1 and A2A adenosine receptors (AR) in regulating oligodendrocyte precursor cells (OPCs) and myelinating oligodendrocyte (OL) in the demyelinated hippocampus using the cuprizone model of MS. Spatial learning and memory were assessed in wild type C57BL/6 mice (WT) or C57BL/6 mice with global deletion of A1 (A1AR-/-) or A2A AR (A2AAR-/-) fed standard or cuprizone diet (CD) for four weeks. Histology, immunofluorescence, Western blot and TUNEL assays were performed to evaluate the extent of demyelination and apoptosis in the hippocampus. Deletion of A1 and A2A AR alters spatial learning and memory. In A1AR-/- mice, cuprizone feeding led to severe hippocampal demyelination, A2AAR-/- mice had a significant increase in myelin whereas WT mice had intermediate demyelination. The A1AR-/- CD-fed mice displayed significant astrocytosis and decreased expression of NeuN and MBP, whereas these proteins were increased in the A2AAR-/- CD mice. Furthermore, Olig2 was upregulated in A1AR-/- CD-fed mice compared to WT mice fed the standard diet. TUNEL staining of brain sections revealed a fivefold increase in the hippocampus of A1AR-/- CD-fed mice. Also, WT mice fed CD showed a significant decrease expression of A1 AR. A1 and A2A AR are involved in OPC/OL functions with opposing roles in myelin regulation in the hippocampus. Thus, the neuropathological findings seen in MS may be connected to the depletion of A1 AR.
Collapse
Affiliation(s)
- Olamide E Adebiyi
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
- Department of Veterinary Physiology and Biochemistry, Faculty of Veterinary Medicine, University of Ibadan, Ibadan, Nigeria.
| | - Margaret S Bynoe
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY, USA.
| |
Collapse
|
46
|
Rowhanirad S, Taherianfard M. The neuroprotective effects of Chalcones from Ashitaba on cuprizone-induced demyelination via modulation of brain-derived neurotrophic factor and tumor necrosis factor α. Brain Behav 2023; 13:e3144. [PMID: 37403256 PMCID: PMC10498084 DOI: 10.1002/brb3.3144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 06/04/2023] [Accepted: 06/21/2023] [Indexed: 07/06/2023] Open
Abstract
INTRODUCTION Multiple sclerosis (MS) is the most common demyelinating disease of the central nervous system. However, the limitations of available therapeutic strategies are frustrating, both in terms of their low efficacy and multiple side effects. Previous studies showed that natural compounds such as Chalcones possess neuroprotective effects on neurodegenerative disorders. However, few studies have so far been published on the potential effects of Chalcones on treating demyelinating disease. The present study was designed to investigate the effects of Chalcones from Ashitaba (ChA) on cuprizone-induced noxious changes in the C57BL6 mice model of MS. METHODS The mice received normal diets (Control group: CNT), or Cuprizone-supplemented diets either without ChA (Cuprizone group: CPZ) or with low or high (300, 600 mg/kg/day) doses of ChA (ChA-treated groups: CPZ+ChA300/600). Brain-derived neurotrophic factor (BDNF) and tumor necrosis factor alpha (TNFα) levels, demyelination scores in the corpus callosum (CC), and cognitive impairment were evaluated using the enzyme-linked immunosorbent assay, histological, and Y-maze tests, respectively. RESULTS The findings showed that ChA Co-treatment significantly reduced the extent of demyelination in the CC and the serum and brain levels of TNFα in the ChA-treated groups compared to the CPZ group. Besides, treatment with a higher dose of ChA significantly improved the behavioral responses and BDNF levels in the serum and brain of the CPZ+ChA600 group when compared with the CPZ group. CONCLUSION The present study provided evidence for the neuroprotective effects of ChA on cuprizone-induced demyelination and behavioral dysfunction in C57BL/6 mice, possibly by modulating TNFα secretion and BDNF expression.
Collapse
Affiliation(s)
- Soodeh Rowhanirad
- Division of Physiology, Department of Basic Science, School of Veterinary MedicineShiraz UniversityShirazIran
| | - Mahnaz Taherianfard
- Division of Physiology, Department of Basic Science, School of Veterinary MedicineShiraz UniversityShirazIran
| |
Collapse
|
47
|
Lee MJ, Choi JH, Kwon TW, Jo HS, Ha Y, Nah SY, Cho IH. Korean Red Ginseng extract ameliorates demyelination by inhibiting infiltration and activation of immune cells in cuprizone-administrated mice. J Ginseng Res 2023; 47:672-680. [PMID: 37720568 PMCID: PMC10499591 DOI: 10.1016/j.jgr.2023.05.001] [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: 08/08/2022] [Revised: 04/16/2023] [Accepted: 05/09/2023] [Indexed: 09/19/2023] Open
Abstract
Background Korean Red Ginseng (KRG), the steamed root of Panax ginseng, has pharmacological activities for immunological and neurodegenerative disorders. But, the role of KRGE in multiple sclerosis (MS) remains unclear. Purpose To determine whether KRG extract (KRGE) could inhibit demyelination in corpus callosum (CC) of cuprizone (CPZ)-induced murine model of MS. Methods Male adult mice were fed with a standard chow diet or a chow diet supplemented with 0.2% (w/w) CPZ ad libitum for six weeks to induce demyelination while were simultaneously administered with distilled water (DW) alone or KRGE-DW (0.004%, 0.02 and 0.1% of KRGE) by drinking. Results Administration with KRGE-DW alleviated demyelination and oligodendrocyte degeneration associated with inhibition of infiltration and activation of resident microglia and monocyte-derived macrophages as well as downregulation of proinflammatory mediators in the CC of CPZ-fed mice. KRGE-DW also attenuated the level of infiltration of Th1 and Th17) cells, in line with inhibited mRNA expression of IFN-γ and IL-17, respectively, in the CC. These positive effects of KRGE-DW mitigated behavioral dysfunction based on elevated plus maze and the rotarod tests. Conclusion The results strongly suggest that KRGE-DW may inhibit CPZ-induced demyelination due to its oligodendroglial protective and anti-inflammatory activities by inhibiting infiltration/activation of immune cells. Thus, KRGE might have potential in therapeutic intervention for MS.
Collapse
Affiliation(s)
- Min Jung Lee
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Vivian L. Smith Department of Neurosurgery, McGovern Medical School, The University of Texas Health Science Center at Houston, USA
| | - Jong Hee Choi
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Tae Woo Kwon
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Hyo-Sung Jo
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Yujeong Ha
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory and Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, Seoul, Republic of Korea
| | - Ik-Hyun Cho
- Department of Convergence Medical Science, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
48
|
Dedoni S, Scherma M, Camoglio C, Siddi C, Dazzi L, Puliga R, Frau J, Cocco E, Fadda P. An overall view of the most common experimental models for multiple sclerosis. Neurobiol Dis 2023:106230. [PMID: 37453561 DOI: 10.1016/j.nbd.2023.106230] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 07/01/2023] [Accepted: 07/12/2023] [Indexed: 07/18/2023] Open
Abstract
Multiple sclerosis (MS) is a complex chronic disease with an unknown etiology. It is considered an inflammatory demyelinating and neurodegenerative disorder of the central nervous system (CNS) characterized, in most cases, by an unpredictable onset of relapse and remission phases. The disease generally starts in subjects under 40; it has a higher incidence in women and is described as a multifactorial disorder due to the interaction between genetic and environmental risk factors. Unfortunately, there is currently no definitive cure for MS. Still, therapies can modify the disease's natural history, reducing the relapse rate and slowing the progression of the disease or managing symptoms. The limited access to human CNS tissue slows down. It limits the progression of research on MS. This limit has been partially overcome over the years by developing various experimental models to study this disease. Animal models of autoimmune demyelination, such as experimental autoimmune encephalomyelitis (EAE) and viral and toxin or transgenic MS models, represent the most significant part of MS research approaches. These models have now been complemented by ex vivo studies, using organotypic brain slice cultures and in vitro, through induced Pluripotent Stem cells (iPSCs). We will discuss which clinical features of the disorders might be reproduced and investigated in vivo, ex vivo, and in vitro in models commonly used in MS research to understand the processes behind the neuropathological events occurring in the CNS of MS patients. The primary purpose of this review is to give the reader a global view of the main paradigms used in MS research, spacing from the classical animal models to transgenic mice and 2D and 3D cultures.
Collapse
Affiliation(s)
- S Dedoni
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - M Scherma
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Camoglio
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy.
| | - C Siddi
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy
| | - L Dazzi
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - R Puliga
- Department of Life and Environmental Sciences, Section of Neuroscience and Anthropology, University of Cagliari, Monserrato (Cagliari), Italy.
| | - J Frau
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy
| | - E Cocco
- Regional Multiple Sclerosis Center, ASSL Cagliari, ATS Sardegna, Italy; Department Medical Science and Public Health, University of Cagliari, Italy.
| | - P Fadda
- Department of Biomedical Sciences, Division of Neuroscience and Clinical Pharmacology, University of Cagliari, Italy; Neuroscience Institute, Section of Cagliari, National Research Council of Italy (CNR), Cagliari, Italy.
| |
Collapse
|
49
|
Gharagozloo M, Galleguillos D, Jank L, Sotirchos ES, Smith MD, Garton T, Kumar S, Hussein O, Potluri S, Taylor M, Siu C, Mace JW, Dawson T, Dawson VL, Lee S, Calabresi PA. The Effects of NLY01, a Novel Glucagon-Like Peptide-1 Receptor Agonist, on Cuprizone-Induced Demyelination and Remyelination: Challenges and Future Perspectives. Neurotherapeutics 2023; 20:1229-1240. [PMID: 37296356 PMCID: PMC10457267 DOI: 10.1007/s13311-023-01390-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/13/2023] [Indexed: 06/12/2023] Open
Abstract
Recent evidence suggests that the glucagon-like peptide-1 receptor (GLP-1R) agonists have neuroprotective activities in the CNS in animal models of Parkinson's disease, Alzheimer's disease, and multiple sclerosis (MS). This study aimed to investigate whether a novel long-acting GLP-1R agonist, NLY01, could limit demyelination or improve remyelination as occurs in MS using the cuprizone (CPZ) mouse model. Herein, we assessed the expression of GLP-1R on oligodendrocytes in vitro and found that mature oligodendrocytes (Olig2+PDGFRa-) express GLP-1R. We further confirmed this observation in the brain by immunohistochemistry and found that Olig2+CC1+ cells express GLP-1R. We next administered NLY01 twice per week to C57B6 mice while on CPZ chow diet and found that NLY01 significantly reduced demyelination with greater weight loss than vehicle-treated controls. Because GLP-1R agonists are known to have anorexigenic effect, we then administered CPZ by oral gavage and treated the mice with NLY01 or vehicle to ensure the dose consistency of CPZ ingestion among mice. Using this modified approach, NLY01 was no longer effective in reducing demyelination of the corpus callosum (CC). We next sought to examine the effects of NLY01 treatment on remyelination after CPZ intoxication and during the recovery period using an adoptive transfer-CPZ (AT-CPZ) model. We found no significant differences between the NLY01 and vehicle groups in the amount of myelin or the number of mature oligodendrocytes in the CC. In summary, despite the promising anti-inflammatory and neuroprotective effects of GLP-1R agonists that have been previously described, our experiments provided no evidence to support a beneficial effect of NLY01 on limiting demyelination or enhancing remyelination. This information may be useful in selecting proper outcome measures in clinical trials of this promising class of drugs in MS.
Collapse
Affiliation(s)
- Marjan Gharagozloo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, Pathology Building 509, 600 N. Wolfe St, Baltimore, MD, 21287, USA.
| | - Danny Galleguillos
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Larissa Jank
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Elias S Sotirchos
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Thomas Garton
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Swati Kumar
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Omar Hussein
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Saahith Potluri
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Michelle Taylor
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Catherine Siu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jackson W Mace
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ted Dawson
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Valina L Dawson
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Neuraly Inc, Gaithersburg, MD, USA
| | | | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, 21205, USA.
- Division of Neuroimmunology and Neurological Infections, Johns Hopkins Hospital, Pathology Building 509, 600 N. Wolfe St, Baltimore, MD, 21287, USA.
| |
Collapse
|
50
|
Lindsay SL, McCanney GA, Zhan J, Scheld M, Smith RS, Goodyear CS, Yates EA, Kipp M, Turnbull JE, Barnett SC. Low sulfated heparan sulfate mimetic differentially affects repair in immune-mediated and toxin-induced experimental models of demyelination. Glia 2023; 71:1683-1698. [PMID: 36945189 PMCID: PMC10952530 DOI: 10.1002/glia.24363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/23/2023]
Abstract
There is an urgent need for therapies that target the multicellular pathology of central nervous system (CNS) disease. Modified, nonanticoagulant heparins mimic the heparan sulfate glycan family and are known regulators of multiple cellular processes. In vitro studies have demonstrated that low sulfated modified heparin mimetics (LS-mHeps) drive repair after CNS demyelination. Herein, we test LS-mHep7 (an in vitro lead compound) in experimental autoimmune encephalomyelitis (EAE) and cuprizone-induced demyelination. In EAE, LS-mHep7 treatment resulted in faster recovery and rapidly reduced inflammation which was accompanied by restoration of animal weight. LS-mHep7 treatment had no effect on remyelination or on OLIG2 positive oligodendrocyte numbers within the corpus callosum in the cuprizone model. Further in vitro investigation confirmed that LS-mHep7 likely mediates its pro-repair effect in the EAE model by sequestering inflammatory cytokines, such as CCL5 which are upregulated during immune-mediated inflammatory attacks. These data support the future clinical translation of this next generation modified heparin as a treatment for CNS diseases with active immune system involvement.
Collapse
Affiliation(s)
- Susan L. Lindsay
- School of Infection and ImmunityUniversity of Glasgow120 University PlaceGlasgowG12 8TAUK
| | - George A. McCanney
- School of Infection and ImmunityUniversity of Glasgow120 University PlaceGlasgowG12 8TAUK
| | - Jiangshan Zhan
- Institute of AnatomyUniversity of RostockGertrudenstrasse 918057RostockGermany
| | - Miriam Scheld
- Institute of Neuroanatomy, Faculty of MedicineRWTH Aachen University52074AachenGermany
| | - Rebecca Sherrard Smith
- School of Infection and ImmunityUniversity of Glasgow120 University PlaceGlasgowG12 8TAUK
| | - Carl S. Goodyear
- School of Infection and ImmunityUniversity of Glasgow120 University PlaceGlasgowG12 8TAUK
| | - Edwin A. Yates
- Institute of Systems, Molecules and Integrative BiologyUniversity of LiverpoolLiverpoolL69 7ZBUK
| | - Markus Kipp
- Institute of AnatomyUniversity of RostockGertrudenstrasse 918057RostockGermany
| | - Jeremy E. Turnbull
- Institute of Systems, Molecules and Integrative BiologyUniversity of LiverpoolLiverpoolL69 7ZBUK
- Centre for GlycosciencesKeele UniversityKeeleST5 5BGUK
| | - Susan C. Barnett
- School of Infection and ImmunityUniversity of Glasgow120 University PlaceGlasgowG12 8TAUK
| |
Collapse
|