1
|
He Y, Xu Z, He Y, Liu J, Li J, Wang S, Xiao L. Preventing production of new oligodendrocytes impairs remyelination and sustains behavioural deficits after demyelination. Biochem Biophys Res Commun 2024; 733:150592. [PMID: 39213705 DOI: 10.1016/j.bbrc.2024.150592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024]
Abstract
Damage to oligodendrocytes (OLs) and myelin sheaths (demyelination) has been shown to be associated with numerous neurological and psychiatric disorders. Remyelination is a rare and reliable regenerative response that occurs in the central nervous system (CNS). It is generally believed that OL progenitor cells (OPCs) are the cell source to generate new OLs to remyelinate the demyelinated axons. However, several recent studies have argued that pre-existing mature OLs that survive within the demyelinated area are responsible for remyelination. Here, by conditional knock-out (KO) of a transcription factor gene that is essential for OPC differentiation, namely myelin regulatory factor (Myrf), to block the production of adult new OLs and examined its effect on remyelination after cuprizone (CPZ)-induced demyelination. We found that OPCs specific Myrf cKO mice show dramatic impairment in remyelination after 4 weeks of recovery from 5 weeks of CPZ diet and they leave over significant behavioral deficits such as anxiety-like behavior, decreased motor skills, and impaired memory compared to control mice that have recovered for the same time. Our data support the idea that OPCs are the major cell sources for myelin regeneration, suggesting that targeting the activation of OPCs and promoting their differentiation to boost new OLs production is critical for therapeutic intervention for demyelinating diseases such as multiple sclerosis (MS).
Collapse
Affiliation(s)
- Yuehua He
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Zhengtao Xu
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Yongxiang He
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Junhong Liu
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Jiong Li
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Shuming Wang
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China
| | - Lin Xiao
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou, 510631, China.
| |
Collapse
|
2
|
Rizoli C, Dos Santos NM, Maróstica Júnior MR, da Cruz-Höfling MA, Mendonça MCP, de Jesus MB. The therapeutic potential of reduced graphene oxide in attenuating cuprizone-induced demyelination in mice. NANOTECHNOLOGY 2024; 36:025102. [PMID: 39389086 DOI: 10.1088/1361-6528/ad857e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 10/10/2024] [Indexed: 10/12/2024]
Abstract
Reduced graphene oxide (rGO) has unique physicochemical properties that make it suitable for therapeutic applications in neurodegenerative scenarios. This study investigates the therapeutic potential of rGO in a cuprizone-induced demyelination model in mice through histomorphological techniques and analysis of biochemical parameters. We demonstrate that daily intraperitoneal administration of rGO (1 mg ml-1) for 21 days tends to reduce demyelination in theCorpus callosumby decreasing glial cell recruitment during the repair mechanism. Additionally, rGO interferes with oxidative stress markers in the brain and liver indicating potential neuroprotective effects in the central nervous system. No significant damage to vital organs was observed, suggesting that multiple doses could be used safely. However, further long-term investigations are needed to understand rGO distribution, metabolism, routes of action and associated challenges in central neurodegenerative therapies. Overall, these findings contribute to the comprehension of rGO effectsin vivo, paving the way for possible future clinical research.
Collapse
Affiliation(s)
- Cintia Rizoli
- Departmento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| | | | | | - Maria Alice da Cruz-Höfling
- Department of Structural and Functional Biology, Institute of Biology, University of Campinas, Campinas, SP, Brazil
| | | | - Marcelo Bispo de Jesus
- Departmento de Bioquímica e Biologia Tecidual, Instituto de Biologia, Universidade Estadual de Campinas (UNICAMP), Campinas, SP, Brazil
| |
Collapse
|
3
|
Friesen E, Sheft M, Hari K, Palmer V, Zhu S, Herrera S, Buist R, Jiang D, Li XM, Del Bigio MR, Thiessen JD, Martin M. Quantitative Analysis of Early White Matter Damage in Cuprizone Mouse Model of Demyelination Using 7.0 T MRI Multiparametric Approach. ASN Neuro 2024; 16:2404366. [PMID: 39400556 DOI: 10.1080/17590914.2024.2404366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
Magnetic Resonance Imaging (MRI) is commonly used to follow the progression of neurodegenerative conditions, including multiple sclerosis (MS). MRI is limited by a lack of correlation between imaging results and clinical presentations, referred to as the clinico-radiological paradox. Animal models are commonly used to mimic the progression of human neurodegeneration and as a tool to help resolve the paradox. Most studies focus on later stages of white matter (WM) damage whereas few focus on early stages when oligodendrocyte apoptosis has just begun. The current project focused on these time points, namely weeks 2 and 3 of cuprizone (CPZ) administration, a toxin which induces pathophysiology similar to MS. In vivo T2-weighted (T2W) and Magnetization Transfer Ratio (MTR) maps and ex vivo Diffusion Tensor Imaging (DTI), Magnetization Transfer Imaging (MTI), and relaxometry (T1 and T2) values were obtained at 7 T. Significant changes in T2W signal intensity and non-significant changes in MTR were observed to correspond to early WM damage, whereas significant changes in both corresponded with full demyelination. Some DTI metrics decrease with simultaneous increase in others, indicating acute demyelination. MTI metrics T2A, T2B, f and R were observed to have contradictory changes across CPZ administration. T1 relaxation times were observed to have stronger correlations to disease states during later stages of CPZ treatment, whereas T2 had weak correlations to early WM damage. These results all suggest the need for multiple metrics and further studies at early and late time points of demyelination. Further research is required to continue investigating the interplay between various MR metrics during all weeks of CPZ administration.
Collapse
Affiliation(s)
- Emma Friesen
- Department of Chemistry, University of Winnipeg, Winnipeg, Canada
| | - Maxina Sheft
- Department of Physics, University of Winnipeg, Winnipeg, Canada
- Massachusetts Institute of Technology, Cambridge, USA
| | - Kamya Hari
- Department of Physics, University of Winnipeg, Winnipeg, Canada
- Electronics and Communication Engineering, SSN College of Engineering, Chennai, India
| | - Vanessa Palmer
- Department of Biomedical Engineering, University of Manitoba, Winnipeg, Canada
- Cubresa Inc, Winnipeg, Canada
| | - Shenghua Zhu
- Department of Neuroradiology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sheryl Herrera
- Department of Physics, University of Winnipeg, Winnipeg, Canada
- Cubresa Inc, Winnipeg, Canada
| | - Richard Buist
- Department of Radiology, University of Manitoba, Winnipeg, Canada
| | - Depeng Jiang
- Department of Community Health Sciences, University of Manitoba, Winnipeg, Canada
| | - Xin-Min Li
- Department of Psychiatry, University of Alberta, Edmonton, Canada
| | - Marc R Del Bigio
- Department of Pathology, University of Manitoba, Winnipeg, Canada
| | - Jonathan D Thiessen
- Imaging Program, Lawson Health Research Institute, London, Canada
- Department of Medical Biophysics, Western University, London, Canada
| | - Melanie Martin
- Department of Physics, University of Winnipeg, Winnipeg, Canada
| |
Collapse
|
4
|
Altunay ZM, Biswas J, Cheung HW, Pijewski RS, Papile LE, Akinlaja YO, Tang A, Kresic LC, Schouw AD, Ugrak MV, Caro K, Peña Palomino PA, Ressl S, Nishiyama A, Crocker SJ, Martinelli DC. C1ql1 expression in oligodendrocyte progenitor cells promotes oligodendrocyte differentiation. FEBS J 2024. [PMID: 39257292 DOI: 10.1111/febs.17256] [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: 03/14/2024] [Revised: 07/16/2024] [Accepted: 08/14/2024] [Indexed: 09/12/2024]
Abstract
Myelinating oligodendrocytes arise from the stepwise differentiation of oligodendrocyte progenitor cells (OPCs). Approximately 5% of all adult brain cells are OPCs. Why would a mature brain need such a large number of OPCs? New myelination is possibly required for higher-order functions such as cognition and learning. Additionally, this pool of OPCs represents a source of new oligodendrocytes to replace those lost during injury, inflammation, or in diseases such as multiple sclerosis (MS). How OPCs are instructed to differentiate into oligodendrocytes is poorly understood, and for reasons presently unclear, resident pools of OPCs are progressively less utilized in MS. The complement component 1, q subcomponent-like (C1QL) protein family has been studied for their functions at neuron-neuron synapses, but we show that OPCs express C1ql1. We created OPC-specific conditional knockout mice and show that C1QL1 deficiency reduces the differentiation of OPCs into oligodendrocytes and reduces myelin production during both development and recovery from cuprizone-induced demyelination. In vivo over-expression of C1QL1 causes the opposite phenotype: increased oligodendrocyte density and myelination during recovery from demyelination. We further used primary cultured OPCs to show that C1QL1 levels can bidirectionally regulate the extent of OPC differentiation in vitro. Our results suggest that C1QL1 may initiate a previously unrecognized signaling pathway to promote differentiation of OPCs into oligodendrocytes. This study has relevance for possible novel therapies for demyelinating diseases and may illuminate a previously undescribed mechanism to regulate the function of myelination in cognition and learning.
Collapse
Affiliation(s)
- Zeynep M Altunay
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Joyshree Biswas
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Hiu W Cheung
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Robert S Pijewski
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
- Department of Biology, Anna Maria College, Paxton, MA, USA
| | - Lucille E Papile
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Yetunde O Akinlaja
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
| | - Andrew Tang
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Lyndsay C Kresic
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Alexander D Schouw
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Maksym V Ugrak
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Keaven Caro
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | | | - Susanne Ressl
- Department of Neuroscience, The University of Texas at Austin, TX, USA
| | - Akiko Nishiyama
- Department of Physiology and Neurobiology, University of Connecticut, Storrs, CT, USA
- The Connecticut Institute for the Brain and Cognitive Sciences (IBACS), Storrs, CT, USA
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
- The Connecticut Institute for the Brain and Cognitive Sciences (IBACS), Storrs, CT, USA
| | - David C Martinelli
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
- The Connecticut Institute for the Brain and Cognitive Sciences (IBACS), Storrs, CT, USA
| |
Collapse
|
5
|
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
|
6
|
Rodrigues-Amorim D, Bozzelli PL, Kim T, Liu L, Gibson O, Yang CY, Murdock MH, Galiana-Melendez F, Schatz B, Davison A, Islam MR, Shin Park D, Raju RM, Abdurrob F, Nelson AJ, Min Ren J, Yang V, Stokes MP, Tsai LH. Multisensory gamma stimulation mitigates the effects of demyelination induced by cuprizone in male mice. Nat Commun 2024; 15:6744. [PMID: 39112447 PMCID: PMC11306744 DOI: 10.1038/s41467-024-51003-7] [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: 09/02/2023] [Accepted: 07/08/2024] [Indexed: 08/10/2024] Open
Abstract
Demyelination is a common pathological feature in a wide range of diseases, characterized by the loss of myelin sheath and myelin-supporting oligodendrocytes. These losses lead to impaired axonal function, increased vulnerability of axons to damage, and result in significant brain atrophy and neuro-axonal degeneration. Multiple pathomolecular processes contribute to neuroinflammation, oligodendrocyte cell death, and progressive neuronal dysfunction. In this study, we use the cuprizone mouse model of demyelination to investigate long-term non-invasive gamma entrainment using sensory stimulation as a potential therapeutic intervention for promoting myelination and reducing neuroinflammation in male mice. Here, we show that multisensory gamma stimulation mitigates demyelination, promotes oligodendrogenesis, preserves functional integrity and synaptic plasticity, attenuates oligodendrocyte ferroptosis-induced cell death, and reduces brain inflammation. Thus, the protective effects of multisensory gamma stimulation on myelin and anti-neuroinflammatory properties support its potential as a therapeutic approach for demyelinating disorders.
Collapse
Grants
- R01 AG069232 NIA NIH HHS
- R01 AT011460 NCCIH NIH HHS
- R01 NS122742 NINDS NIH HHS
- R56 AG069232 NIA NIH HHS
- We would like to acknowledge the following individuals and organizations for their support: Fundacion Bancaria la Caixa, The JPB Foundation, Carol and Gene Ludwig Family Foundation, Lester A. Gimpelson, Eduardo Eurnekian, The Dolby Family, Kathy and Miguel Octavio, the Marc Haas Foundation, Ben Lenail and Laurie Yoler, and NIH RO1 grants AG069232, AT011460 and R01NS122742 to L.-H.T.
Collapse
Affiliation(s)
- Daniela Rodrigues-Amorim
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - P Lorenzo Bozzelli
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - TaeHyun Kim
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Liwang Liu
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Cheng-Yi Yang
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Mitchell H Murdock
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Fabiola Galiana-Melendez
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Brooke Schatz
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Alexis Davison
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Md Rezaul Islam
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Dong Shin Park
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ravikiran M Raju
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Newborn Medicine, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Fatema Abdurrob
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Jian Min Ren
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA, USA
| | - Vicky Yang
- Cell Signaling Technology, 3 Trask Lane, Danvers, MA, USA
| | | | - Li-Huei Tsai
- Department of Brain and Cognitive Sciences, Picower Institute for Learning and Memory, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
7
|
Pouzol L, Sassi A, Tunis M, Zurbach A, Baumlin N, Gnerre C, Strasser DS, Marrie J, Vezzali E, Martinic MM. ACKR3 Antagonism Enhances the Repair of Demyelinated Lesions Through Both Immunomodulatory and Remyelinating Effects. Neurochem Res 2024; 49:2087-2104. [PMID: 38819698 PMCID: PMC11233362 DOI: 10.1007/s11064-024-04173-1] [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: 01/27/2024] [Revised: 04/16/2024] [Accepted: 05/22/2024] [Indexed: 06/01/2024]
Abstract
Addressing inflammation, demyelination, and associated neurodegeneration in inflammatory demyelinating diseases like multiple sclerosis (MS) remains challenging. ACT-1004-1239, a first-in-class and potent ACKR3 antagonist, currently undergoing clinical development, showed promise in preclinical MS models, reducing neuroinflammation and demyelination. However, its effectiveness in treating established disease and impact on remyelination after the occurrence of demyelinated lesions remain unexplored. This study assessed the therapeutic effect of ACT-1004-1239 in two demyelinating disease models. In the proteolipid protein (PLP)-induced experimental autoimmune encephalomyelitis (EAE) model, ACT-1004-1239 administered upon the detection of the first signs of paralysis, resulted in a dose-dependent reduction in EAE disease severity, concomitant with diminished immune cell infiltrates in the CNS and reduced demyelination. Notably, efficacy correlated with elevated plasma concentrations of CXCL11 and CXCL12, two pharmacodynamic biomarkers of ACKR3 antagonism. Combining ACT-1004-1239 with siponimod, an approved immunomodulatory treatment for MS, synergistically reduced EAE severity. In the cuprizone-induced demyelination model, ACT-1004-1239 administered after 5 weeks of cuprizone exposure, significantly accelerated remyelination, already quantifiable one week after cuprizone withdrawal. Additionally, ACT-1004-1239 penetrated the CNS, elevating brain CXCL12 concentrations. These results demonstrate that ACKR3 antagonism significantly reduces the severity of experimental demyelinating diseases, even when treatment is initiated therapeutically, after the occurrence of lesions. It confirms the dual mode of action of ACT-1004-1239, exhibiting both immunomodulatory effects by reducing neuroinflammation and promyelinating effects by accelerating myelin repair. The results further strengthen the rationale for evaluating ACT-1004-1239 in clinical trials for patients with demyelinating diseases.
Collapse
Affiliation(s)
- Laetitia Pouzol
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland.
| | - Anna Sassi
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Mélanie Tunis
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Anaïs Zurbach
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Nadège Baumlin
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Carmela Gnerre
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Daniel S Strasser
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Julia Marrie
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Enrico Vezzali
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| | - Marianne M Martinic
- Idorsia Pharmaceuticals Ltd, Hegenheimermattweg 91, Allschwil 4123, Basel-Landschaft, Switzerland
| |
Collapse
|
8
|
Amoah R, Danquah J, Mante PK. Antiaris africana aqueous extract inhibits chronic demyelination and seizures in mice. Heliyon 2024; 10:e34296. [PMID: 39100489 PMCID: PMC11296038 DOI: 10.1016/j.heliyon.2024.e34296] [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: 04/03/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
Demyelinating diseases are commonly associated with epileptic seizures and have limited management options. Hence, the need to investigate potential options for management of such seizures. Antiaris Africana extract (AE) was investigated for effect in chronic demyelinating seizures. Cuprizone treatment induced short but frequent spike discharges in mice. Antiaris Africana extract (300 mg/kg) treatment abolished epileptiform discharges. Cuprizone administration caused severe demyelination in the corpus callosum. After the demyelination phase, myelin content decreased to 22.86 ± 1.92 % in the cuprizone-only group. However, there was an increase to 52.14 ± 3.91 % in cuprizone-only group and 62.00 ± 2.78 % in the Antiaris africana extract group respectively, after a 4-week cuprizone cessation period. Treatment with AE and LEV visibly altered myelin growth. Antiaris africana extract treatment produced significant (P < 0.001, F (3, 16) = 698.4) increase in locomotor activity similar to LEV (P < 0.001,F (2, 12) = 678.7) and DZP (P < 0.001, F (2, 12) = 620.4) and improved beam traversal time (18.71 ± 2.244 s; 95 % CI: 13.22-24.20) while causing significantly (P < 0.05, F (2, 15) = 6.667) fewer stepping errors. Antiaris africana extract inhibits seizures induced by chronic demyelination and has beneficial effects on motor coordination.
Collapse
Affiliation(s)
- Ransford Amoah
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - John Danquah
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Priscilla Kolibea Mante
- Department of Pharmacology, Faculty of Pharmacy and Pharmaceutical Sciences, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| |
Collapse
|
9
|
He Y, Xie H, Xu Z, Zhang L, Feng Y, Long Y, Wang S, He Y, Li J, Zou Y, Zheng W, Xiao L. Rapid and prolonged response of oligodendrocyte lineage cells in standard acute cuprizone demyelination model revealed by in situ hybridization. Neurosci Lett 2024; 836:137869. [PMID: 38852766 DOI: 10.1016/j.neulet.2024.137869] [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: 03/15/2024] [Revised: 05/29/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Dietary administration of a copper chelator, cuprizone (CPZ), has long been reported to induce intense and reproducible demyelination of several brain structures such as the corpus callosum. Despite the widespread use of CPZ as an animal model for demyelinating diseases such as multiple sclerosis (MS), the mechanism by which it induces demyelination and then allows robust remyelination is still unclear. An intensive mapping of the cell dynamics of oligodendrocyte (OL) lineage during the de- and remyelination course would be particularly important for a deeper understanding of this model. Here, using a panel of OL lineage cell markers as in situ hybridization (ISH) probes, including Pdgfra, Plp, Mbp, Mog, Enpp6, combined with immunofluorescence staining of CC1, SOX10, we provide a detailed dynamic profile of OL lineage cells during the entire course of the model from 1, 2, 3.5 days, 1, 2, 3, 4,5 weeks of CPZ treatment, as well as after 1, 2, 3, 4 weeks of recovery from CPZ treatment. The result showed an unexpected early death of mature OLs and response of OL progenitor cells (OPCs) in vivo upon CPZ challenge, and a prolonged upregulation of myelin-forming OLs compared to the intact control even 4 weeks after CPZ withdrawal. These data may serve as a basic reference system for future studies of the effects of any intervention on de- and remyelination using the CPZ model, and imply the need to optimize the timing windows for the introduction of pro-remyelination therapies in demyelinating diseases such as MS.
Collapse
Affiliation(s)
- Yuehua He
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Hua Xie
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - ZhengTao Xu
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Liuning Zhang
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Yuanyu Feng
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Yu Long
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Shuming Wang
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Yongxiang He
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Jiong Li
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Yanping Zou
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Wei Zheng
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China
| | - Lin Xiao
- Key Laboratory of Brain, Cognition and Education Sciences of Ministry of Education, Institute for Brain Research and Rehabilitation, Guangdong Key Laboratory of Mental Health and Cognitive Science, and Center for Studies of Psychological Application, South China Normal University, Guangzhou 510631, China.
| |
Collapse
|
10
|
Barbaresi P, Fabri M, Lorenzi T, Sagrati A, Morroni M. Intrinsic organization of the corpus callosum. Front Physiol 2024; 15:1393000. [PMID: 39035452 PMCID: PMC11259024 DOI: 10.3389/fphys.2024.1393000] [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: 02/28/2024] [Accepted: 05/16/2024] [Indexed: 07/23/2024] Open
Abstract
The corpus callosum-the largest commissural fiber system connecting the two cerebral hemispheres-is considered essential for bilateral sensory integration and higher cognitive functions. Most studies exploring the corpus callosum have examined either the anatomical, physiological, and neurochemical organization of callosal projections or the functional and/or behavioral aspects of the callosal connections after complete/partial callosotomy or callosal lesion. There are no works that address the intrinsic organization of the corpus callosum. We review the existing information on the activities that take place in the commissure in three sections: I) the topographical and neurochemical organization of the intracallosal fibers, II) the role of glia in the corpus callosum, and III) the role of the intracallosal neurons.
Collapse
Affiliation(s)
- Paolo Barbaresi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Marche Polytechnic University, Ancona, Italy
| | - Mara Fabri
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Teresa Lorenzi
- Department of Experimental and Clinical Medicine, Section of Neuroscience and Cell Biology, Marche Polytechnic University, Ancona, Italy
| | - Andrea Sagrati
- Department of Life and Environmental Sciences, Marche Polytechnic University, Ancona, Italy
| | - Manrico Morroni
- Electron Microscopy Unit, Azienda Ospedaliero-Universitaria, Ancona, Italy
| |
Collapse
|
11
|
Mathieu PA, Sampertegui YR, Elias F, Silva AS, de Luján Calcagno M, López R, Adamo AM. Oligodeoxynucleotide IMT504: Effects on Central Nervous System Repair Following Demyelination. Mol Neurobiol 2024; 61:4146-4165. [PMID: 38064102 DOI: 10.1007/s12035-023-03825-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 11/21/2023] [Indexed: 07/11/2024]
Abstract
Multiple sclerosis (MS) is an immune-mediated central nervous system (CNS) disease characterized by demyelination resulting from oligodendrocyte loss and inflammation. Cuprizone (CPZ) administration experimentally replicates MS pattern-III lesions, generating an inflammatory response through microgliosis and astrogliosis. Potentially remyelinating agents include oligodeoxynucleotides (ODN) with a specific immunomodulatory sequence consisting of the active motif PyNTTTTGT. In this work, the remyelinating effects of ODN IMT504 were evaluated through immunohistochemistry and qPCR analyses in a rat CPZ-induced demyelination model. Subcutaneous IMT504 administration exacerbated the pro-inflammatory response to demyelination and accelerated the transition to an anti-inflammatory state. IMT504 reduced microgliosis in general and the number of phagocytic microglia in particular and expanded the population of oligodendroglial progenitor cells (OPCs), later reflected in an increase in mature oligodendrocytes. The intracranial injection of IMT504 and intravenous inoculation of IMT504-treated B lymphocytes rendered comparable results. Altogether, these findings unveil potentially beneficial properties of IMT504 in the regulation of neuroinflammation and oligodendrogenesis, which may aid the development of therapies for demyelinating diseases such as MS.
Collapse
Affiliation(s)
- Patricia A Mathieu
- Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Universidad de Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina
- CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Universidad de Buenos Aires Junín 956, C1113AAD, Buenos Aires, Argentina
| | - Yim Rodriguez Sampertegui
- Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Universidad de Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina
- CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Universidad de Buenos Aires Junín 956, C1113AAD, Buenos Aires, Argentina
| | - Fernanda Elias
- Instituto de Ciencia y Tecnología Dr. César Milstein (CONICET-Fundación Pablo Cassará), Saladillo 2468, C1440FFX, Buenos Aires, Argentina
| | - Alexis Silva Silva
- Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Universidad de Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina
- CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Universidad de Buenos Aires Junín 956, C1113AAD, Buenos Aires, Argentina
| | - María de Luján Calcagno
- Facultad de Farmacia y Bioquímica, Departamento de Fisicomatemática, Universidad de Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina
| | | | - Ana M Adamo
- Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Universidad de Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina.
- CONICET, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Universidad de Buenos Aires Junín 956, C1113AAD, Buenos Aires, Argentina.
| |
Collapse
|
12
|
Strohm AO, Majewska AK. Physical exercise regulates microglia in health and disease. Front Neurosci 2024; 18:1420322. [PMID: 38911597 PMCID: PMC11192042 DOI: 10.3389/fnins.2024.1420322] [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: 04/19/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024] Open
Abstract
There is a well-established link between physical activity and brain health. As such, the effectiveness of physical exercise as a therapeutic strategy has been explored in a variety of neurological contexts. To determine the extent to which physical exercise could be most beneficial under different circumstances, studies are needed to uncover the underlying mechanisms behind the benefits of physical activity. Interest has grown in understanding how physical activity can regulate microglia, the resident immune cells of the central nervous system. Microglia are key mediators of neuroinflammatory processes and play a role in maintaining brain homeostasis in healthy and pathological settings. Here, we explore the evidence suggesting that physical activity has the potential to regulate microglia activity in various animal models. We emphasize key areas where future research could contribute to uncovering the therapeutic benefits of engaging in physical exercise.
Collapse
Affiliation(s)
- Alexandra O. Strohm
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, United States
| | - Ania K. Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, NY, United States
- Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY, United States
- Center for Visual Science, University of Rochester Medical Center, Rochester, NY, United States
| |
Collapse
|
13
|
Du C, Zhang T, Feng C, Sun Q, Chen Z, Shen X, Liu Y, Dai G, Zhang X, Tang N. The effects of venlafaxine on depressive-like behaviors and gut microbiome in cuprizone-treated mice. Front Psychiatry 2024; 15:1347867. [PMID: 38899045 PMCID: PMC11186413 DOI: 10.3389/fpsyt.2024.1347867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 05/13/2024] [Indexed: 06/21/2024] Open
Abstract
Background Cuprizone (CPZ)-treated mice show significant demyelination, altered gut microbiome, and depressive-like behaviors. However, the effects of venlafaxine (Ven) on the gut microbiome and depressive-like behavior of CPZ-treated mice are largely unclear. Methods Male C57BL/6J mice were fed a chow containing 0.2% cuprizone (w/w) for 5 weeks to induce a model of demyelination. Meanwhile, the gut microbiota and depressive-like behaviors were assessed after the mice were fed with Ven (20 mg/kg/day) or equal volumes of distilled water for 2 weeks by oral gavage from the third week onward during CPZ treatment. Results CPZ treatment decreased the sucrose preference rate in the sucrose preference test and increased the immobility time in the tail-suspension test, and it also induced an abnormality in β-diversity and changes in microbial composition. Ven alleviated the depressive-like behavior and regulated the composition of the gut microbiota, such as the increase of Lactobacillus and Bifidobacterium in CPZ-treated mice. Conclusion The anti-depressant effects of Ven might be related to the regulation of gut microbiota in the CPZ-treated mice.
Collapse
Affiliation(s)
- Chunhai Du
- Department of Oncology, Hengshui Hospital of Traditional Chinese Medicine, Hengshui, Hebei, China
| | - Tian Zhang
- Department of Psychiatry, Xijing Hospital, Air Force Medical University, Xi’an, Shaanxi, China
| | - Chong Feng
- Department of Psychiatry, The 907th Hospital of the PLA Joint Logistics Support Force, Nanping, Fujian, China
| | - Qian Sun
- Department of Oncology, Hengshui Hospital of Traditional Chinese Medicine, Hengshui, Hebei, China
| | - ZhiGuo Chen
- Department of Psychiatry, The 907th Hospital of the PLA Joint Logistics Support Force, Nanping, Fujian, China
| | - Xin Shen
- Department of Psychiatry, The 907th Hospital of the PLA Joint Logistics Support Force, Nanping, Fujian, China
| | - Ying Liu
- Department of Psychiatry, The 907th Hospital of the PLA Joint Logistics Support Force, Nanping, Fujian, China
| | - Gengwu Dai
- Department of Psychiatry, The 907th Hospital of the PLA Joint Logistics Support Force, Nanping, Fujian, China
| | - Xuan Zhang
- Institute for Hospital Management Research, Chinese PLA General Hospital, Beijing, China
| | - Nailong Tang
- Department of Psychiatry, The 907th Hospital of the PLA Joint Logistics Support Force, Nanping, Fujian, China
| |
Collapse
|
14
|
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
|
15
|
Gharighnia S, Omidi A, Ragerdi Kashani I, Sepand MR, Pour Beiranvand S. Ameliorative effects of acetyl-L-carnitine on corpus callosum and functional recovery in demyelinated mouse model. Int J Neurosci 2024; 134:409-419. [PMID: 35912879 DOI: 10.1080/00207454.2022.2107515] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 10/16/2022]
Abstract
AIM Multiple sclerosis (MS) is the most common chronic inflammatory demyelinating disease of the central nervous system. Oxidative stress via distinct pathobiological pathways plays a pivotal role in the formation and persistence of MS lesions. Acetyl-L-carnitine (ALC) facilitates the uptake of acetyl coenzyme-A into the mitochondria by a fatty acid oxidation process. ALC could be a therapeutic antioxidant in the myelin repair process. This study explored the potential neuroprotective effects of ALC in cuprizone (CPZ) intoxicated mice. MATERIALS AND METHODS Thirty male C57BL/6 mice were divided into three groups. The control animals received a normal diet. The CPZ and CPZ + ALC groups were fed with a 0.2% cuprizone diet for 12 weeks. In the CPZ + ALC group, animals received ALC (300 mg/kg/day) from the 10th -12th weeks. Animals were evaluated functionally by beam walking test (BWT) weekly. Eventually, the corpus callosum (CC) was extracted for histological, biochemical, and molecular studies. RESULTS BWT data showed ALC significantly improves balance and gait in the demyelinating mouse model. Histological staining represented ALC effectively increased remyelination in the CC. Biochemical evaluations demonstrated ALC decreased the malondialdehyde level with a parallel increase in the reduced glutathione and catalase activity levels in the CC. Molecular analysis revealed that ALC significantly increased the expression of oligodendrocyte transcription-2 (Olig-2) and Poly lipoproteins (Plp) genes in the CC. CONCLUSIONS ALC improved balance and motor coordination in the demyelinated mouse model. It may be by reducing the levels of free radicals and increasing the expression of Olig-2 and Plp as myelin-related genes.
Collapse
Affiliation(s)
- Sanaz Gharighnia
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ameneh Omidi
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Iraj Ragerdi Kashani
- Department of Anatomical Sciences, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Sepand
- Toxicology and Poisoning Research Centre, Department of Toxicology and Pharmacology, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahram Pour Beiranvand
- Department of Anatomical Sciences, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Vinnenberg L, Rychlik N, Oniani T, Williams B, White JA, Kovac S, Meuth SG, Budde T, Hundehege P. Assessing neuroprotective effects of diroximel fumarate and siponimod via modulation of pacemaker channels in an experimental model of remyelination. Exp Neurol 2024; 371:114572. [PMID: 37852467 DOI: 10.1016/j.expneurol.2023.114572] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/04/2023] [Accepted: 10/14/2023] [Indexed: 10/20/2023]
Abstract
Cuprizone (CPZ)-induced alterations in axonal myelination are associated with a period of neuronal hyperexcitability and increased activity of hyperpolarization-activated and cyclic nucleotide-gated (HCN) channels in the thalamocortical (TC) system. Substances used for the treatment of multiple sclerosis (MS) have been shown to normalize neuronal excitability in CPZ-treated mice. Therefore, we aimed to examine the effects of diroximel fumarate (DRF) and the sphingosine 1-phospate receptor (S1PR) modulator siponimod on action potential firing and the inward current (Ih) carried by HCN ion channels in naive conditions and during different stages of de- and remyelination. Here, DRF application reduced Ih current density in ex vivo patch clamp recordings from TC neurons of the ventrobasal thalamic complex (VB), thereby counteracting the increase of Ih during early remyelination. Siponimod reduced Ih in VB neurons under control conditions but had no effect in neurons of the auditory cortex (AU). Furthermore, siponimod increased and decreased AP firing properties of neurons in VB and AU, respectively. Computational modeling revealed that both DRF and siponimod influenced thalamic bursting during early remyelination by delaying the onset and decreasing the interburst frequency. Thus, substances used in MS treatment normalize excitability in the TC system by influencing AP firing and Ih.
Collapse
Affiliation(s)
- Laura Vinnenberg
- Department of Neurology with Institute of Translational Neurology, Münster University, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Nicole Rychlik
- Institute of Physiology I, Münster University, Robert-Koch-Str. 27a, D-48149 Münster, Germany.
| | - Tengiz Oniani
- Institute of Physiology I, Münster University, Robert-Koch-Str. 27a, D-48149 Münster, Germany
| | - Brandon Williams
- Department of Biomedical Engineering, Center for Systems Neuroscience, Neurophotonics Center, Boston University, 610 Commonwealth Ave, Boston MA-02215, USA
| | - John A White
- Department of Biomedical Engineering, Center for Systems Neuroscience, Neurophotonics Center, Boston University, 610 Commonwealth Ave, Boston MA-02215, USA
| | - Stjepana Kovac
- Department of Neurology with Institute of Translational Neurology, Münster University, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| | - Sven G Meuth
- Neurology Clinic, Medical Faculty, University Clinic Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany
| | - Thomas Budde
- Institute of Physiology I, Münster University, Robert-Koch-Str. 27a, D-48149 Münster, Germany
| | - Petra Hundehege
- Department of Neurology with Institute of Translational Neurology, Münster University, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany
| |
Collapse
|
19
|
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
|
20
|
Oizumi H, Miyamoto Y, Seiwa C, Yamamoto M, Yoshioka N, Iizuka S, Torii T, Ohbuchi K, Mizoguchi K, Yamauchi J, Asou H. Lethal adulthood myelin breakdown by oligodendrocyte-specific Ddx54 knockout. iScience 2023; 26:107448. [PMID: 37720086 PMCID: PMC10502337 DOI: 10.1016/j.isci.2023.107448] [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: 02/21/2022] [Revised: 05/08/2023] [Accepted: 07/18/2023] [Indexed: 09/19/2023] Open
Abstract
Multiple sclerosis (MS) is a leading disease that causes disability in young adults. We have previously shown that a DEAD-box RNA helicase Ddx54 binds to mRNA and protein isoforms of myelin basic protein (MBP) and that Ddx54 siRNA blocking abrogates oligodendrocyte migration and myelination. Herein, we show that MBP-driven Ddx54 knockout mice (Ddx54 fl/fl;MBP-Cre), after the completion of normal postnatal myelination, gradually develop abnormalities in behavioral profiles and learning ability, inner myelin sheath breakdown, loss of myelinated axons, apoptosis of oligodendrocytes, astrocyte and microglia activation, and they die within 7 months but show minimal peripheral immune cell infiltration. Myelin in Ddx54fl/fl;MBP-Cre is highly vulnerable to the neurotoxicant cuprizone and Ddx54 knockdown greatly impairs myelination in vitro. Ddx54 expression in oligodendrocyte-lineage cells decreased in corpus callosum of MS patients. Our results demonstrate that Ddx54 is indispensable for myelin homeostasis, and they provide a demyelinating disease model based on intrinsic disintegration of adult myelin.
Collapse
Affiliation(s)
- Hiroaki Oizumi
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | - Yuki Miyamoto
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Chika Seiwa
- Glovia Myelin Research Institute, Tsurumi-ku, Yokohama, Kanagawa 230-0046, Japan
| | - Masahiro Yamamoto
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | - Nozomu Yoshioka
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Chuo-ku, Niigata 951-8510, Japan
| | - Seiichi Iizuka
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | - Tomohiro Torii
- Laboratory of Ion Channel Pathophysiology, Graduate School of Brain Science, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| | - Katsuya Ohbuchi
- Tsumura Kampo Laboratories, Tsumura & Co, Ami, Ibaraki 300-1192, Japan
| | | | - Junji Yamauchi
- Department of Pharmacology, National Research Institute for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo 192-0392, Japan
| | - Hiroaki Asou
- Glovia Myelin Research Institute, Tsurumi-ku, Yokohama, Kanagawa 230-0046, Japan
| |
Collapse
|
21
|
Tripathi A, Rai N, Perles A, Jones C, Dutta R. Dicer deficiency in microglia leads to accelerated demyelination and failed remyelination. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.17.562812. [PMID: 37905110 PMCID: PMC10614879 DOI: 10.1101/2023.10.17.562812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Microglia are the resident immune cells of the central nervous system (CNS) and are important regulators of normal brain functions. In CNS demyelinating diseases like multiple sclerosis (MS), the functions of these cells are of particular interest. Here we probed the impact of microRNA (miRNA)-mediated post-transcriptional gene regulation using a mouse model lacking microglia/macrophage-specific Dicer expression during demyelination and remyelination. Conditional Dicer ablation and loss of miRNAs in adult microglia led to extensive demyelination and impaired myelin processing. Interestingly, demyelination was accompanied by increased apoptosis of mature oligodendrocytes (OLs) and arresting OL progenitor cells (OPCs) in the precursor stage. At the transcriptional level, Dicer -deficient microglia led to downregulation of microglial homeostatic genes, increased cell proliferation, and a shift towards a disease-associated phenotype. Loss of remyelination efficiency in these mice was accompanied by stalling of OPCs in the precursor stage. Collectively, these results highlight a new role of microglial miRNAs in promoting a pro-regenerative phenotype in addition to promoting OPC maturation and differentiation during demyelination and remyelination.
Collapse
|
22
|
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
|
23
|
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
|
24
|
Atkinson KC, Osunde M, Tiwari-Woodruff SK. The complexities of investigating mitochondria dynamics in multiple sclerosis and mouse models of MS. Front Neurosci 2023; 17:1144896. [PMID: 37559701 PMCID: PMC10409489 DOI: 10.3389/fnins.2023.1144896] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Accepted: 06/23/2023] [Indexed: 08/11/2023] Open
Abstract
Multiple sclerosis (MS) is a demyelinating, degenerating disorder of the central nervous system (CNS) that is accompanied by mitochondria energy production failure. A loss of myelin paired with a deficit in energy production can contribute to further neurodegeneration and disability in patients in MS. Mitochondria are essential organelles that produce adenosine triphosphate (ATP) via oxidative phosphorylation in all cells in the CNS, including neurons, oligodendrocytes, astrocytes, and immune cells. In the context of demyelinating diseases, mitochondria have been shown to alter their morphology and undergo an initial increase in metabolic demand. This is followed by mitochondrial respiratory chain deficiency and abnormalities in mitochondrial transport that contribute to progressive neurodegeneration and irreversible disability. The current methodologies to study mitochondria are limiting and are capable of providing only a partial snapshot of the true mitochondria activity at a particular timepoint during disease. Mitochondrial functional studies are mostly performed in cell culture or whole brain tissue, which prevents understanding of mitochondrial pathology in distinct cell types in vivo. A true understanding of cell-specific mitochondrial pathophysiology of MS in mouse models is required. Cell-specific mitochondria morphology, mitochondria motility, and ATP production studies in animal models of MS will help us understand the role of mitochondria in the normal and diseased CNS. In this review, we present currently used methods to investigate mitochondria function in MS mouse models and discuss the current advantages and caveats with using each technique. In addition, we present recently developed mitochondria transgenic mouse lines expressing Cre under the control of CNS specific promoters to relate mitochondria to disease in vivo.
Collapse
Affiliation(s)
| | | | - Seema K. Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine, University of California, Riverside, Riverside, CA, United States
| |
Collapse
|
25
|
Martinović K, Bauer J, Kunze M, Berger J, Forss-Petter S. Abcd1 deficiency accelerates cuprizone-induced oligodendrocyte loss and axonopathy in a demyelinating mouse model of X-linked adrenoleukodystrophy. Acta Neuropathol Commun 2023; 11:98. [PMID: 37331971 PMCID: PMC10276915 DOI: 10.1186/s40478-023-01595-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: 04/19/2023] [Accepted: 05/30/2023] [Indexed: 06/20/2023] Open
Abstract
X-linked adrenoleukodystrophy (X-ALD), the most frequent, inherited peroxisomal disease, is caused by mutations in the ABCD1 gene encoding a peroxisomal lipid transporter importing very long-chain fatty acids (VLCFAs) from the cytosol into peroxisomes for degradation via β-oxidation. ABCD1 deficiency results in accumulation of VLCFAs in tissues and body fluids of X-ALD patients with a wide range of phenotypic manifestations. The most severe variant, cerebral X-ALD (CALD) is characterized by progressive inflammation, loss of the myelin-producing oligodendrocytes and demyelination of the cerebral white matter. Whether the oligodendrocyte loss and demyelination in CALD are caused by a primary cell autonomous defect or injury to oligodendrocytes or by a secondary effect of the inflammatory reaction remains unresolved. To address the role of X-ALD oligodendrocytes in demyelinating pathophysiology, we combined the Abcd1 deficient X-ALD mouse model, in which VLCFAs accumulate without spontaneous demyelination, with the cuprizone model of toxic demyelination. In mice, the copper chelator cuprizone induces reproducible demyelination in the corpus callosum, followed by remyelination upon cuprizone removal. By immunohistochemical analyses of oligodendrocytes, myelin, axonal damage and microglia activation during de-and remyelination, we found that the mature oligodendrocytes of Abcd1 KO mice are more susceptible to cuprizone-induced cell death compared to WT mice in the early demyelinating phase. Furthermore, this effect was mirrored by a greater extent of acute axonal damage during demyelination in the KO mice. Abcd1 deficiency did not affect the function of microglia in either phase of the treatment. Also, the proliferation and differentiation of oligodendrocyte precursor cells and remyelination progressed at similar rates in both genotypes. Taken together, our findings point to an effect of Abcd1 deficiency on mature oligodendrocytes and the oligodendrocyte-axon unit, leading to increased vulnerability in the context of a demyelinating insult.
Collapse
Affiliation(s)
- Ksenija Martinović
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Markus Kunze
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Johannes Berger
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| | - Sonja Forss-Petter
- Department of Pathobiology of the Nervous System, Center for Brain Research, Medical University of Vienna, Spitalgasse 4, 1090 Vienna, Austria
| |
Collapse
|
26
|
Gilli F, Ceccarelli A. Magnetic resonance imaging approaches for studying mouse models of multiple sclerosis: A mini review. J Neurosci Res 2023. [DOI: 10.1002/jnr.25193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 01/30/2023] [Accepted: 03/10/2023] [Indexed: 04/03/2023]
Affiliation(s)
- Francesca Gilli
- Department of Neurology, Dartmouth Hitchcock Medical Center Geisel School of Medicine at Dartmouth Lebanon New Hampshire USA
| | - Antonia Ceccarelli
- Department of Neurology EpiCURA Centre Hospitalier Ath Belgium
- Hearthrhythmanagement, UZB Brussels Belgium
| |
Collapse
|
27
|
Wang Y, Sun J, Zhu K, Wang D, Zhao X, Zhang H, Wu S, Wang Y, Wang J. Microglial aryl hydrocarbon receptor enhances phagocytic function via SYK and promotes remyelination in the cuprizone mouse model of demyelination. J Neuroinflammation 2023; 20:83. [PMID: 36966295 PMCID: PMC10040134 DOI: 10.1186/s12974-023-02764-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 03/11/2023] [Indexed: 03/27/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory-mediated demyelinating disease of the central nervous system (CNS). Although studies have demonstrated that microglia facilitate remyelination in demyelinating diseases, the underlying mechanisms are still not fully characterized. We found that aryl hydrocarbon receptor (AhR), an environment sensor, was upregulated within the corpus callosum in the cuprizone model of CNS demyelination, and upregulated AhR was mainly confined to microglia. Deletion of AhR in adult microglia inhibited efficient remyelination. Transcriptome analysis using RNA-seq revealed that AhR-deficient microglia displayed impaired gene expression signatures associated with lysosome and phagocytotic pathways. Furthermore, AhR-deficient microglia showed impaired clearance of myelin debris and defected phagocytic capacity. Further investigation of target genes of AhR revealed that spleen tyrosine kinase (SYK) is the downstream effector of AhR and mediated the phagocytic capacity of microglia. Additionally, AhR deficiency in microglia aggravated CNS inflammation during demyelination. Altogether, our study highlights an essential role for AhR in microglial phagocytic function and suggests the therapeutic potential of AhR in demyelinating diseases.
Collapse
Affiliation(s)
- Yumeng Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jingxian Sun
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Keying Zhu
- Department of Clinical Neuroscience, Karolinska Institute, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Danjie Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xiaoqiang Zhao
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongyu Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shuai Wu
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanqing Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Jun Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
28
|
Molinari YA, Byrne AJ, Pérez MJ, Silvestroff L, Franco PG. The Effects of Cuprizone on Murine Subventricular Zone-Derived Neural Stem Cells and Progenitor Cells Grown as Neurospheres. Mol Neurobiol 2023; 60:1195-1213. [PMID: 36424468 DOI: 10.1007/s12035-022-03096-8] [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: 05/10/2022] [Accepted: 10/17/2022] [Indexed: 11/25/2022]
Abstract
Despite the extensive use of the cuprizone (CPZ) demyelination animal model, there is little evidence regarding the effects of CPZ on a cellular level. Initial studies have suggested that oligodendrocytes (OL) are the main cell targets for CPZ toxicity. However, recent data have revealed additional effects on neural stem cells and progenitor cells (NSC/NPC), which constitute a reservoir for OL regeneration during brain remyelination. We cultured NSC/NPC as neurospheres to investigate CPZ effects on cell mechanisms which are thought to be involved in demyelination and remyelination processes in vivo. Proliferating NSC/NPC cultures exposed to CPZ showed overproduction of intracellular reactive oxygen species and increased progenitor migration at the expense of a significant inhibition of cell proliferation. Although NSC/NPC survival was not affected by CPZ in proliferative conditions, we found that CPZ-treated cultures undergoing cell differentiation were more prone to cell death than controls. The commitment and cell differentiation towards neural lineages did not seem to be affected by CPZ, as shown by the conserved proportions of OL, astrocytes, and neurons. Nevertheless, when CPZ treatment was performed after cell differentiation, we detected a significant reduction in the number and the morphological complexity of OL, astrogliosis, and neuronal damage. We conclude that, in addition to damaging mature OL, CPZ also reduces NSC/NPC proliferation and activates progenitor migration. These results shed light on CPZ direct effects on NSC proliferation and the progression of in vitro differentiation.
Collapse
Affiliation(s)
- Yamila Azul Molinari
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Buenos Aires, Argentina.,CONICET- Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - Agustín Jesús Byrne
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Buenos Aires, Argentina.,CONICET- Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - María Julia Pérez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Buenos Aires, Argentina.,CONICET- Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - Lucas Silvestroff
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Buenos Aires, Argentina.,CONICET- Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - Paula Gabriela Franco
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Química Biológica, Cátedra de Química Biológica Patológica, Buenos Aires, Argentina. .,CONICET- Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina.
| |
Collapse
|
29
|
Noori T, Dehpour AR, Alavi SD, Hosseini SZ, Korani S, Sureda A, Esmaeili J, Shirooie S. Synthesis and evaluation of the effects of solid lipid nanoparticles of ivermectin and ivermectin on cuprizone-induced demyelination via targeting the TRPA1/NF-kB/GFAP signaling pathway. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2023; 26:1272-1282. [PMID: 37886003 PMCID: PMC10598811 DOI: 10.22038/ijbms.2023.71309.15493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 05/30/2023] [Indexed: 10/28/2023]
Abstract
Objectives Multiple sclerosis (MS) is a chronic disease of the central nervous system (CNS) and its cause is unknown. Several environmental and genetic factors may have roles in the pathogenesis of MS. The synthesis of solid lipid nanoparticles (SLNs) for ivermectin (IVM) loading was performed to increase its efficiency and bioavailability and evaluate its ability in improving the behavioral and histopathological changes induced by cuprizone (CPZ) in the male C57BL/6 mice. Materials and Methods Four groups of 7 adult C57BL/6 mice including control (normal diet), CPZ, IVM, and nano-IVM groups were chosen. After synthesis of nano-ivermectin, demyelination was induced by adding 0.2% CPZ to animal feed for 6 weeks. IVM and nano-IVM (1 mg/kg/day, IP) were given for the final 14 days of the study. At last, behavioral tests, histochemical assays, and immunohistochemistry of TRPA1, NF-kB p65, and GFAP were done. Results The time of immobility of mice in the IVM and nano-IVM groups was reduced compared to the CPZ group. Histopathological examination revealed demyelination in the CPZ group, which was ameliorated by IVM and nano-IVM administration. In IVM and nano-IVM groups corpus callosum levels of TRPA1, NF-kB p65, and GFAP were decreased compared to the CPZ group. In the IVM and nano-IVM groups, the levels of MBP were significantly higher than in the CPZ group. Conclusion The results evidenced that IVM and nano-IVM administration is capable of reducing demyelination in mice.
Collapse
Affiliation(s)
- Tayebeh Noori
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Experimental medicine research center, Tehran University of medical sciences, Tehran, Iran
| | - Seyede Darya Alavi
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Seyede Zahra Hosseini
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Sina Korani
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress (NUCOX) and Health Research Institute of Balearic Islands (IdISBa), University of Balearic Islands-IUNICS, Palma de Mallorca E-07122, Balearic Islands, Spain
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
| | - Jamileh Esmaeili
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Samira Shirooie
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
30
|
Ahmad I, Wergeland S, Oveland E, Bø L. An Association of Chitinase-3 Like-Protein-1 With Neuronal Deterioration in Multiple Sclerosis. ASN Neuro 2023; 15:17590914231198980. [PMID: 38062768 PMCID: PMC10710113 DOI: 10.1177/17590914231198980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 07/28/2023] [Accepted: 08/16/2023] [Indexed: 12/18/2023] Open
Abstract
Elevated levels of Chitinase-3-like protein-1 (CHI3L1) in cerebrospinal fluid have previously been linked to inflammatory activity and disease progression in multiple sclerosis (MS) patients. This study aimed to investigate the presence of CHI3L1 in the brains of MS patients and in the cuprizone model in mice (CPZ), a model of toxic/metabolic demyelination and remyelination in different brain areas. In MS gray matter (GM), CHI3L1 was detected primarily in astrocytes and in a subset of pyramidal neurons. In neurons, CHI3L1 immunopositivity was associated with lipofuscin-like substance accumulation, a sign of cellular aging that can lead to cell death. The density of CHI3L1-positive neurons was found to be significantly higher in normal-appearing MS GM tissue compared to that of control subjects (p = .014). In MS white matter (WM), CHI3L1 was detected in astrocytes located within lesion areas, as well as in perivascular normal-appearing areas and in phagocytic cells from the initial phases of lesion development. In the CPZ model, the density of CHI3L1-positive cells was strongly associated with microglial activation in the WM and choroid plexus inflammation. Compared to controls, CHI3L1 immunopositivity in WM was increased from an early phase of CPZ exposure. In the GM, CHI3L1 immunopositivity increased later in the CPZ exposure phase, particularly in the deep GM region. These results indicate that CHI3L1 is associated with neuronal deterioration, pre-lesion pathology, along with inflammation in MS.
Collapse
Affiliation(s)
- Intakhar Ahmad
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| | - Stig Wergeland
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Norwegian MS-registry and biobank, Department of Neurology, Haukeland University Hospital, Bergen, Norway
- Neuro-SysMed, Haukeland University Hospital, Bergen, Norway
| | - Eystein Oveland
- Proteomics Unit at the University of Bergen (PROBE), Department of Biomedicine, University of Bergen, Bergen, Norway
- Institute of Marine Research, IMR, Bergen, Norway
| | - Lars Bø
- Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Norwegian Multiple Sclerosis Competence Centre, Department of Neurology, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
31
|
Labunets I, Rodnichenko A, Savosko S, Pivneva T. Reaction of different cell types of the brain on neurotoxin cuprizone and hormone melatonin treatment in young and aging mice. Front Cell Neurosci 2023; 17:1131130. [PMID: 37153635 PMCID: PMC10157497 DOI: 10.3389/fncel.2023.1131130] [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: 12/24/2022] [Accepted: 03/30/2023] [Indexed: 05/10/2023] Open
Abstract
Introduction The brain myelin and neurons destruction in multiple sclerosis may be associated with the production of neuroinflammatory cells (macrophages, astrocytes, T-lymphocytes) of pro-inflammatory cytokines and free radicals. The age-associated changes of the above cells can influence on the response of nervous system cells to toxic damaging and regulatory factors of humoral/endocrine nature, in particular pineal hormone melatonin. The study aim was (1) to evaluate changes of the brain macrophages, astrocytes, T-cells, neural stem cells, neurons, and central nervous system (CNS) functioning in the neurotoxin cuprizone-treated mice of different age; and (2) to assess in such mice the effects of exogenous melatonin and possible courses of its action. Methods A toxic demyelination and neurodegeneration model was induced in 129/Sv mice aged 3-5 and 13-15 months by adding cuprizone neurotoxin to their food for 3 weeks. From the 8th day of the cuprizone treatment, melatonin was injected intraperitoneally at 6 p.m. daily, at a dose of 1 mg/kg. The brain GFPA + -cells were evaluated by immunohistochemical method, the proportion of CD11b+, CD3+CD11b+, CD3+, CD3+CD4+, CD3+CD8+, Nestin+-cells was determined via flow cytometry. Macrophage activity was evaluated by their ability to phagocytose latex beads Morphometric analysis of the brain neurons and the behavioral reactions ("open field" and rotarod tests) were performed. To assess the involvement of the bone marrow and thymus in the action of melatonin, the amount of granulocyte/macrophage colony-forming cells (GM-CFC), and blood monocytes and thymic hormone thymulin were evaluated. Results and discussion The numbers of the GFAP+-, CD3+-, CD3+CD4+, CD3+CD8+, CD11b+, CD3+CD11b+, Nestin+-cells and macrophages phagocytic latex beads and malondialdehyde (MDA) content were increased in the brain of young and aging mice under cuprizone influence. The proportion of undamaged neurons within the brain, motor, affective, and exploratory activities, and muscle tone decreased in mice of both ages. Introducing melatonin to mice of any age reduced the number of GFAP+-, CD3+- cells and their subpopulations, macrophage activation, and MDA content. At the same time, the percentage of brain neurons that were unchanged increased as the number of Nestin+ cells decreased. The behavioral responses were also improved. Besides, the number of bone marrow GM-CFC and the blood level of monocytes and thymulin increased. The effects of both neurotoxin and melatonin on the brain astrocytes, macrophages T-cells, and immune system organs as well as the structure and functioning of neurons were more pronounced in the young mice. Conclusion We have observed the involvement of the astrocytes, macrophages, T-cells, neural stem cells, and neurons in the brain reaction of mice different age after administration of neurotoxin cuprizone and melatonin. The brain cell composition reaction has the age features. The neuroprotective effects of melatonin in cuprizone-treated mice have been realized through an improvement of the brain cell composition and oxidative stress factors and functioning of bone marrow and thymus.
Collapse
Affiliation(s)
- Irina Labunets
- Cell and Tissue Technologies Department, Institute of Genetic and Regenerative Medicine, National Scientific Center “M.D. Strazhesko Institute of Cardiology”, Clinical and Regenerative Medicine of the National Academy of Medical Sciences of Ukraine, Kyiv, Ukraine
- Laboratory of Pathophysiology and Immunology, D. F. Chebotarev Institute of Gerontology of the National Academy of Medical Sciences of Ukraine, Kyiv, Ukraine
- *Correspondence: Irina Labunets,
| | - Anzhela Rodnichenko
- Cell and Tissue Technologies Department, Institute of Genetic and Regenerative Medicine, National Scientific Center “M.D. Strazhesko Institute of Cardiology”, Clinical and Regenerative Medicine of the National Academy of Medical Sciences of Ukraine, Kyiv, Ukraine
| | - Sergey Savosko
- Cell and Tissue Technologies Department, Institute of Genetic and Regenerative Medicine, National Scientific Center “M.D. Strazhesko Institute of Cardiology”, Clinical and Regenerative Medicine of the National Academy of Medical Sciences of Ukraine, Kyiv, Ukraine
| | - Tetyana Pivneva
- Department of Sensory Signaling, Bogomoletz Institute of Physiology, National Academy of Sciences of Ukraine, Kyiv, Ukraine
- Department of Biomedicine and Neuroscience, Kyiv Academic University, Kyiv, Ukraine
| |
Collapse
|
32
|
Inhomogeneous Magnetization Transfer (ihMT) imaging in the acute cuprizone mouse model of demyelination/remyelination. Neuroimage 2023; 265:119785. [PMID: 36464096 DOI: 10.1016/j.neuroimage.2022.119785] [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: 09/03/2022] [Revised: 10/31/2022] [Accepted: 12/01/2022] [Indexed: 12/05/2022] Open
Abstract
BACKGROUND To investigate the association of ihMT (inhom signals with the demyelination and remyelination phases of the acute cuprizone mouse model in comparison with histology, and to assess the extent of tissue damage and repair from MRI data. METHODS Acute demyelination by feeding 0.2% cuprizone for five weeks, followed by a four-week remyelination period was applied on genetically modified plp-GFP mice. Animals were scanned at different time points of the demyelination and remyelination phases of the cuprizone model using a multimodal MRI protocol, including ihMT T1D-filters, MPF (Macromolecular Proton Fraction) and R1 (longitudinal relaxation rate). For histology, plp-GFP (proteolipid protein - Green Fluorescent Protein) microscopy and LFB (Luxol Fast Blue) staining were employed as references for the myelin content. Comparison of MRI with histology was performed in the medial corpus callosum (mCC) and cerebral cortex (CTX) at two brain levels whereas ROI-wise and voxel-based analyses of the MRI metrics allowed investigating in vivo the spatial extent of myelin alterations. RESULTS IhMT high-pass T1D-filters, targeted toward long T1D components, showed significant temporal variations in the mCC consistent with the effects induced by the cuprizone toxin. In addition, the corresponding signals correlated strongly and significantly with the myelin content assessed by GFP fluorescence and LFB staining over the demyelination and the remyelination phases. The signal of the band-pass T1D-filter, which isolates short T1D components, showed changes over time that were poorly correlated with histology, hence suggesting a sensitivity to pathological processes possibly not related to myelin. Although MPF was also highly correlated to histology, ihMT high-pass T1D-filters showed better capability to characterize the spatial-temporal patterns during the demyelination and remyelination phases of the acute cuprizone model (e.g., rostro-caudal gradient of demyelination in the mCC previously described in the literature). CONCLUSIONS IhMT sequences selective for long T1D components are specific and sensitive in vivo markers of demyelination and remyelination and have successfully captured the spatially heterogeneous pattern of the demyelination and remyelination mechanisms in the cuprizone model. Interestingly, differences in signal variations between the ihMT high-pass and band-pass T1D-filter, suggest a sensitivity of the ihMT sequences targeted to short T1Ds to alterations other than those of myelin. Future studies will need to further address these differences by examining more closely the origin of the short T1D components and the variation of each T1D component in pathology.
Collapse
|
33
|
Zhang H, Wang D, Sun J, Wang Y, Wu S, Wang J. Huperzine-A Improved Animal Behavior in Cuprizone-Induced Mouse Model by Alleviating Demyelination and Neuroinflammation. Int J Mol Sci 2022; 23:ijms232416182. [PMID: 36555825 PMCID: PMC9785798 DOI: 10.3390/ijms232416182] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 12/12/2022] [Accepted: 12/13/2022] [Indexed: 12/23/2022] Open
Abstract
Huperzine A (HupA) is a natural acetylcholinesterase inhibitor (AChEI) with the advantages of high efficiency, selectivity as well as reversibility and can exhibit significant therapeutic effects against certain neurodegenerative diseases. It is also beneficial in reducing the neurological impairment and neuroinflammation of experimental autoimmune encephalomyelitis (EAE), a classic model for multiple sclerosis (MS). However, whether HupA can directly regulate oligodendrocyte differentiation and maturation and promote remyelination has not been investigated previously. In this study, we have analyzed the potential protective effects of HupA on the demylination model of MS induced by cuprizone (CPZ). It was found that HupA significantly attenuated anxiety-like behavior, as well as augmented motor and cognitive functions in CPZ mice. It also decreased demyelination and axonal injury in CPZ mice. Moreover, in CPZ mice, HupA increased mRNA levels of the various anti-inflammatory cytokines (Arg1, CD206) while reducing the levels of different pro-inflammatory cytokines (iNOS, IL-1β, IL-18, CD16, and TNF-α). Mecamylamine, a nicotinic acetylcholinergic receptor antagonist, could effectively reverse the effects of HupA. Therefore, we concluded that HupA primarily exerts its therapeutic effects on multiple sclerosis through alleviating demyelination and neuroinflammation.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Danjie Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jingxian Sun
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yumeng Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Department of Integrative Medicine and Neurobiology, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Institutes of Brain Science, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shuai Wu
- Department of Neurology, Zhongshan Hospital, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Correspondence: (S.W.); (J.W.); Tel.: +86-15921977760 (S.W.); +86-17721371757 (J.W.)
| | - Jun Wang
- Department of Integrative Medicine and Neurobiology, School of Basic Medical Science, Institutes of Integrative Medicine, Shanghai Medical College, Fudan University, Shanghai 200032, China
- Correspondence: (S.W.); (J.W.); Tel.: +86-15921977760 (S.W.); +86-17721371757 (J.W.)
| |
Collapse
|
34
|
Wuzi Yanzong Pill relieves CPZ-induced demyelination by improving the microenvironment in the brain. Heliyon 2022; 8:e12277. [PMID: 36578409 PMCID: PMC9791345 DOI: 10.1016/j.heliyon.2022.e12277] [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: 12/17/2021] [Revised: 08/28/2022] [Accepted: 12/02/2022] [Indexed: 12/14/2022] Open
Abstract
Ethnopharmacology relevance Wuzi Yanzong Pill (WYP), a well-known prescription for invigorating the kidney and essence, which is widely used to treat infertility such as oligoasthenospermia. Studies have shown that WYP can be used to treat neurological diseases, but its therapeutic effects and mechanisms for multiple sclerosis (MS) remain unclear. Aim of the study Based on the establishment of Cuprizone (CPZ)-induced demyelination model, this study determined the effect of WYP on remyelination by detecting changes in the microenvironment of the central nervous system. Materials and methods C57BL/6 mice were divided into three groups. The CPZ group and CPZ + WYP group were fed with 0.2% CPZ feed, and the control group was fed normal feed, for 6 weeks. At the end of the second week, the CPZ + WYP group was gavaged with WYP solution (16 g/kg/d), and the other two groups were gavaged with normal saline twice a day with an interval of 12 h each time, for 4 weeks. Forced swimming and elevated plus maze were used to detect changes in anxiety and depression before and after treatment. Luxol fast blue staining and the expression of MBP were used to evaluate the demyelination of the brain. Western blot was used to detect the expression of microglia and their subtype markers Iba-1, Arg-1, iNOS, the expression of neurotrophic factors BDNF, GDNF, CNTF, and the expression of oligodendrocyte precursor cells NG2. ELISA detected the content of IL-6, IL-1β, IL-10, TGF-β, BDNF, GDNF, CNTF in the brain. The distribution of Iba-1 in the corpus callosum was observed by immunofluorescence. Results The results showed that on the basis of improving mood abnormalities and demyelination, WYP reduced the protein content of Iba-1 and iNOS, increased the protein content of Arg-1, and reduce accumulation of microglia in the corpus callosum. In addition, WYP reduced the secretion of IL-6 and IL-1β while promoting the secretion of IL-10 and TGF-β. After WYP intervention treatment, the levels of neurotrophic factors BDNF, GDNF, CNTF increased. Due to the improvement of inflammatory and nutritional environment in the CNS, promoting the proliferation of NG2 oligodendrocyte, increased the expression of MBP, and repairing myelin sheath. Conclusion Our results indicated that WYP promoted the proliferation and development of oligodendrocytes by improving the CNS microenvironment, effectively alleviating demyelination.
Collapse
|
35
|
Sex Differences in the Behavioural Aspects of the Cuprizone-Induced Demyelination Model in Mice. Brain Sci 2022; 12:brainsci12121687. [PMID: 36552147 PMCID: PMC9775311 DOI: 10.3390/brainsci12121687] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/28/2022] [Accepted: 12/06/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis is an autoimmune disease characterised by demyelination in the central nervous system. The cuprizone-induced demyelination model is often used in mice to test novel treatments for multiple sclerosis. However, despite significant demyelination, behavioural deficits may be subtle or have mixed results depending on the paradigm used. Furthermore, the sex differences within the model are not well understood. In the current study, we have sought to understand the behavioural deficits associated with the cuprizone-induced demyelination model in both male and female C57BL/6J mice. Using Black gold II stain, we found that cuprizone administration over 6 weeks caused significant demyelination in the corpus callosum that was consistent across both sexes. Cuprizone administration caused increased mechanical sensitivity when measured using an electronic von Frey aesthesiometer, with no sex differences observed. However, cuprizone administration decreased motor coordination, with more severe deficits seen in males in the horizontal bar and passive wire hang tests. In contrast, female mice showed more severe deficits in the motor skill sequence test. Cuprizone administration caused more anxiety-like behaviours in males compared to females in the elevated zero maze. Therefore, this study provides a better understanding of the sex differences involved in the behavioural aspects of cuprizone-induced demyelination, which could allow for a better translation of results from the laboratory to the clinic.
Collapse
|
36
|
Sun W, Wen M, Liu M, Wang Q, Liu Q, Li L, Siebert HC, Loers G, Zhang R, Zhang N. Effect of β-hydroxybutyrate on behavioral alterations, molecular and morphological changes in CNS of multiple sclerosis mouse model. Front Aging Neurosci 2022; 14:1075161. [PMID: 36533180 PMCID: PMC9752847 DOI: 10.3389/fnagi.2022.1075161] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 11/14/2022] [Indexed: 09/29/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory and degenerative disease of central nervous system (CNS). Aging is the most significant risk factor for the progression of MS. Dietary modulation (such as ketogenic diet) and caloric restriction, can increase ketone bodies, especially β-hydroxybutyrate (BHB). Increased BHB has been reported to prevent or improve age-related disease. The present studies were performed to understand the therapeutic effect and potential mechanisms of exogenous BHB in cuprizone (CPZ)-induced demyelinating model. In this study, a continuous 35 days CPZ mouse model with or without BHB was established. The changes of behavior function, pathological hallmarks of CPZ, and intracellular signal pathways in mice were detected by Open feld test, Morris water maze, RT-PCR, immuno-histochemistry, and western blot. The results showed that BHB treatment improved behavioral performance, prevented myelin loss, decreased the activation of astrocyte as well as microglia, and up-regulated the neurotrophin brain-derived neurotrophic factor in both the corpus callosum and hippocampus. Meanwhile, BHB treatment increased the number of MCT1+ cells and APC+ oligodendrocytes. Furthermore, the treatment decreased the expression of HDAC3, PARP1, AIF and TRPA1 which is related to oligodendrocyte (OL) apoptosis in the corpus callosum, accompanied by increased expression of TrkB. This leads to an increased density of doublecortin (DCX)+ neuronal precursor cells and mature NeuN+ neuronal cells in the hippocampus. As a result, BHB treatment effectively promotes the generation of PDGF-Ra+ (oligodendrocyte precursor cells, OPCs), Sox2+ cells and GFAP+ (astrocytes), and decreased the production of GFAP+ TRAP1+ cells, and Oligo2+ TRAP1+ cells in the corpus callosum of mouse brain. Thus, our results demonstrate that BHB treatment efficiently supports OPC differentiation and decreases the OLs apoptosis in CPZ-intoxicated mice, partly by down-regulating the expression of TRPA1 and PARP, which is associated with the inhibition of the p38-MAPK/JNK/JUN pathway and the activation of ERK1/2, PI3K/AKT/mTOR signaling, supporting BHB treatment adjunctive nutritional therapy for the treatment of chronic demyelinating diseases, such as multiple sclerosis (MS).
Collapse
Affiliation(s)
- Wei Sun
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Min Wen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Min Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Qingpeng Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Quiqin Liu
- Shandong Donkey Industry, Technology Collaborative Innovation Center, Liaocheng University, Liaocheng, China
| | - Lanjie Li
- Shandong Donkey Industry, Technology Collaborative Innovation Center, Liaocheng University, Liaocheng, China
| | - Hans-Christian Siebert
- Schauenburgerstr, RI-B-NT - Research Institute of Bioinformatics and Nanotechnology, Kiel University, Kiel, Germany
| | - Gabriele Loers
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, University of Hamburg, Hamburg, Germany
| | - Ruiyan Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong, China
- Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| |
Collapse
|
37
|
Gharagozloo M, Mace JW, Calabresi PA. Animal models to investigate the effects of inflammation on remyelination in multiple sclerosis. Front Mol Neurosci 2022; 15:995477. [PMID: 36407761 PMCID: PMC9669474 DOI: 10.3389/fnmol.2022.995477] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Accepted: 09/22/2022] [Indexed: 09/19/2023] Open
Abstract
Multiple sclerosis (MS) is a chronic inflammatory, demyelinating, and neurodegenerative disease of the central nervous system (CNS). In people with MS, impaired remyelination and axonal loss lead to debilitating long-term neurologic deficits. Current MS disease-modifying drugs mainly target peripheral immune cells and have demonstrated little efficacy for neuroprotection or promoting repair. To elucidate the pathological mechanisms and test therapeutic interventions, multiple animal models have been developed to recapitulate specific aspects of MS pathology, particularly the acute inflammatory stage. However, there are few animal models that facilitate the study of remyelination in the presence of inflammation, and none fully replicate the biology of chronic demyelination in MS. In this review, we describe the animal models that have provided insight into the mechanisms underlying demyelination, myelin repair, and potential therapeutic targets for remyelination. We highlight the limitations of studying remyelination in toxin-based demyelination models and discuss the combinatorial models that recapitulate the inflammatory microenvironment, which is now recognized to be a major inhibitor of remyelination mechanisms. These models may be useful in identifying novel therapeutics that promote CNS remyelination in inflammatory diseases such as MS.
Collapse
Affiliation(s)
- Marjan Gharagozloo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Jackson W. Mace
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Peter A. Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, United States
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
38
|
Lee J, Villarreal OD, Wang YC, Ragoussis J, Rivest S, Gosselin D, Richard S. PRMT1 is required for the generation of MHC-associated microglia and remyelination in the central nervous system. Life Sci Alliance 2022; 5:5/10/e202201467. [PMID: 35705491 PMCID: PMC9201232 DOI: 10.26508/lsa.202201467] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Revised: 06/03/2022] [Accepted: 06/03/2022] [Indexed: 11/24/2022] Open
Abstract
PRMT1 regulates MHC-associated microglia cluster during de/remyelination. Remyelination failure in multiple sclerosis leads to progressive demyelination and inflammation, resulting in neurodegeneration and clinical decline. Microglia are innate immune cells that can acquire a regenerative phenotype to promote remyelination, yet little is known about the regulators controlling the regenerative microglia activation. Herein, using a cuprizone (CPZ)-diet induced de- and remyelination mice model, we identify PRMT1 as a driver for MHC-associated microglia population required for remyelination in the central nervous system. The loss of PRMT1, but not PRMT5, in microglia resulted in impairment of the remyelination with a reduction of oligoprogenitor cell number and prolonged microgliosis and astrogliosis. Using single-cell RNA sequencing, we found eight distinct microglial clusters during the CPZ diet, and PRMT1 depleted microglia hindered the formation of the MHC-associated cluster, expressing MHCII and CD11c. Mechanistically, PRMT1-KO microglia displayed reduced the H3K27ac peaks at the promoter regions of the MHC- and IFN-associated genes and further suppressed gene expression during CPZ diet. Overall, our findings demonstrate that PRMT1 is a critical regulator of the MHC- and IFN-associated microglia, necessary for central nervous system remyelination.
Collapse
Affiliation(s)
- Jeesan Lee
- Segal Cancer Center, Lady Davis Institute for Medical Research and Gerald Bronfman Department of Oncology and Departments of Biochemistry, Human Genetics, and Medicine, McGill University, Montreal, Canada
| | - Oscar David Villarreal
- Segal Cancer Center, Lady Davis Institute for Medical Research and Gerald Bronfman Department of Oncology and Departments of Biochemistry, Human Genetics, and Medicine, McGill University, Montreal, Canada
| | - Yu Chang Wang
- McGill Genome Centre, Department of Human Genetics, McGill University, Montreal, Canada
| | - Jiannis Ragoussis
- McGill Genome Centre, Department of Human Genetics, McGill University, Montreal, Canada
| | - Serge Rivest
- Neuroscience Laboratory, CHU de Quebec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, Canada
| | - David Gosselin
- Neuroscience Laboratory, CHU de Quebec Research Center and Department of Molecular Medicine, Faculty of Medicine, Laval University, Quebec City, Canada
| | - Stéphane Richard
- Segal Cancer Center, Lady Davis Institute for Medical Research and Gerald Bronfman Department of Oncology and Departments of Biochemistry, Human Genetics, and Medicine, McGill University, Montreal, Canada
| |
Collapse
|
39
|
Marzban M, Rustamzadeh A, Asghari A, Terme Y, Amichi AG, Ghanbarzehi V, Holaso AS, Hosseini F, Shahraki M, Sadafi P, Hashemzahi E, Honardar M, Iravankhah M, Baloochi M, Yarmohammadi A, Ebrahimi P. Stem cell therapy for cuprizone model of multiple sclerosis focusing on the effectiveness of different injection methods and cell labeling. Acta Histochem 2022; 124:151953. [PMID: 36116321 DOI: 10.1016/j.acthis.2022.151953] [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: 08/05/2022] [Revised: 09/06/2022] [Accepted: 09/12/2022] [Indexed: 11/01/2022]
Abstract
Multiple Sclerosis (MS) is a chronic and autoimmune disease of the central nervous system that causes inflammation in the brain and spinal cord, progressive degeneration of central nervous system tissue, damage to neuronal axons, and loss of function of central nervous system neurons. Experimental encephalomyelitis is an alternative animal model of MS that can simulate the symptoms of this disease. Cuprizone is one of the factors creating this model. Various researchers are testing the use of different cells to reduce the symptoms of cuprizone-demyelinated mice. The different injection methods explained in this article include intracerebral, intraperitoneal, intravenous, and intranasal. The intracerebral method, in contrast to the intranasal method, was widely employed by researchers. In each technique, the researchers try to inject a specific type of stem cell (SC) and monitor their efficiency. For monitoring SCs various labeling procedures are available, however, there is an upward trend in using magnetic resonance imaging (MRI). Two main barriers to using this method are high cost and complexity. In the current review, we try to make review cell therapy in the cuprizone model of MS.
Collapse
Affiliation(s)
- Mohsen Marzban
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran.
| | - Auob Rustamzadeh
- Department of Anatomical Sciences, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Aria Asghari
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Yousef Terme
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Vahid Ghanbarzehi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | | | - Fateme Hosseini
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mahya Shahraki
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Paniz Sadafi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Erfan Hashemzahi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Minoo Honardar
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Marziyeh Iravankhah
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Mehdi Baloochi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Amin Yarmohammadi
- Student Research Committee, Iranshahr University of Medical Sciences, Iranshahr, Iran
| | - Pirooz Ebrahimi
- Department of Pharmacy, Health and Nutrition Sciences, University of Calabria, Italy
| |
Collapse
|
40
|
Roczkowsky A, Doan MAL, Hlavay BA, Mamik MK, Branton WG, McKenzie BA, Saito LB, Schmitt L, Eitzen G, Di Cara F, Wuest M, Wuest F, Rachubinski R, Power C. Peroxisome Injury in Multiple Sclerosis: Protective Effects of 4-Phenylbutyrate in CNS-Associated Macrophages. J Neurosci 2022; 42:7152-7165. [PMID: 35940876 PMCID: PMC9480879 DOI: 10.1523/jneurosci.0312-22.2022] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 06/14/2022] [Accepted: 07/30/2022] [Indexed: 11/21/2022] Open
Abstract
Multiple sclerosis (MS) is a progressive and inflammatory demyelinating disease of the CNS. Peroxisomes perform critical functions that contribute to CNS homeostasis. We investigated peroxisome injury and mitigating effects of peroxisome-restorative therapy on inflammatory demyelination in models of MS. Human autopsied CNS tissues (male and female), human cell cultures, and cuprizone-mediated demyelination mice (female) were examined by RT-PCR, Western blotting, and immunolabeling. The therapeutic peroxisome proliferator, 4-phenylbutyrate (4-PBA) was investigated in vitro and in vivo White matter from MS patients showed reduced peroxisomal transcript and protein levels, including PMP70, compared with non-MS controls. Cultured human neural cells revealed that human microglia contained abundant peroxisomal proteins. TNF-α-exposed microglia displayed reduced immunolabeling of peroxisomal proteins, PMP70 and PEX11β, which was prevented with 4-PBA. In human myeloid cells exposed to TNF-α or nigericin, suppression of PEX11β and catalase protein levels were observed to be dependent on NLRP3 expression. Hindbrains from cuprizone-exposed mice showed reduced Abcd1, Cat, and Pex5l transcript levels, with concurrent increased Nlrp3 and Il1b transcript levels, which was abrogated by 4-PBA. In the central corpus callosum, Iba-1 in CNS-associated macrophages and peroxisomal thiolase immunostaining after cuprizone exposure was increased by 4-PBA. 4-PBA prevented decreased myelin basic protein and neurofilament heavy chain immunoreactivity caused by cuprizone exposure. Cuprizone-induced neurobehavioral deficits were improved by 4-PBA treatment. Peroxisome injury in CNS-associated macrophages contributed to neuroinflammation and demyelination that was prevented by 4-PBA treatment. A peroxisome-targeted therapy might be valuable for treating inflammatory demyelination and neurodegeneration in MS.SIGNIFICANCE STATEMENT Multiple sclerosis (MS) is a common and disabling disorder of the CNS with no curative therapies for its progressive form. The present studies implicate peroxisome impairment in CNS-associated macrophages (CAMs), which include resident microglia and blood-derived macrophages, as an important contributor to inflammatory demyelination and neuroaxonal injury in MS. We also show that the inflammasome molecule NLRP3 is associated with peroxisome injury in vitro and in vivo, especially in CAMs. Treatment with the peroxisome proliferator 4-phenylbutyrate exerted protective effects with improved molecular, morphologic, and neurobehavioral outcomes that were associated with a neuroprotective CAM phenotype. These findings offer novel insights into the contribution of peroxisome injury in MS together with preclinical testing of a rational therapy for MS.
Collapse
Affiliation(s)
| | - Matthew A L Doan
- The Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | | | | | | | | | | | | | | | - Francesca Di Cara
- Department of Microbiology & Immunology, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | | | | | | | - Christopher Power
- Departments of Medicine
- Medical Microbiology & Immunology
- The Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| |
Collapse
|
41
|
Sun W, Zhang N, Liu B, Yang J, Loers G, Siebert HC, Wen M, Zheng X, Wang Z, Han J, Zhang R. HDAC3 Inhibitor RGFP966 Ameliorated Neuroinflammation in the Cuprizone-Induced Demyelinating Mouse Model and LPS-Stimulated BV2 Cells by Downregulating the P2X7R/STAT3/NF-κB65/NLRP3 Activation. ACS Chem Neurosci 2022; 13:2579-2598. [PMID: 35947794 DOI: 10.1021/acschemneuro.1c00826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Suppression of excessive microglial overactivation can prevent the progression of multiple sclerosis (MS). Histone deacetylases 3 inhibitor (HDAC3i) has been demonstrated to exert anti-inflammatory effects by suppressing microglia (M1-liked) activation. Here, we demonstrate that the RGFP966 (a selective inhibitor of HDAC3) protects white matter after cuprizone-induced demyelination, as shown by reductions in neurological behavioral deficits and increases in myelin basic protein. Moreover, in this study, we found that RGFP966 caused a significant reduction in the levels of inflammatory cytokines, including IL-1β, TNF-α, as well as iNOS, and inhibited microglial (M1-liked) activation in the experimental cuprizone model and LPS-stimulated BV2 cells. Meanwhile, RGFP966 alleviated apoptosis of LPS-induced BV2 cells in vitro. Furthermore, RGFP966 suppressed the expression of P2X7R, NLRP3, ASC, IL-18, IL-1β, and caspase-1, inhibited the ratio of phosphorylated-STAT3/STAT3 and phosphorylated NF-κB p65/NF-κB p65, as well as increased acetylated NF-κB p65 in vitro and in vivo. Furthermore, we confirmed that brilliant blue G (antagonists of P2X7R) suppressed the expression of microglial NLRP3, IL-18, IL-1β, caspase-1, NF-κB p65 (including phosphorylated NF-κB p65), and STAT3 (including phosphorylated STAT3) in vitro. These findings demonstrated that RFFP966 alleviated the inflammatory response and exerted a neuroprotective effect possibly by modulating P2X7R/STAT3/NF-κB65/NLRP3 signaling pathways. Thus, HDAD3 might be considered a promising intervention target for neurodegenerative diseases, such as MS.
Collapse
Affiliation(s)
- Wei Sun
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Ning Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China.,State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Bingyi Liu
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Junrong Yang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Gabriele Loers
- Center for Molecular Neurobiology Hamburg, University Medical Center Hamburg-Eppendorf, University of Hamburg, Falkenried 94, 20251 Hamburg, Germany
| | - Hans-Christian Siebert
- RI-B-NT - Research Institute of Bioinformatics and Nanotechnology, Schauenburgerstr. 116, 24118 Kiel, Germany
| | - Min Wen
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Xuexing Zheng
- Department of Virology, School of Public Health, Shandong University, Jinan 250012, China
| | - Zhengping Wang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Jun Han
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China
| | - Ruiyan Zhang
- Institute of Biopharmaceutical Research, Liaocheng University, Liaocheng, Shandong 252000, China
| |
Collapse
|
42
|
Naeem AG, El-Naga RN, Michel HE. Nebivolol elicits a neuroprotective effect in the cuprizone model of multiple sclerosis in mice: emphasis on M1/M2 polarization and inhibition of NLRP3 inflammasome activation. Inflammopharmacology 2022; 30:2197-2209. [PMID: 35948811 DOI: 10.1007/s10787-022-01045-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/23/2022] [Indexed: 11/05/2022]
Abstract
BACKGROUND AND AIM Multiple sclerosis (MS) is a demyelinating neurodegenerative inflammatory disease affecting mainly young adults. Microgliosis-derived neuroinflammation represents a key hallmark in MS pathology and progression. Nebivolol (Neb) demonstrated antioxidant, anti-inflammatory and neuroprotective properties in several brain pathologies. This study was conducted to investigate the potential neuroprotective effect of Neb in the cuprizone (Cup) model of MS. METHODS C57Bl/6 mice were fed 0.2% Cup mixed into rodent chow for 5 weeks. Neb (5 and 10 mg/kg/day) was administered by oral gavage during the last 2 weeks. RESULTS Neb prevented Cup-induced weight loss and motor deficits as evidenced by increased latency to fall in the rotarod test and enhanced locomotor activity as compared to Cup-intoxicated mice. Neb reversed Cup-induced demyelination as confirmed by Luxol fast blue staining and myelin basic protein western blotting. Administration of Neb modulated microglial activation status by suppressing M1 markers (Iba-1, CD86, iNOS, NO and TNF-α) and increasing M2 markers (Arg-1 and IL-10) as compared to Cup-fed mice. Furthermore, Neb hindered NLRP3/caspase-1/IL-18 inflammatory cascade and alleviated oxidative stress by reducing lipid peroxidation, as well as increasing catalase and superoxide dismutase activities. CONCLUSION These findings suggest the potential neuroprotective effect of Neb in the Cup-induced model of MS in mice, at least partially by virtue of shifting microglia towards M2 phenotype, mitigation of NLRP3 inflammasome activation and alleviation of oxidative stress.
Collapse
Affiliation(s)
- Antoinette G Naeem
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Reem N El-Naga
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Haidy E Michel
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
43
|
Dupree JL, Paez PM, Tiwari-Woodruff SK, Denton TT, Hensley K, Angeliu CG, Boullerne AI, Kalinin S, Egge S, Cheli VT, Denaroso G, Atkinson KC, Feri M, Feinstein DL. Lanthionine Ketimine Ethyl Ester Accelerates Remyelination in a Mouse Model of Multiple Sclerosis. ASN Neuro 2022; 14:17590914221112352. [PMID: 35791633 PMCID: PMC9272172 DOI: 10.1177/17590914221112352] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Although over 20 disease modifying therapies are approved to treat Multiple Sclerosis (MS), these do not increase remyelination of demyelinated axons or mitigate axon damage. Previous studies showed that lanthionine ketenamine ethyl ester (LKE) reduces clinical signs in the experimental autoimmune encephalomyelitis (EAE) mouse model of MS and increased maturation of oligodendrocyte (OL) progenitor cells (OPCs) in vitro. In the current study, we used the cuprizone (CPZ) demyelination model of MS to test if LKE could increase remyelination. The corpus callosum (CC) and somatosensory cortex was examined by immunohistochemistry (IHC), electron microscopy and for mRNA expression changes in mice provided 5 weeks of CPZ diet followed by 2 weeks of normal diet in the presence of LKE or vehicle. A significant increase in the number of myelinated axons, and increased myelin thickness was observed in the CC of LKE-treated groups compared to vehicle-treated groups. LKE also increased myelin basic protein and proteolipid protein expression in the CC and cortex, and increased the number of mature OLs in the cortex. In contrast, LKE did not increase the percentage of proliferating OPCs suggesting effects on OPC survival and differentiation but not proliferation. The effects of LKE on OL maturation and remyelination were supported by similar changes in their relative mRNA levels. Interestingly, LKE did not have significant effects on GFAP or Iba1 immunostaining or mRNA levels. These findings suggest that remyelinating actions of LKE can potentially be formulated to induce remyelination in neurological diseases associated with demyelination including MS.
Collapse
Affiliation(s)
- Jeffrey L. Dupree
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA,Research Service, HH McGuire VA Medical Center, Richmond, VA, USA
| | - Pablo M. Paez
- Institute for Myelin and Glia Exploration, Department of Pharmacology and
Toxicology, University at Buffalo, NY, USA
| | - Seema K. Tiwari-Woodruff
- Division of Biomedical Sciences, School of Medicine at the University of California
Riverside, Riverside, CA, USA
| | - Travis T. Denton
- Department of Pharmaceutical Sciences, College of Pharmacy &
Pharmaceutical Sciences, Washington State University Health Sciences Spokane, Spokane, WA,
USA,Department of Translational Medicine and Physiology, Elson S. Floyd College
of Medicine, Washington State University Health Sciences Spokane, Spokane, WA, USA,Steve Gleason Institute for Neuroscience, Washington State University Health Sciences
Spokane, Spokane, WA, USA
| | - Kenneth Hensley
- Arkansas College of Osteopathic
Medicine, Fort Smith, AR, USA
| | - Christina G. Angeliu
- Institute for Myelin and Glia Exploration, Department of Pharmacology and
Toxicology, University at Buffalo, NY, USA
| | | | - Sergey Kalinin
- Department Anesthesiology, University of Illinois, Chicago, IL, USA
| | - Sophia Egge
- Department of Anatomy and Neurobiology, Virginia Commonwealth University, Richmond, VA, USA
| | - Veronica T. Cheli
- Institute for Myelin and Glia Exploration, Department of Pharmacology and
Toxicology, University at Buffalo, NY, USA
| | - Giancarlo Denaroso
- Institute for Myelin and Glia Exploration, Department of Pharmacology and
Toxicology, University at Buffalo, NY, USA
| | - Kelley C. Atkinson
- Division of Biomedical Sciences, School of Medicine at the University of California
Riverside, Riverside, CA, USA
| | - Micah Feri
- Division of Biomedical Sciences, School of Medicine at the University of California
Riverside, Riverside, CA, USA
| | - Douglas L. Feinstein
- Department Anesthesiology, University of Illinois, Chicago, IL, USA,Jesse Brown VA Medical Center, Chicago, IL, USA,Douglas L. Feinstein, Department of Anesthesiology,
University of Illinois, 835 South Wolcott Avenue, MC 513, Chicago IL, 60612, USA.
| |
Collapse
|
44
|
Oniani T, Vinnenberg L, Chaudhary R, Schreiber JA, Riske K, Williams B, Pape HC, White JA, Junker A, Seebohm G, Meuth SG, Hundehege P, Budde T, Zobeiri M. Effects of Axonal Demyelination, Inflammatory Cytokines and Divalent Cation Chelators on Thalamic HCN Channels and Oscillatory Bursting. Int J Mol Sci 2022; 23:ijms23116285. [PMID: 35682964 PMCID: PMC9181513 DOI: 10.3390/ijms23116285] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 05/31/2022] [Accepted: 05/31/2022] [Indexed: 12/13/2022] Open
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system that is characterized by the progressive loss of oligodendrocytes and myelin and is associated with thalamic dysfunction. Cuprizone (CPZ)-induced general demyelination in rodents is a valuable model for studying different aspects of MS pathology. CPZ feeding is associated with the altered distribution and expression of different ion channels along neuronal somata and axons. However, it is largely unknown whether the copper chelator CPZ directly influences ion channels. Therefore, we assessed the effects of different divalent cations (copper; zinc) and trace metal chelators (EDTA; Tricine; the water-soluble derivative of CPZ, BiMPi) on hyperpolarization-activated cyclic nucleotide-gated (HCN) channels that are major mediators of thalamic function and pathology. In addition, alterations of HCN channels induced by CPZ treatment and MS-related proinflammatory cytokines (IL-1β; IL-6; INF-α; INF-β) were characterized in C57Bl/6J mice. Thus, the hyperpolarization-activated inward current (Ih) was recorded in thalamocortical (TC) neurons and heterologous expression systems (mHCN2 expressing HEK cells; hHCN4 expressing oocytes). A number of electrophysiological characteristics of Ih (potential of half-maximal activation (V0.5); current density; activation kinetics) were unchanged following the extracellular application of trace metals and divalent cation chelators to native neurons, cell cultures or oocytes. Mice were fed a diet containing 0.2% CPZ for 35 days, resulting in general demyelination in the brain. Withdrawal of CPZ from the diet resulted in rapid remyelination, the effects of which were assessed at three time points after stopping CPZ feeding (Day1, Day7, Day25). In TC neurons, Ih was decreased on Day1 and Day25 and revealed a transient increased availability on Day7. In addition, we challenged naive TC neurons with INF-α and IL-1β. It was found that Ih parameters were differentially altered by the application of the two cytokines to thalamic cells, while IL-1β increased the availability of HCN channels (depolarized V0.5; increased current density) and the excitability of TC neurons (depolarized resting membrane potential (RMP); increased the number of action potentials (APs); produced a larger voltage sag; promoted higher input resistance; increased the number of burst spikes; hyperpolarized the AP threshold), INF-α mediated contrary effects. The effect of cytokine modulation on thalamic bursting was further assessed in horizontal slices and a computational model of slow thalamic oscillations. Here, IL-1β and INF-α increased and reduced oscillatory bursting, respectively. We conclude that HCN channels are not directly modulated by trace metals and divalent cation chelators but are subject to modulation by different MS-related cytokines.
Collapse
Affiliation(s)
- Tengiz Oniani
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| | - Laura Vinnenberg
- Department of Neurology with Institute of Translational Neurology, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany; (L.V.); (P.H.)
| | - Rahul Chaudhary
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| | - Julian A. Schreiber
- Institute of Pharmaceutical and Medicinal Chemistry, Westfälische Wilhelms-Universität, Corren-Str. 48, D-48149 Münster, Germany;
- Cellular Electrophysiology and Molecular Biology, Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany;
| | - Kathrin Riske
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität, Waldeyer-Str. 15, D-48149 Münster, Germany; (K.R.); (A.J.)
| | - Brandon Williams
- Center for Systems Neuroscience, Neurophotonics Center, Department of Biomedical Engineering, Boston University, 610 Commonwealth Ave., Boston, MA 02215, USA; (B.W.); (J.A.W.)
| | - Hans-Christian Pape
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| | - John A. White
- Center for Systems Neuroscience, Neurophotonics Center, Department of Biomedical Engineering, Boston University, 610 Commonwealth Ave., Boston, MA 02215, USA; (B.W.); (J.A.W.)
| | - Anna Junker
- European Institute for Molecular Imaging (EIMI), Westfälische Wilhelms-Universität, Waldeyer-Str. 15, D-48149 Münster, Germany; (K.R.); (A.J.)
| | - Guiscard Seebohm
- Cellular Electrophysiology and Molecular Biology, Department of Cardiovascular Medicine, Institute for Genetics of Heart Diseases (IfGH), University Hospital Münster, Robert-Koch-Str. 45, D-48149 Münster, Germany;
| | - Sven G. Meuth
- Neurology Clinic, University Clinic Düsseldorf, Moorenstraße 5, D-40225 Düsseldorf, Germany;
| | - Petra Hundehege
- Department of Neurology with Institute of Translational Neurology, Albert-Schweitzer-Campus 1, D-48149 Münster, Germany; (L.V.); (P.H.)
| | - Thomas Budde
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
- Correspondence:
| | - Mehrnoush Zobeiri
- Institute of Physiology I, Westfälische Wilhelms-Universität, Robert-Koch-Str. 27a, D-48149 Münster, Germany; (T.O.); (R.C.); (H.-C.P.); (M.Z.)
| |
Collapse
|
45
|
Dehpour AR, Khaledi E, Noori T, Mohammadi-Farani A, Delphi L, Sureda A, Sobarzo-Sanchez E, Shirooie S. Dapsone reduced cuprizone-induced demyelination via targeting Nrf2 and IKB in C57BL/6 mice. IRANIAN JOURNAL OF BASIC MEDICAL SCIENCES 2022; 25:675-682. [PMID: 35949308 PMCID: PMC9320209 DOI: 10.22038/ijbms.2022.64993.14310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Accepted: 05/31/2022] [Indexed: 11/21/2022]
Abstract
Objectives Multiple Sclerosis (MS) is an inflammatory disorder wherein the myelin of nerve cells in the central nervous system is damaged. In the current study, we assessed the effect of Dapsone (DAP) on the improvement of behavioral dysfunction and preservation of myelin in the cuprizone (CPZ) induced demyelination model via targeting Nrf2 and IKB. Materials and Methods MS was induced in C57BL/6 mice through diet supplementation of CPZ (0.2%) for 6 weeks, and DAP (12.5 mg/kg/day; IP) was administered for the last 2 weeks of treatment. Pole test and rotarod performance test, LFB and H&E staining, and Immunohistochemistry (IHC) staining of p-Nrf2 and p-IKB were performed. Furthermore, superoxide dismutase (SOD) and nitrite were measured. Results DAP treatment prevented body loss induced by CPZ (P<0.001). Pole test showed that CPZ increased latency time to fall (P<0.0001) but the latency to reach the floor in the DAP-CPZ group was significantly shorter (P<0.0001). Rotarod performance test showed the effect of CPZ in reducing fall time in the CPZ group (P<0.0014); however, DAP significantly increased fall time (P=0.0012). In LFB staining, DAP reduced demyelination induced by CPZ. CPZ significantly decreased p-Nrf2 and elevated p-IKB levels compared with the control group (P<0.0001), but in DAP-treated groups markedly modified these changes (P<0.0001). CPZ increased the brain nitrite levels and reduced SOD activity, but in DAP-treated considerably reversed CPZ-induced changes. Conclusion These data support the suggestion that the beneficial properties of DAP on the CPZ-induced demyelination are mediated by targeting Nrf2 and NF-kB pathways.
Collapse
Affiliation(s)
- Ahmad Reza Dehpour
- Department of Pharmacology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran,Experimental Medicine Research Center, Tehran University of medical sciences, Tehran, Iran
| | - Ehsan Khaledi
- Student Research Committee, Faculty of Pharmacy, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Tayebeh Noori
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Ahmad Mohammadi-Farani
- Medical Plant Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran,Department of Physiology and Pharmacology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Ladan Delphi
- Animal Biology Department, Faculty of Biology, College of Sciences, University of Tehran, Tehran, Iran
| | - Antoni Sureda
- Research Group on Community Nutrition and Oxidative Stress (NUCOX) and Health Research Institute of Balearic Islands (IdISBa), University of Balearic Islands, Palma de Mallorca E-07122, Balearic Islands, Spain,CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III (ISCIII), 28029 Madrid
| | - Eduardo Sobarzo-Sanchez
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Chile,Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Spain
| | - Samira Shirooie
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran,Corresponding author: Samira Shirooie. Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
46
|
Ramasamy R, Hardy CC, Crocker SJ, Smith PP. Cuprizone-mediated demyelination reversibly degrades voiding behavior in mice while sparing brainstem reflex. J Neurosci Res 2022; 100:1707-1720. [PMID: 35596557 DOI: 10.1002/jnr.25065] [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: 02/02/2022] [Revised: 03/30/2022] [Accepted: 04/22/2022] [Indexed: 11/12/2022]
Abstract
Multiple sclerosis (MS) is a chronic, progressively debilitating demyelinating disease of the central nervous system (CNS). Nearly 80% of MS patients experience lower urinary tract dysfunction early in their diagnosis. This significantly affects the quality of life, and in latter stages of disease is a leading cause of hospitalization. Previously, animal models have shown that inflammatory demyelination in the CNS causes profound bladder dysfunction, but the confounding influence of systemic inflammation limits the potential interpretation of the contribution of CNS demyelination to bladder dysfunction. Since the micturition circuit has myelinated neuronal connections in the cortex, brainstem, and spinal cord, we examined alterations in bladder function in the cuprizone model characterized by demyelinating lesions in the cortex and corpus callosum that are independent of T-cell-mediated autoimmunity. Herein, we report that a 4-week dietary cuprizone treatment in C57Bl/6J mice induced alterations in voiding behavior with increased micturition frequency and reduced volume voided, similar to human MS bladder dysfunction. Subsequently, recovery from cuprizone treatment restored normal bladder function. Demyelination and remyelination were confirmed by Luxol Fast Blue staining of the corpus callosum. Additionally, we also determined that an 8-week cuprizone treatment, resulting in chronic demyelination lacking spontaneous remyelination potential, is associated with an exacerbated voiding phenotype. Interestingly, while cuprizone-induced CNS demyelination severely affected conscious (cortical) urinary behavior, the brainstem and spinal cord reflex remained unchanged, as confirmed by urethane-anesthetized cystometry. This is the first study to show that cortical demyelination independent of inflammation can negatively impact urinary function.
Collapse
Affiliation(s)
- Ramalakshmi Ramasamy
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA.,UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Cara C Hardy
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA.,UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA.,UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| | - Phillip P Smith
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut, USA.,UConn Center on Aging, University of Connecticut School of Medicine, Farmington, Connecticut, USA.,Department of Surgery, University of Connecticut School of Medicine, Farmington, Connecticut, USA
| |
Collapse
|
47
|
Zirngibl M, Assinck P, Sizov A, Caprariello AV, Plemel JR. Oligodendrocyte death and myelin loss in the cuprizone model: an updated overview of the intrinsic and extrinsic causes of cuprizone demyelination. Mol Neurodegener 2022; 17:34. [PMID: 35526004 PMCID: PMC9077942 DOI: 10.1186/s13024-022-00538-8] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 04/08/2022] [Indexed: 12/15/2022] Open
Abstract
The dietary consumption of cuprizone – a copper chelator – has long been known to induce demyelination of specific brain structures and is widely used as model of multiple sclerosis. Despite the extensive use of cuprizone, the mechanism by which it induces demyelination are still unknown. With this review we provide an updated understanding of this model, by showcasing two distinct yet overlapping modes of action for cuprizone-induced demyelination; 1) damage originating from within the oligodendrocyte, caused by mitochondrial dysfunction or reduced myelin protein synthesis. We term this mode of action ‘intrinsic cell damage’. And 2) damage to the oligodendrocyte exerted by inflammatory molecules, brain resident cells, such as oligodendrocytes, astrocytes, and microglia or peripheral immune cells – neutrophils or T-cells. We term this mode of action ‘extrinsic cellular damage’. Lastly, we summarize recent developments in research on different forms of cell death induced by cuprizone, which could add valuable insights into the mechanisms of cuprizone toxicity. With this review we hope to provide a modern understanding of cuprizone-induced demyelination to understand the causes behind the demyelination in MS.
Collapse
Affiliation(s)
- Martin Zirngibl
- Faculty of Medicine & Dentistry, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Peggy Assinck
- Wellcome Trust- MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.,Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK
| | - Anastasia Sizov
- Faculty of Medicine & Dentistry, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada
| | - Andrew V Caprariello
- Department of Clinical Neurosciences, Hotchkiss Brain Institute, University of Calgary, Cumming School of Medicine, Calgary, Canada
| | - Jason R Plemel
- Faculty of Medicine & Dentistry, Neuroscience and Mental Health Institute, University of Alberta, Edmonton, Canada. .,Department of Medical Microbiology and Immunology, University of Alberta, Edmonton, Canada. .,Department of Medicine, Division of Neurology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
48
|
Palavra F, Viana SD, Henriques S, Dinis J, Martins J, Madeira MH, Santiago R, Petrella L, Sereno J, Castelo-Branco M, Pereira FC, Almeida L, Ambrósio AF, Reis F. Defining milestones for the study of remyelination using the cuprizone mouse model: how early is early? Mult Scler Relat Disord 2022; 63:103886. [DOI: 10.1016/j.msard.2022.103886] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 04/06/2022] [Accepted: 05/13/2022] [Indexed: 10/18/2022]
|
49
|
Phenytoin promotes the proliferation of oligodendrocytes and enhances the expression of myelin basic protein in the corpus callosum of mice demyelinated by cuprizone. Exp Brain Res 2022; 240:1617-1627. [PMID: 35362723 DOI: 10.1007/s00221-022-06356-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 03/21/2022] [Indexed: 11/04/2022]
Abstract
Oligodendrocyte loss and myelin sheet destruction are crucial characteristics of demyelinating diseases. Phenytoin promotes the proliferation of endogenous neural precursor cells in the ventricular-subventricular zone in the postnatal brain that help restore the oligodendroglial population. This study aimed to evaluate whether phenytoin promotes myelin recovery of the corpus callosum of demyelinated adult mice. CD1 male mice were exposed to a demyelinating agent (0.2% cuprizone) for 8 weeks. We assembled two groups: the phenytoin-treated group and the control-vehicle group. The treated group received oral phenytoin (10 mg/kg) for 4 weeks. We quantified the number of Olig2 + and NG2 + oligodendrocyte precursor cells (OPCs), Rip + oligodendrocytes, the expression level of myelin basic protein (MBP), and the muscle strength and motor coordination. The oligodendroglial lineage (Olig2 + cells, NG2 + cells, and RIP + cells) significantly increases by the phenytoin administration when compared to the control-vehicle group. The phenytoin-treated group also showed an increased expression of MBP in the corpus callosum and better functional scores in the horizontal bar test. These findings suggest that phenytoin stimulates the proliferation of OPCs, re-establishes the oligodendroglial population, promotes myelin recovery in the corpus callosum, and improves motor coordination and muscle strength.
Collapse
|
50
|
Gharagozloo M, Bannon R, Calabresi PA. Breaking the barriers to remyelination in multiple sclerosis. Curr Opin Pharmacol 2022; 63:102194. [PMID: 35255453 PMCID: PMC8995341 DOI: 10.1016/j.coph.2022.102194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 12/22/2022]
Abstract
Chronically demyelinated axons are rendered susceptible to degeneration through loss of trophic support from oligodendrocytes and myelin, and this process underlies disability progression in multiple sclerosis. Promoting remyelination is a promising neuroprotective therapeutic strategy, but to date, has not been achieved through simply promoting oligodendrocyte precursor cell differentiation, and it is clear that a detailed understanding of the molecular mechanisms underlying failed remyelination is required to guide future therapeutic approaches. In multiple sclerosis, remyelination is impaired by extrinsic inhibitory cues in the lesion microenvironment including secreted effector molecules released from compartmentalized immune cells and reactive glia, as well as by intrinsic defects in oligodendrocyte lineage cells, most notably increased metabolic demands causing oxidative stress and accelerated cellular senescence. Promising advances in our understanding of the cellular and molecular mechanisms underlying these processes offers hope for strategically designed interventions to facilitate remyelination thereby resulting in robust clinical benefits.
Collapse
Affiliation(s)
- Marjan Gharagozloo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Riley Bannon
- 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
| | - 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.
| |
Collapse
|