1
|
Chen G, Zhang K, Sun M, Xie N, Wu L, Zhang G, Guo B, Huang C, Man Hoi MP, Zhang G, Shi C, Sun Y, Zhang Z, Wang Y. Multi-functional memantine nitrate attenuated cognitive impairment in models of vascular dementia and Alzheimer's disease through neuroprotection and increased cerebral blood flow. Neuropharmacology 2025; 272:110410. [PMID: 40081796 DOI: 10.1016/j.neuropharm.2025.110410] [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: 12/07/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Alzheimer's disease (AD) and vascular dementia (VaD) are two prevalent forms of dementia. VaD is linked to cerebrovascular lesions, such as those from white matter ischemia and chronic cerebral hypoperfusion, which can also occur in AD. Nitric oxide (NO) regulates cerebral blood flow (CBF) in the central nervous system. Memantine is an NMDA receptor antagonist approved for AD treatment. This study investigated the efficacy and molecular mechanism of MN-08, a novel memantine nitrate, in one VaD model (2VO) and two AD models (APP/PS1 mice and Aβ1-42-induced mice). MN-08 increased CBF, ameliorated cognitive and memory functions in VaD and AD, and was more effective than memantine. MN-08 increased the survival rate of CA1 neurons and mitigated white matter lesions and axonal damage. Moreover, MN-08 protected neurons from OGD-induced loss and promoted axonal outgrowth in the hippocampus by upregulating phosphorylated Akt (p-Akt), glycogen synthase kinase-3β (p-GSK3β), and high-molecular-weight neurofilaments (p-NFH). The beneficial effects of MN-08 were attenuated by carboxy-PTIO, a potent NO scavenger, suggesting that MN-08-derived NO may alleviate cognitive impairment from cerebral hypoperfusion. Taken together, our studies demonstrate that MN-08 is a promising therapeutic agent for the treatment of dementia including VaD and AD.
Collapse
Affiliation(s)
- Guangying Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Kexin Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Minghua Sun
- Department of Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China; Department of Radiology, The Fuyang Hospital of Anhui Medical University, Fuyang, 236000, China
| | - Ningqing Xie
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Liangmiao Wu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China; Department of Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Guiliang Zhang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao Special Administrative Region, China
| | - Baojian Guo
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Chunhui Huang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Maggie Pui Man Hoi
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao Special Administrative Region, China
| | - Gaoxiao Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| | - Changzheng Shi
- Department of Medical Imaging Center, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China.
| | - Yewei Sun
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China.
| | - Zaijun Zhang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China.
| | - Yuqiang Wang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangzhou Key Laboratory of Innovative Chemical Drug Research in Cardio-cerebrovascular Diseases, and Institute of New Drug Research, Jinan University College of Pharmacy, Guangzhou, 510632, China
| |
Collapse
|
2
|
Magurova M, Bacova M, Papcunova S, Kiss Bimbova K, Kuruc T, Kisucka A, Ihnatova L, Kucharova K, Lukacova N, Galik J. Exploring synergistic effects: Atorvastatin and electrical stimulation in spinal cord injury therapy. IBRO Neurosci Rep 2025; 18:389-399. [PMID: 40124115 PMCID: PMC11927724 DOI: 10.1016/j.ibneur.2025.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/23/2025] [Indexed: 03/25/2025] Open
Abstract
Spinal cord trauma represents a significant clinical challenge, and improving patient outcomes is a main priority for many scientific teams globally. Despite advances in the understanding its pathogenesis, the overall mechanisms occurring in the spinal cord after traumatic injury remain unclear. This study explores the possible synergistic effects of a regenerative therapy that combines electrical stimulation with the anti-inflammatory drug Atorvastatin (ATR) after spinal cord injury (SCI). SCI was induced at the T9 segment under isoflurane anesthesia and applying a compression force of 40 g for 15 minutes. An oscillating field stimulator (OFS) was implanted subcutaneously, delivering a weak electric current (50 µA) that changed polarity every 15 minutes for six weeks to promote axonal growth at the injury site. Female Wistar albino rats were divided into four groups: SCI with non-functional stimulator (SCI + nOFS), SCI with functional stimulator (SCI+OFS), and two groups that received ATR together with stimulator for 7 days after injury (SCI+OFS+ATR, SCI+nOFS+ATR). Behavioral tests (hot-plate test and BBB scale) showed improvement in sensory and motor performance in animals treated with the combination therapy. The protein levels of astrocytes (GFAP), neurofilaments (NF-L), newly sprouting axons (GAP-43), and oligodendrocytes (PLP -1, CNPase) were analysed by Western blot. The results showed increased neurofilaments, newly sprouting axons and oligodendrocytes in groups receiving both individual and combination therapies, with a decrease in their concentrations in the following order: SCI+OFS+ATR, SCI+nOFS+ATR, SCI+OFS, SCI+nOFS. In addition, astrocyte protein levels were lower in the SCI+OFS+ATR group compared with others. Histological analysis showed a significant reduction in white and gray matter after SCI, but less white and gray matter volume loss was found in the groups receiving therapies (SCI+OFS+ATR, SCI+nOFS+ATR, SCI+OFS). These results suggest that the combination of Atorvastatin with OFS stimulation promotes neural recovery after SCI, highlighting the potential of combination therapies in enhancing regenerative outcomes.
Collapse
Affiliation(s)
- Martina Magurova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Maria Bacova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Stefania Papcunova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Katarina Kiss Bimbova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Tomas Kuruc
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Alexandra Kisucka
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Lenka Ihnatova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Karolina Kucharova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Nadezda Lukacova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| | - Jan Galik
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4-6, Kosice 040 01, Slovakia
| |
Collapse
|
3
|
Arrazola R, Espinosa-Jeffrey A, Serafín N, Harmony T, Quirarte GL. Excitotoxic lesion in the corpus callosum of neonatal rats: A model for encephalopathy of prematurity. Neuroscience 2025; 573:198-213. [PMID: 40096962 DOI: 10.1016/j.neuroscience.2025.03.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/19/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Encephalopathy of prematurity (EP) can develop in preterm infants exposed to risk factors like extreme prematurity, low birth weight, hypoxia, infections, and inflammation. These factors can induce excitotoxicity in the brain's gray and white matter, leading to the death of neurons and oligodendrocyte progenitors. Understanding the brain mechanisms of EP requires animal models. In this study, we generated an EP model by injecting N-methyl-D-aspartic acid (NMDA) into the corpus callosum (CC) of neonatal male rats on postnatal day (PND) 5. Rats were divided into five groups: Intact, Vehicle, and three doses of NMDA (3, 4, or 5 μg). On PND 20, we measured the volumes of the CC, motor cortex (MC), and lateral ventricles. The 5 µg NMDA dose caused the largest lesion. We later assessed these structures on PNDs 6, 10, 20, and 30 to monitor lesion progression. We also analyzed myelin basic protein (MBP) expression and counted NeuN-positive cells using immunofluorescent markers. NMDA groups showed reduced MBP expression and fewer NeuN-positive cells in the MC. Additionally, NMDA-treated rats exhibited increased motor activity in the open field and reduced fall latencies in the rotarod task compared to controls. In conclusion, our perinatal excitotoxic lesion model in rats demonstrates structural abnormalities, including decreased MBP and loss of NeuN-positive cells, alongside motor and habituation impairments, resembling those seen in human EP.
Collapse
Affiliation(s)
- Rafael Arrazola
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro. Mexico
| | - Araceli Espinosa-Jeffrey
- Intellectual and Developmental Disabilities Research Center, Jane and Terry Semel Institute for Neuroscience and Human Behavior, Departments of Neurobiology, Psychiatry & Biobehavioral Sciences, University of California, Los Angeles, CA, USA
| | - Norma Serafín
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro. Mexico
| | - Thalía Harmony
- Unidad de Investigación en Neurodesarrollo, Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro, Mexico.
| | - Gina L Quirarte
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Querétaro, Qro. Mexico.
| |
Collapse
|
4
|
Zhu S, Huszar IN, Cottaar M, Daubney G, Eichert N, Hanayik T, Khrapitchev AA, Mars RB, Mollink J, Sallet J, Scott C, Smart A, Jbabdi S, Miller KL, Howard AFD. Imaging the structural connectome with hybrid MRI-microscopy tractography. Med Image Anal 2025; 102:103498. [PMID: 40086183 DOI: 10.1016/j.media.2025.103498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/20/2025] [Accepted: 02/05/2025] [Indexed: 03/16/2025]
Abstract
Mapping how neurons are structurally wired into whole-brain networks can be challenging, particularly in larger brains where 3D microscopy is not available. Multi-modal datasets combining MRI and microscopy provide a solution, where high resolution but 2D microscopy can be complemented by whole-brain but lowresolution MRI. However, there lacks unified approaches to integrate and jointly analyse these multi-modal data in an insightful way. To address this gap, we introduce a data-fusion method for hybrid MRI-microscopy fibre orientation and connectome reconstruction. Specifically, we complement precise "in-plane" orientations from microscopy with "through-plane" information from MRI to construct 3D hybrid fibre orientations at resolutions far exceeding that of MRI whilst preserving microscopy's myelin specificity, resulting in superior fibre tracking. Our method is openly available, can be deployed on standard 2D microscopy, including different microscopy contrasts, and is species agnostic, facilitating neuroanatomical investigation in both animal models and human brains.
Collapse
Affiliation(s)
- Silei Zhu
- Wellcome Centre for Integrative Neuroimaging, FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom.
| | - Istvan N Huszar
- Wellcome Centre for Integrative Neuroimaging, FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Michiel Cottaar
- Wellcome Centre for Integrative Neuroimaging, FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Greg Daubney
- Wellcome Centre for Integrative Neuroimaging, Experimental Psychology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Nicole Eichert
- Wellcome Centre for Integrative Neuroimaging, FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Taylor Hanayik
- Wellcome Centre for Integrative Neuroimaging, FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | | | - Rogier B Mars
- Wellcome Centre for Integrative Neuroimaging, FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Donders Institute for Brain, Cognition and Behaviour, Radboud University Nijmegen, Nijmegen, the Netherlands
| | - Jeroen Mollink
- Wellcome Centre for Integrative Neuroimaging, Experimental Psychology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Jerome Sallet
- Wellcome Centre for Integrative Neuroimaging, Experimental Psychology, Medical Sciences Division, University of Oxford, Oxford, United Kingdom; INSERM U1208, Stem Cell and Brain Research Institute, University Lyon, Bron, France
| | - Connor Scott
- Division of Clinical Neurology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Adele Smart
- Wellcome Centre for Integrative Neuroimaging, FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Division of Clinical Neurology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Saad Jbabdi
- Wellcome Centre for Integrative Neuroimaging, FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Karla L Miller
- Wellcome Centre for Integrative Neuroimaging, FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom
| | - Amy F D Howard
- Wellcome Centre for Integrative Neuroimaging, FMRIB Centre, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, United Kingdom; Department of Bioengineering, Imperial College London, London, United Kingdom
| |
Collapse
|
5
|
Okutan B, Frederiksen JL, Houen G, Sellebjerg F, Kyllesbech C, Magyari M, Paunovic M, Sørensen PS, Jacobsen C, Lassmann H, Bramow S. Subcortical plaques and inflammation reflect cortical and meningeal pathologies in progressive multiple sclerosis. Brain Pathol 2025; 35:e13314. [PMID: 39460678 PMCID: PMC11961212 DOI: 10.1111/bpa.13314] [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: 05/13/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
It remains elusive whether lesions and inflammation in the sub/juxtacortical white matter reflect cortical and/or meningeal pathologies. Elucidating this could have implications for MRI monitoring as sub/juxtacortical lesions are detectable by routine MRI, while cortical lesions and meningeal inflammation are not. By large-area microscopy, we quantified total and mixed active plaque loads along with densities and sizes of perivascular mononuclear infiltrates (infiltrates) in the sub/juxtacortical white matter ≤2 mm from the cortex, intra-cortically and in the meninges. Data were related to ante-mortem clinical parameters in a false discovery rate-corrected analysis. We compared 12 patients with primary progressive multiple sclerosis (PPMS) and 15 with secondary progressive MS to 22 controls. Fifteen patients and 11 controls contributed with hemispheric sections. Sections were stained with haematoxylin-eosin, for myelin and for microglia/macrophages. B cells and T cells were confirmed in a subset. Immunoglobulin G depositions in selected cortical plaques resembled depositions described before in "slowly expanding" plaques in the white matter. We quantified plaque activity by measuring microglia-dominated and macrophage-dominated areas. Sub/juxtacortical plaques (load and activity) reflected plaque activity in the cerebral cortex. Plaque activity and infiltrates were more pronounced in the sub/juxtacortical white matter than in the cerebral cortex while conversely, the total plaque load was highest in the cortex. Infiltrates correlated trans-cortically and sub/juxtacortical plaque activity reflected cortical and meningeal infiltrates. Sub/juxtacortical infiltrate sizes correlated with shorter survival after progression onset. Two patients with PPMS and putatively fatal brain stem lesions argue against incidental findings. Trans-cortical inflammatory flares and plaque activity may be pathogenic in progressive MS. We suggest emphasis on sub/juxtacortical MRI lesions as plausible surrogates for cortical and meningeal pathologies and, when present, as indicators for cognitive testing.
Collapse
Affiliation(s)
- Betül Okutan
- Department of Neurology, Danish Multiple Sclerosis CenterCopenhagen University Hospital – RigshospitaletGlostrupDenmark
| | - Jette L. Frederiksen
- Department of Neurology, Danish Multiple Sclerosis CenterCopenhagen University Hospital – RigshospitaletGlostrupDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Gunnar Houen
- Department of Neurology, Danish Multiple Sclerosis CenterCopenhagen University Hospital – RigshospitaletGlostrupDenmark
- Department of Biochemistry and Molecular Biology, Faculty of Health SciencesUniversity of Southern DenmarkOdenseDenmark
| | - Finn Sellebjerg
- Department of Neurology, Danish Multiple Sclerosis CenterCopenhagen University Hospital – RigshospitaletGlostrupDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Cecilie Kyllesbech
- Department of Neurology, Danish Multiple Sclerosis CenterCopenhagen University Hospital – RigshospitaletGlostrupDenmark
- Department of Biochemistry and Molecular Biology, Faculty of Health SciencesUniversity of Southern DenmarkOdenseDenmark
| | - Melinda Magyari
- Department of Neurology, Danish Multiple Sclerosis CenterCopenhagen University Hospital – RigshospitaletGlostrupDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Neurology, Danish Multiple Sclerosis RegistryCopenhagen University Hospital – RigshospitaletGlostrupDenmark
| | - Manuela Paunovic
- Department of NeurologyErasmus Medical CentreRotterdamThe Netherlands
- Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Per S. Sørensen
- Department of Neurology, Danish Multiple Sclerosis CenterCopenhagen University Hospital – RigshospitaletGlostrupDenmark
- Department of Clinical Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Christina Jacobsen
- Section of Forensic Pathology, Department of Forensic Medicine, Faculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Hans Lassmann
- Center for Brain ResearchMedical University of ViennaViennaAustria
| | - Stephan Bramow
- Department of Neurology, Danish Multiple Sclerosis CenterCopenhagen University Hospital – RigshospitaletGlostrupDenmark
- Department of PathologyCopenhagen University Hospital – RigshospitaletCopenhagenDenmark
| |
Collapse
|
6
|
Donkels C, Häussler U, Huber S, Tiesmeyer N, Demerath T, Scheiwe C, Shah MJ, Heers M, Urbach H, Schulze‐Bonhage A, Prinz M, Vlachos A, Beck J, Nakagawa JM, Haas CA. Dysregulation of Myelination in Focal Cortical Dysplasia Type II of the Human Frontal Lobe. Glia 2025; 73:928-947. [PMID: 39719691 PMCID: PMC11920682 DOI: 10.1002/glia.24662] [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: 06/28/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 12/26/2024]
Abstract
Focal cortical dysplasias (FCDs) are local malformations of the human neocortex and a leading cause of intractable epilepsy. FCDs are classified into different subtypes including FCD IIa and IIb, characterized by a blurred gray-white matter boundary or a transmantle sign indicating abnormal white matter myelination. Recently, we have shown that myelination is also compromised in the gray matter of FCD IIa of the temporal lobe. Since myelination is key for brain function, which is imbalanced in epilepsy, in the current study, we investigated myelination in the gray matter of FCD IIa and IIb from the frontal lobe on the morphological, ultrastructural, and transcriptional level. We found that FCD IIa presents with an ordinary radial myelin fiber pattern, but with a reduced thickness of myelin sheaths of 500-1000 nm thick axons in comparison to FCD IIb and with an attenuation of the myelin synthesis machinery. In contrast, FCD IIb showed an irregular and disorganized myelination pattern covering an enlarged area in comparison to FCD IIa and controls and with increased numbers of myelinating oligodendrocytes (OLs). FCD IIb had significantly thicker myelin sheaths of large caliber axons (above 1000 nm) when compared to FCD IIa. Accordingly, FCD IIb showed a significant up-regulation of myelin-associated mRNAs in comparison to FCD IIa and enhanced binding capacities of the transcription factor MYRF to target sites in myelin-associated genes. These data indicate that FCD IIa and IIb are characterized by a differential dysregulation of myelination in the gray matter of the frontal lobe.
Collapse
Affiliation(s)
- Catharina Donkels
- Faculty of Medicine, Experimental Epilepsy Research, Department of NeurosurgeryMedical Center ‐ University of FreiburgFreiburgGermany
| | - Ute Häussler
- Faculty of Medicine, Experimental Epilepsy Research, Department of NeurosurgeryMedical Center ‐ University of FreiburgFreiburgGermany
- Faculty of Medicine, Translational Epilepsy Research, Department of NeurosurgeryMedical Center ‐ University of FreiburgFreiburgGermany
- BrainLinks‐BrainTools CenterUniversity of FreiburgFreiburgGermany
| | - Susanne Huber
- Faculty of Medicine, Experimental Epilepsy Research, Department of NeurosurgeryMedical Center ‐ University of FreiburgFreiburgGermany
| | - Nina Tiesmeyer
- Faculty of Medicine, Experimental Epilepsy Research, Department of NeurosurgeryMedical Center ‐ University of FreiburgFreiburgGermany
| | - Theo Demerath
- Faculty of Medicine, Department of NeuroradiologyMedical Center‐University of FreiburgFreiburgGermany
| | - Christian Scheiwe
- Faculty of Medicine, Department of NeurosurgeryMedical Center ‐ University of FreiburgFreiburgGermany
| | - Mukesch J. Shah
- Faculty of Medicine, Department of NeurosurgeryMedical Center ‐ University of FreiburgFreiburgGermany
| | - Marcel Heers
- Faculty of Medicine, Epilepsy Center FreiburgMedical Center ‐ University of FreiburgFreiburgGermany
| | - Horst Urbach
- Faculty of Medicine, Department of NeuroradiologyMedical Center‐University of FreiburgFreiburgGermany
| | - Andreas Schulze‐Bonhage
- BrainLinks‐BrainTools CenterUniversity of FreiburgFreiburgGermany
- Faculty of Medicine, Epilepsy Center FreiburgMedical Center ‐ University of FreiburgFreiburgGermany
| | - Marco Prinz
- Faculty of Medicine, Institute of NeuropathologyMedical Center ‐ University of FreiburgFreiburgGermany
- Signalling Research Centers BIOSS and CIBSSUniversity of FreiburgFreiburgGermany
| | - Andreas Vlachos
- BrainLinks‐BrainTools CenterUniversity of FreiburgFreiburgGermany
- Department of Neuroanatomy, Institute of Anatomy and Cell Biology, Faculty of MedicineUniversity of FreiburgFreiburgGermany
| | - Jürgen Beck
- Faculty of Medicine, Department of NeurosurgeryMedical Center ‐ University of FreiburgFreiburgGermany
| | - Julia M. Nakagawa
- Faculty of Medicine, Translational Epilepsy Research, Department of NeurosurgeryMedical Center ‐ University of FreiburgFreiburgGermany
- Faculty of Medicine, Department of NeurosurgeryMedical Center ‐ University of FreiburgFreiburgGermany
| | - Carola A. Haas
- Faculty of Medicine, Experimental Epilepsy Research, Department of NeurosurgeryMedical Center ‐ University of FreiburgFreiburgGermany
- BrainLinks‐BrainTools CenterUniversity of FreiburgFreiburgGermany
| |
Collapse
|
7
|
Dubois J, Grotheer M, Yang JYM, Tournier JD, Beaulieu C, Lebel C. Small brains but big challenges: white matter tractography in early life samples. Brain Struct Funct 2025; 230:58. [PMID: 40293528 DOI: 10.1007/s00429-025-02922-8] [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: 04/10/2025] [Accepted: 04/19/2025] [Indexed: 04/30/2025]
Abstract
In the human brain, white matter development is a complex and long-lasting process involving intermingling micro- and macrostructural mechanisms, such as fiber growth, pruning and myelination. Did you know that all these neurodevelopmental changes strongly affect MRI signals, with consequences on tractography performances and reliability? This communication aims to elaborate on these aspects, highlighting the importance of tracking and studying the developing connections with dedicated approaches.
Collapse
Affiliation(s)
- Jessica Dubois
- Université Paris Cité, Inserm, Paris, F-75019, NeuroDiderot, France.
- Université Paris-Saclay, CEA, UNIACT, Gif-sur-Yvette, F-91191, NeuroSpin, France.
| | - Mareike Grotheer
- Department of Psychology, Phillips-Universität Marburg, 35039, Marburg, Germany
- Center for Mind, Brain and Behavior, Phillips-Universität Marburg, Justus-Liebig Universität Giessen, Technische Universität Darmstadt, 35039, Marburg, Germany
| | - Joseph Yuan-Mou Yang
- Department of Neurosurgery, Neuroscience Advanced Clinical Imaging Service (NACIS), The Royal Children's Hospital, Melbourne, Australia
- Neuroscience Research, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, University of Melbourne, Melbourne, Australia
| | - Jacques-Donald Tournier
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
- Centre for the Developing Brain, School of Biomedical Engineering and Imaging Sciences, King's College London, King's Health Partners, St. Thomas' Hospital, London, UK
| | - Christian Beaulieu
- Departments of Radiology and Diagnostic Imaging & Biomedical Engineering, University of Alberta, Edmonton, AB, Canada
| | - Catherine Lebel
- Department of Radiology, University of Calgary, Calgary, Canada
| |
Collapse
|
8
|
Zou Y, Jin Y, Yang Y, Zhang L, Feng Y, Long Y, Xu Z, He Y, Zheng W, Wang S, He Y, Li J, Li H, Luo Z, Hu C, Xiao L. Effect of Cytoskeletal Linker Protein GAS2L1 on Oligodendrocyte and Myelin Development. Glia 2025; 73:840-856. [PMID: 39743758 DOI: 10.1002/glia.24658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 11/25/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025]
Abstract
Oligodendrocytes (OLs), the myelin-forming cells of the central nervous system (CNS), develop from OL precursor cells (OPCs) through a complex process involving significant morphological changes that are critically dependent on the dynamic interactions between cytoskeletal networks. Growth arrest-specific 2-like protein 1 (GAS2L1) is a cytoskeletal linker protein that mediates the cross-talk between actin filaments and microtubules. However, its role in OL and myelin development remains unknown. Here, we report that GAS2L1 is expressed in both OPCs and mature OLs, and that overexpression or knockdown of Gas2l1 in cultured OPCs in vitro impaired or enhanced their differentiation, respectively, while both inhibited their proliferation. We generated a Gas2l1 fl/fl mouse line and found that mice with conditional knockout of Gas2l1 in OL lineage cells (Olig1-Cre;Gas2l1 fl/fl , cKO) showed an increased number of mature OLs and enhanced myelination, as well as a reduction in the branching complexity of OPCs. In addition, an alternative mouse line with postnatally induced Gas2l1 ablation specifically in OPCs (Pdfgra-CreER T2 ;Gas2l1 fl/fl , iKO) recapitulated the acceleration of OL and myelin development as well as the inhibition of OPC process branching. Furthermore, EdU tracking in Gas2l1 iKO mice in vivo and in their OPC cultures in vitro showed both a reduction in OPC proliferation and an increase in OL maturation. Finally, cultured OPCs from iKO mice showed an increase in filopodia extension. Taken together, our results demonstrate an effect of GAS2L1 on the regulation of OL/myelin development and may provide a novel potential therapeutic target for various diseases involving OL/myelin pathology.
Collapse
Affiliation(s)
- 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, China
| | - Yili Jin
- 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, China
| | - Yuqian Yang
- 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, 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, 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, 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, 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, China
| | - 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, 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, 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, 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, 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, China
| | - Huiliang Li
- Wolfson Institute for Biomedical Research, University College London, London, UK
| | - Zhigang Luo
- Department of Experimental Medicine, The Third People's Hospital of Sichuan Province, Chengdu, Sichuan, China
| | - Chun Hu
- 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, 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, China
| |
Collapse
|
9
|
Horvat S, Fonović UP, Mitrović A, Zidar N, Kos J, Pišlar A. The α- to γ-enolase switch: The role and regulation of γ-enolase during oligodendrocyte differentiation. Int J Biol Macromol 2025; 301:140464. [PMID: 39884600 DOI: 10.1016/j.ijbiomac.2025.140464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/11/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
The glycolytic enzyme γ-enolase is a highly specific neuronal marker that is known to replace ubiquitously expressed α-enolase in the brain. Moreover, γ-enolase has been shown to exert neurotrophic activity, which is regulated by cathepsin X, a lysosomal peptidase. This study investigates the role of γ-enolase and its regulation by cathepsin X during the differentiation of oligodendrocytes, which are essential for normal brain function. We established a differentiation protocol for the human oligodendroglioma (HOG) cell line and demonstrated for the first time that an α- to γ-enolase switch occurs during HOG cell differentiation. This switch was confirmed by the expression of specific markers underscoring the role of γ-enolase in oligodendrocyte differentiation. Moreover, γ-enolase overexpression enhanced oligodendrocyte differentiation, while silencing of γ-enolase by siRNA significantly decreased maturation marker. Further, the regulatory role of cysteine peptidase cathepsin X on γ-enolase function was found. Silencing cathepsin X significantly changed cell morphology, enhanced oligodendrocyte differentiation, altered the expression of oligodendrocyte markers, and increased levels of the active form of γ-enolase. Inhibiting cathepsin X similarly changed cell morphology and enhanced oligodendrocyte differentiation. These findings suggest that cathepsin X modulates γ-enolase activity and thereby influences oligodendrocyte differentiation and thus neuronal function.
Collapse
Affiliation(s)
- Selena Horvat
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| | - Urša Pečar Fonović
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| | - Ana Mitrović
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia; Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia.
| | - Nace Zidar
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| | - Janko Kos
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia; Department of Biotechnology, Jožef Stefan Institute, Jamova 39, 1000 Ljubljana, Slovenia.
| | - Anja Pišlar
- Department of Pharmaceutical Biology, Faculty of Pharmacy, University of Ljubljana, Aškerčeva 7, 1000 Ljubljana, Slovenia.
| |
Collapse
|
10
|
Zacher AC, Grabinski M, Console-Meyer L, Felmy F, Pätz-Warncke C. Oligodendrocyte arrangement, identification and morphology in the developing superior olivary complex. Front Cell Neurosci 2025; 19:1561312. [PMID: 40226299 PMCID: PMC11985757 DOI: 10.3389/fncel.2025.1561312] [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: 01/15/2025] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Oligodendrocytes provide myelination, metabolic and developmental support for neurons and circuits. Within the auditory superior olivary complex (SOC), relevant for sound localization and spectro-temporal integration, oligodendrocytes are fundamental for fast neuronal communication and accurate timing of sound signals. Despite their important role in function and development, an assessment of their developmental arrangement and morphology is missing for the SOC. Here, immunofluorescence labeling and single cell electroporation was used to quantify their distribution, identification and morphology between postnatal day (P) 5 and ~ P54 in the SOC of Mongolian gerbils (Meriones unguiculatus). Oligodendrocytes show developmental, region-specific accumulations, redistributions and density profiles. Their identification by Olig2 and SOX10 appears age specific, while myelinating oligodendrocytes are detected by co-labeling with S100 irrespective of age. Comparison of oligodendrocyte density and identification between mature gerbil and Etruscan shrew (Suncus etruscus), revealed species-specific differences. Morphologically, the number of myelinating processes decreased, while process length, diameter and coverage area of oligodendrocytes increased during development. Together, oligodendrocyte developmental alterations occur at moments of SOC circuit refinement supporting functions beyond myelination.
Collapse
Affiliation(s)
- Alina Carola Zacher
- Institute for Zoology, University of Veterinary Medicine Foundation, Hannover, Germany
- Hannover Graduate School for Neurosciences, Infection Medicine and Veterinary Sciences (HGNI), Hannover, Germany
| | - Melissa Grabinski
- Institute for Zoology, University of Veterinary Medicine Foundation, Hannover, Germany
| | - Laura Console-Meyer
- Institute for Zoology, University of Veterinary Medicine Foundation, Hannover, Germany
- Hannover Graduate School for Neurosciences, Infection Medicine and Veterinary Sciences (HGNI), Hannover, Germany
| | - Felix Felmy
- Institute for Zoology, University of Veterinary Medicine Foundation, Hannover, Germany
| | | |
Collapse
|
11
|
Gong J, Li J, Li J, He A, Ren B, Zhao M, Li K, Zhang Y, He M, Liu Y, Wang Z. Impact of Microglia-Derived Extracellular Vesicles on Resident Central Nervous System Cell Populations After Acute Brain Injury Under Various External Stimuli Conditions. Mol Neurobiol 2025:10.1007/s12035-025-04858-w. [PMID: 40126599 DOI: 10.1007/s12035-025-04858-w] [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: 11/19/2024] [Accepted: 03/14/2025] [Indexed: 03/25/2025]
Abstract
Acute brain injuries (ABI) caused by various emergencies can lead to structural and functional damage to brain tissue. Common causes include traumatic brain injury, cerebral hemorrhage, ischemic stroke, and heat stroke. Globally, ABI represent a significant portion of neurosurgical cases. Previous studies have emphasized the significant therapeutic potential of stem cell-derived extracellular vesicles (EVs). Recent research indicates that EVs extracted from resident cells in the central nervous system (CNS) also show therapeutic potential following brain injury. Microglia, as innate immune cells of the CNS, respond to changes in the internal environment by altering their phenotype and secreting EVs that impact various CNS cells, including neurons, astrocytes, oligodendrocytes, endothelial cells, neural stem cells (NSCs), and microglia themselves. Notably, under different external stimuli, microglia can either promote neuronal survival, angiogenesis, and myelin regeneration while reducing glial scarring and inflammation, or they can exert opposite effects. This review summarizes and evaluates the current research findings on how microglia-derived EVs influence various CNS cells after ABI under different external stimuli. It analyzes the interaction mechanisms between EVs and resident CNS cells and discusses potential future research directions and clinical applications.
Collapse
Affiliation(s)
- Junjie Gong
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Jing Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Jian Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Anqi He
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, Tianjin, China
| | - Bingcheng Ren
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Mingyu Zhao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Kexin Li
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Yuchi Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Mengyao He
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China
| | - Yuheng Liu
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China.
| | - Zengguang Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China.
- Ministry of Education and Tianjin, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Tianjin, China.
| |
Collapse
|
12
|
Kobayashi S, Yokoi T, Omata T, Yako H, Miyamoto Y, Yamauchi J. Claudin-11, a hypomyelinating leukodystrophy 22 (HLD22)-responsible protein, uniquely interacts with shroom-2 to change cell phenotypes. BBA ADVANCES 2025; 7:100159. [PMID: 40230506 PMCID: PMC11995805 DOI: 10.1016/j.bbadva.2025.100159] [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: 12/24/2024] [Revised: 03/11/2025] [Accepted: 03/24/2025] [Indexed: 04/16/2025] Open
Abstract
Oligodendroglial cells are a type of glial cell in the central nervous system (CNS) that wrap neuronal axons with differentiated plasma membranes known as myelin sheaths. While the physiological functions of oligodendrocytes, such as generating saltatory conduction and protecting neuronal axons, are well understood, the physiological and/or pathophysiological molecular mechanisms governing their differentiation before myelination remain unclear. In this study, we describe for the first time that claudin-11, a protein associated with hypomyelinating leukodystrophy 22 (HLD22), interacts with shroom-2, a presumable adaptor protein containing the PSD95, DLG1, and ZO-1 (PDZ) domain. Knockdown of claudin-11 using specific siRNA resulted in a decrease in morphological changes and marker proteins in the FBD-102b oligodendroglial model undergoing differentiation. Transfection of the C-terminal PDZ ligand sequence of claudin-11, which was found to interact with the PDZ domain of shroom-2, also reduced these phenotypic changes. The HLD22-associated mutated sequence in claudin-11 failed to interact with the PDZ domain of shroom-2. Furthermore, knockdown of shroom-2 or transfection of the PDZ domain of shroom-2, which is involved in the interaction with claudin-11, resulted in decreased morphological changes and marker protein expression. These changes were linked to the phosphorylation states of Akt kinase, a key signaling molecule in oligodendroglial cell differentiation and myelination. These results suggest that the interaction between claudin-11 and shroom-2 plays a key role in shaping cell morphology, providing insights into the molecular mechanisms underlying oligodendroglial differentiation before myelination, as well as potential pathological mechanisms associated with HLD22 at the molecular and cellular levels.
Collapse
Affiliation(s)
- Sakurako Kobayashi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Takanori Yokoi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Takeru Omata
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Hideji Yako
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Yuki Miyamoto
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| |
Collapse
|
13
|
Jana R, Das Sarma J. The crosstalk between CNS resident glial cells and peripheral immune cells is critical for age-dependent demyelination and subsequent remyelination. Biogerontology 2025; 26:74. [PMID: 40085264 DOI: 10.1007/s10522-025-10213-2] [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: 12/30/2024] [Accepted: 03/01/2025] [Indexed: 03/16/2025]
Abstract
White-matter diseases like multiple sclerosis begin in young adulthood. Aging, being a risk factor, contributes to the progression of these diseases and makes neurological disabilities worsen. Aging causes white matter alteration due to myelin loss, axonal degeneration, and hyperintensities, resulting in cognitive impairment and neurological disorders. Aging also negatively affects central nervous system resident glial cells and peripheral immune cells, contributing to myelin degeneration and diminished myelin renewal process. Restoration of myelin failure with aging accelerates the progression of cognitive decline. This review will mainly focus on how age-related altered functions of glial and peripheral cells will affect myelin sheath alteration and myelin restoration. This understanding can give us insights into the underlying mechanisms of demyelination and failure of remyelination with aging concerning altered glial and peripheral immune cell function and their crosstalk. Also, we will explain the therapeutic strategies to enhance the remyelination process of an aging brain to improve the cognitive health of an aging person.
Collapse
Affiliation(s)
- Rishika Jana
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, India.
- Departments of Ophthalmology, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| |
Collapse
|
14
|
Balantzategi U, Gaminde-Blasco A, Kearns CA, Bayón-Cordero L, Sánchez-Gómez MV, Zugaza JL, Appel B, Alberdi E. Amyloid-β Dysregulates Oligodendroglial Lineage Cell Dynamics and Myelination via PKC in the Zebrafish Spinal Cord. Glia 2025. [PMID: 40087862 DOI: 10.1002/glia.70015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025]
Abstract
Soluble forms of amyloid-β (Aβ) peptide have been proposed as candidates to induce oligodendrocyte (OL) and myelin dysfunctions in the early stages of Alzheimer's disease (AD) pathology. Nevertheless, little is known about how Aβ affects OL differentiation and myelination in vivo, and the underlying molecular mechanisms. In this study, we explored the effects of a brain intraventricular injection of Aβ on OLs and myelin in the developing spinal cord of zebrafish larvae. Using quantitative fluorescent in situ RNA hybridization assays, we demonstrated that Aβ altered myrf and mbp mRNA levels and the regional distribution of mbp during larval development, suggesting an early differentiation of OLs. Through live imaging of Tg(myrf:mScarlet) and Tg(mbpa:tagRFP) zebrafish lines, both crossed with Tg(olig2:EGFP), we found that Aβ increased the number of myrf+ and mbp+ OLs in the dorsal spinal cord at 72 hpf and 5 dpf, respectively, without affecting total cell numbers. Furthermore, Aβ also increased the number of Sox10+cells, myelin sheaths per OL, and the number of myelinated axons in the dorsal spinal cord at 8 dpf compared to vehicle-injected control animals. Interestingly, the treatment of Aβ-injected zebrafish with the pan-PKC inhibitor Gö6983 restored the aforementioned alterations in OLs and myelin to control levels. Altogether, not only do we demonstrate that Aβ induces a precocious oligodendroglial differentiation leading to dysregulated myelination, but we also identified PKC as a key player in Aβ-induced pathology.
Collapse
Affiliation(s)
- Uxue Balantzategi
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Adhara Gaminde-Blasco
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - Christina A Kearns
- Department of Pediatrics, Section of Developmental Biology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Laura Bayón-Cordero
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - María Victoria Sánchez-Gómez
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| | - José Luis Zugaza
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (UPV/EHU), Leioa, Spain
- IKERBASQUE Basque Foundation for Science, Bilbao, Spain
| | - Bruce Appel
- Department of Pediatrics, Section of Developmental Biology, University of Colorado-Anschutz Medical Campus, Aurora, Colorado, USA
| | - Elena Alberdi
- Department of Neurosciences, University of the Basque Country (UPV/EHU), Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
| |
Collapse
|
15
|
López-Muguruza E, Peiró-Moreno C, Pérez-Cerdá F, Matute C, Ruiz A. Del Río Hortega's insights into oligodendrocytes: recent advances in subtype characterization and functional roles in axonal support and disease. Front Neuroanat 2025; 19:1557214. [PMID: 40145026 PMCID: PMC11936973 DOI: 10.3389/fnana.2025.1557214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
Pío Del Río Hortega (1882-1945) was a giant of modern neuroscience and perhaps the most impactful member of Cajal's School. His contributions to clarifying the structure of the nervous system were key to understanding the brain beyond neurons. He uncovered microglia and oligodendrocytes, the latter until then named mesoglia. Most importantly, the characterization of oligodendroglia subtypes he made has stood the omics revolution that added molecular details relevant to comprehend their biological properties. Astounding as it may seem on today's eyes, he postulated a century ago that oligodendrocytes provide trophic support to axons, an idea that is now beyond doubt and under scrutiny as dysfunction at the axon-myelin unit is key to neurodegeneration. Here, we revised recent key advancements in oligodendrocyte biology that shed light on Hortega's ideas a century ago.
Collapse
Affiliation(s)
- Eneritz López-Muguruza
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIBERNED-Instituto de Salud Carlos III, Leioa, Spain
| | - Carla Peiró-Moreno
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- Achucarro Basque Center for Neuroscience, Leioa, Spain
- CIBERNED-Instituto de Salud Carlos III, Leioa, Spain
| | - Fernando Pérez-Cerdá
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- CIBERNED-Instituto de Salud Carlos III, Leioa, Spain
- Department of Neurosciences, Biobizkaia, Barakaldo, Spain
| | - Carlos Matute
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- CIBERNED-Instituto de Salud Carlos III, Leioa, Spain
- Department of Neurosciences, Biobizkaia, Barakaldo, Spain
| | - Asier Ruiz
- Department of Neurosciences, University of the Basque Country UPV/EHU, Leioa, Spain
- CIBERNED-Instituto de Salud Carlos III, Leioa, Spain
- Department of Neurosciences, Biobizkaia, Barakaldo, Spain
| |
Collapse
|
16
|
Lin ML, Lin W. Thinning of originally-existing, mature myelin represents a nondestructive form of myelin loss in the adult CNS. Front Cell Neurosci 2025; 19:1565913. [PMID: 40134707 PMCID: PMC11933062 DOI: 10.3389/fncel.2025.1565913] [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: 01/23/2025] [Accepted: 02/28/2025] [Indexed: 03/27/2025] Open
Abstract
The main function of oligodendrocytes is to assemble and maintain myelin that wraps and insulates axons in the central nervous system (CNS). Traditionally, myelin structure, particularly its thickness, was believed to remain remarkably stable in adulthood (including early and middle adulthood, but not late adulthood or aging). However, emerging evidence reveals that the thickness of originally-existing, mature myelin (OEM) can undergo dynamic changes in the adult CNS. This overview highlights recent findings on the alteration of OEM thickness in the adult CNS, explores the underlying mechanisms, and proposes that progressive thinning of OEM represents a novel, nondestructive form of myelin loss in myelin disorders of the CNS.
Collapse
Affiliation(s)
- Min Li Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
17
|
Feng Y, Guo M, You T, Zhang M, Li J, Xie J, Han S, Zhao H, Jiang Y, Zhao Y, Yu J, Dong Q, Cui M. Paranodal instability driven by axonal mitochondrial accumulation in ischemic demyelination and cognitive decline. Mol Psychiatry 2025:10.1038/s41380-025-02936-y. [PMID: 40033045 DOI: 10.1038/s41380-025-02936-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 01/20/2025] [Accepted: 02/14/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Subcortical ischemic demyelination is the primary cause of vascular cognitive impairment in the elderly. However, its underlying mechanisms remain elusive. METHODS Using a bilateral common carotid artery stenosis (BACS) mouse model and an in vitro cerebellar slice model treated with low glucose-low oxygen (LGLO), we investigated a novel mechanism of vascular demyelination. RESULTS This work identified syntaphilin-mediated docking of mitochondria as the initial event preceding ischemic demyelination. This axonal insult drives paranodal retraction, myelin instability, and subsequent cognitive impairment through excessive oxidation of protein 4.1B by mitochondrial ROS. Syntaphilin knockdown reestablished the balance of mitochondrial axoplasmic transport, reduced axonal ROS burden, and consequently decreased the abnormal oxidation of protein 4.1B, an essential component that secures the Caspr1/contactin-1/NF155 complex tethered to the axonal cytoskeleton βII-Spectrin within paranodes. This ultimately protected the paranodal structure and myelin and improved cognitive function. CONCLUSIONS Our findings reveal a distinct pathological characteristic of ischemic demyelination and highlight the therapeutic potential of modulating axonal mitochondrial mobility to stabilize myelin structures and improve vascular cognitive impairment.
Collapse
Affiliation(s)
- Yiwei Feng
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Min Guo
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Tongyao You
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Minjie Zhang
- Department of Neurology, The 10th People's Hospital, Tongji University, Shanghai, China
| | - Jincheng Li
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China
| | - Junchao Xie
- Department of Neurology, The 10th People's Hospital, Tongji University, Shanghai, China
| | - Sida Han
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Hongchen Zhao
- Department of Neurology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yanfeng Jiang
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China
| | - Yanxin Zhao
- Department of Neurology, The 10th People's Hospital, Tongji University, Shanghai, China.
| | - Jintai Yu
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Qiang Dong
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| | - Mei Cui
- Department of Neurology, Huashan Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China.
| |
Collapse
|
18
|
Naffaa V, Van Steenwinckel J, Magny R, Regazzetti A, Keime C, Gressens P, Laprévote O, Auzeil N, Schang AL. Bisphenol A exposure during gestation and lactation in mice: Sex-specific consequences on oligodendrocytes and myelination. Toxicology 2025; 512:154041. [PMID: 39742910 DOI: 10.1016/j.tox.2024.154041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/11/2024] [Accepted: 12/27/2024] [Indexed: 01/04/2025]
Abstract
Bisphenol A (BPA), a ubiquitous environmental endocrine disruptor, is suspected of disturbing brain development through largely unknown cellular and molecular mechanisms. In the central nervous system, oligodendrocytes are responsible for forming myelin sheaths, which enhance the propagation of action potentials along axons. Disruption of axon myelination can have lifelong consequences, making oligodendrocyte differentiation and myelination critical stages of brain development. In the present study, mice were exposed to BPA during gestation and lactation through drinking water at concentrations of 25 and 250 μg.L-1. These doses, corresponding to estimated exposures of 4 μg.kg-1.d-1 and 40 μg.kg-1.d-1, respectively, led to disturbances in lipid remodeling associated with myelination in the offspring. Importantly, changes in myelin lipid composition were selectively observed in female mice and were transient, being visible only at post-natal day P15 but not at later stages (P30 and P60). In females exposed to BPA, myelin exhibited a lower proportion of phosphatidylcholines and higher proportions of other glycerophospholipid subclasses, thus resembling more mature myelin. Conversely, male myelin was not affected, likely due to its already more mature lipid composition. Additionally, transcriptomic analysis of female oligodendrocytes at P15 did not reveal any transcriptional changes in genes related to lipid metabolism, further suggesting post-transcriptional effects of BPA via chaperone-mediated protein folding and RNA splicing. In males, the altered genes were mainly associated with synaptic transmission. Finally, alterations in chromatin accessibility were also largely sex dependent and did not correlate with transcription, with the exception of the Cwc22. At this locus, BPA exposure increased chromatin accessibility in half of mice of both sexes, leading to an "unchanged/open" bimodal profile correlated with "unchanged/upregulated" gene expression. Together, these results open new insights into the sex-dependent mechanisms of BPA's effects on brain development.
Collapse
Affiliation(s)
- Vanessa Naffaa
- Université Paris Cité, CNRS, CiTCoM, Paris 75006, France
| | | | - Romain Magny
- Université Paris Cité, CNRS, CiTCoM, Paris 75006, France
| | | | - Céline Keime
- Université de Strasbourg, IGBMC, Inserm U1258, CNRS UMR7104, Illkirch 67404, France
| | - Pierre Gressens
- Université Paris Cité, Inserm, NeuroDiderot, Paris 75019, France
| | | | - Nicolas Auzeil
- Université Paris Cité, CNRS, CiTCoM, Paris 75006, France
| | - Anne-Laure Schang
- Université Paris Cité, Inserm, HERA Team, CRESS UMR 1153, Paris 75006, France.
| |
Collapse
|
19
|
Norris CL, Sandler DP, Pratt GC, Stenzel MR, Stewart PA, Jackson WB, Christenbury KE, Werder EJ, Groth CP, Banerjee S, Lawrence KG, Engel LS. Fine particulate matter from burning oil and gas and associated neurological symptoms among Deepwater Horizon oil spill cleanup workers. ENVIRONMENTAL SCIENCE. PROCESSES & IMPACTS 2025; 27:423-436. [PMID: 39815820 PMCID: PMC11843438 DOI: 10.1039/d4em00469h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 01/06/2025] [Indexed: 01/18/2025]
Abstract
Burning and flaring of oil and gas following the 2010 Deepwater Horizon (DWH) oil spill generated high airborne concentrations of fine particulate matter (PM2.5). Neurological effects of PM2.5 have been previously reported, but this relationship has received limited attention in the context of oil spills. We evaluated associations between burning-related PM2.5 and prevalence of self-reported neurological symptoms during, and 1-3 years after, the DWH disaster cleanup. For 9914 DWH disaster responders in the Gulf Long-term Follow-up Study who worked on the water, we examined aggregate outcomes (central nervous system [CNS; dizziness, sweating, palpitations, nausea, or migraine/severe headache] and peripheral nervous system [PNS; tingling/numbness in extremities, blurred vision, or stumbling] symptoms) and individual symptoms (CNS and PNS symptoms, plus insomnia, vomiting, seizures, and fatigue). We estimated PM2.5 concentrations via Gaussian plume dispersion models and linked these to detailed DWH cleanup work histories. We used log-binomial regression to estimate adjusted prevalence ratios (PR) and 95% confidence intervals, accounting for age, race, ethnicity, and sex, and DWH disaster-related co-exposures to benzene, toluene, ethylbenzene, xylene, and n-hexane (BTEX-H). We examined effect measure modification by age, race, smoking, and BTEX-H exposure. During the disaster, 34% of participants experienced at least one symptom (23% CNS, 12% PNS); 1-3 years later, 30% did (19% CNS, 17% PNS). Evidence of associations with PM2.5 was most consistent for CNS symptoms (PR range: 1.17 to 1.51), although we did not observe exposure-response trends. For PNS, PR ranged from 0.96 to 1.84. Associations with PM were more apparent among those with lower BTEX-H exposure and among older workers. We found some evidence of an association between burning-related PM2.5 and prevalence of neurologic symptoms during the DWH disaster response and 1-3 years later. Understanding these relationships can inform responses to future disasters to better protect human health.
Collapse
Affiliation(s)
- Christina L Norris
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
| | - Dale P Sandler
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Gregory C Pratt
- Division of Environmental Health, School of Public Health, University of Minnesota, Minneapolis, Minnesota, USA
| | - Mark R Stenzel
- Exposure Assessment Applications, LLC, Arlington, Virginia, USA
| | | | | | | | - Emily J Werder
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Caroline P Groth
- Department of Epidemiology and Biostatistics, West Virginia University School of Public Health, Morgantown, WV, USA
| | - Sudipto Banerjee
- Department of Biostatistics, University of California, Los Angeles Fielding School of Public Health, Los Angeles, CA, USA
| | - Kaitlyn G Lawrence
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| | - Lawrence S Engel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA.
- Epidemiology Branch, National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina, USA
| |
Collapse
|
20
|
Falcón P, Brito Á, Escandón M, Roa JF, Martínez NW, Tapia-Godoy A, Farfán P, Matus S. GCN2-Mediated eIF2α Phosphorylation Is Required for Central Nervous System Remyelination. Int J Mol Sci 2025; 26:1626. [PMID: 40004088 PMCID: PMC11855834 DOI: 10.3390/ijms26041626] [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: 12/20/2024] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Under conditions of amino acid deficiency, mammalian cells activate a nutrient-sensing kinase known as general control nonderepressible 2 (GCN2). The activation of GCN2 results in the phosphorylation of the alpha subunit of the eukaryotic initiation factor 2 (eIF2α), which can be phosphorylated by three other three integrated stress response (ISR) kinases, reducing overall protein synthesis. GCN2 activation also promotes the translation of specific mRNAs, some of which encode transcription factors that enhance the transcription of genes involved in the synthesis, transport, and metabolism of amino acids to restore cellular homeostasis. The phosphorylation of eIF2α has been shown to protect oligodendrocytes, the cells responsible for producing myelin in the central nervous system during remyelination. Here, we explore the potential role of the kinase GCN2 in the myelination process. We challenged mice deficient in the GCN2-encoding gene with a pharmacological demyelinating stimulus (cuprizone) and evaluated the recovery of myelin as well as ISR activation through the levels of eIF2α phosphorylation. Our findings indicate that GCN2 controls the establishment of myelin by fine-tuning its abundance and morphology in the central nervous system. We also found that GCN2 is essential for remyelination. Surprisingly, we discovered that GCN2 is necessary to maintain eIF2α levels during remyelination.
Collapse
Affiliation(s)
- Paulina Falcón
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
| | - Álvaro Brito
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
| | - Marcela Escandón
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Ph.D. “Program in Cell Biology and Biomedicine”, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile
| | - Juan Francisco Roa
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Ph.D. “Program in Cell Biology and Biomedicine”, Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile
| | - Nicolas W. Martínez
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580704, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile
| | - Ariel Tapia-Godoy
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580704, Chile
| | - Pamela Farfán
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580704, Chile
| | - Soledad Matus
- Fundación Ciencia & Vida, Avenida del Valle 725, Huechuraba, Santiago 8580704, Chile; (P.F.); (Á.B.); (M.E.); (J.F.R.); (N.W.M.); (A.T.-G.); (P.F.)
- Centro Ciencia & Vida, Fundación Ciencia & Vida, Santiago 8580704, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Santiago 7510157, Chile
| |
Collapse
|
21
|
Vale-Silva R, de Paes de Faria J, Seixas AI, Brakebusch C, Franklin RJM, Relvas JB. RhoA regulates oligodendrocyte differentiation and myelination by orchestrating cortical and membrane tension. Glia 2025; 73:381-398. [PMID: 39495111 DOI: 10.1002/glia.24640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
Timely differentiation and myelin formation by oligodendrocytes are essential for the physiological functioning of the central nervous system (CNS). While the Rho GTPase RhoA has been hinted as a negative regulator of myelin sheath formation, the precise in vivo mechanisms have remained elusive. Here we show that RhoA controls the timing and progression of myelination by oligodendrocytes through a fine-tuned balance between cortical tension, membrane tension and cell shape. Using a conditional mouse model, we observe that Rhoa ablation results in the acceleration of myelination driven by hastened differentiation and facilitated through membrane expansion induced by changes in MLCII activity and in F-actin redistribution and turnover within the cell. These findings reveal RhoA as a central molecular integrator of alterations in actin cytoskeleton, actomyosin contractility and membrane tension underlying precise morphogenesis of oligodendrocytes and normal myelination of the CNS.
Collapse
Affiliation(s)
- Raquel Vale-Silva
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas Abel Salazar (ICBAS), Universidade do Porto, Porto, Portugal
| | - Joana de Paes de Faria
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Ana Isabel Seixas
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
| | - Cord Brakebusch
- Biotech Research and Innovation Centre (BRIC), Københavns Biocenter, Copenhagen, Denmark
| | | | - João B Relvas
- Instituto de Investigação e Inovação em Saúde (i3S), Universidade do Porto, Porto, Portugal
- Department of Biomedicine, Faculdade de Medicina, Universidade do Porto, Porto, Portugal
| |
Collapse
|
22
|
Bizen N, Takebayashi H. Diverse functions of DEAD-box proteins in oligodendrocyte development, differentiation, and homeostasis. J Neurochem 2025; 169:e16238. [PMID: 39374171 DOI: 10.1111/jnc.16238] [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: 05/30/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024]
Abstract
Oligodendrocytes, a type of glial cell in the central nervous system, have a critical role in the formation of myelin around axons, facilitating saltatory conduction, and maintaining the integrity of nerve axons. The dysregulation of oligodendrocyte differentiation and homeostasis have been implicated in a wide range of neurological diseases, including dysmyelinating disorders (e.g., Pelizaeus-Merzbacher disease), demyelinating diseases (e.g., multiple sclerosis), Alzheimer's disease, and psychiatric disorders. Therefore, unraveling the mechanisms of oligodendrocyte development, differentiation, and homeostasis is essential for understanding the pathogenesis of these diseases and the development of therapeutic interventions. Numerous studies have identified and analyzed the functions of transcription factors, RNA metabolic factors, translation control factors, and intracellular and extracellular signals involved in the series of processes from oligodendrocyte fate determination to terminal differentiation. DEAD-box proteins, multifunctional RNA helicases that regulate various intracellular processes, including transcription, RNA processing, and translation, are increasingly recognized for their diverse roles in various aspects of oligodendrocyte development, differentiation, and maintenance of homeostasis. This review introduces the latest insights into the regulatory networks of oligodendrocyte biology mediated by DEAD-box proteins.
Collapse
Affiliation(s)
- Norihisa Bizen
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Center for Anatomical Studies, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
23
|
Sharma T, Mehan S, Tiwari A, Khan Z, Gupta GD, Narula AS. Targeting Oligodendrocyte Dynamics and Remyelination: Emerging Therapies and Personalized Approaches in Multiple Sclerosis Management. Curr Neurovasc Res 2025; 21:359-417. [PMID: 39219420 DOI: 10.2174/0115672026336440240822063430] [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: 06/27/2024] [Revised: 01/01/1970] [Accepted: 07/12/2024] [Indexed: 09/04/2024]
Abstract
Multiple sclerosis (MS) is a progressive autoimmune condition that primarily affects young people and is characterized by demyelination and neurodegeneration of the central nervous system (CNS). This in-depth review explores the complex involvement of oligodendrocytes, the primary myelin- producing cells in the CNS, in the pathophysiology of MS. It discusses the biochemical processes and signalling pathways required for oligodendrocytes to function and remain alive, as well as how they might fail and cause demyelination to occur. We investigate developing therapeutic options that target remyelination, a fundamental component of MS treatment. Remyelination approaches promote the survival and differentiation of oligodendrocyte precursor cells (OPCs), restoring myelin sheaths. This improves nerve fibre function and may prevent MS from worsening. We examine crucial parameters influencing remyelination success, such as OPC density, ageing, and signalling pathway regulation (e.g., Retinoid X receptor, LINGO-1, Notch). The review also examines existing neuroprotective and antiinflammatory medications being studied to see if they can assist oligodendrocytes in surviving and reducing the severity of MS symptoms. The review focuses on medicines that target the myelin metabolism in oligodendrocytes. Altering oligodendrocyte metabolism has been linked to reversing demyelination and improving MS patient outcomes through various mechanisms. We also explore potential breakthroughs, including innovative antisense technologies, deep brain stimulation, and the impact of gut health and exercise on MS development. The article discusses the possibility of personalized medicine in MS therapy, emphasizing the importance of specific medicines based on individual molecular profiles. The study emphasizes the need for reliable biomarkers and improved imaging tools for monitoring disease progression and therapy response. Finally, this review focuses on the importance of oligodendrocytes in MS and the potential for remyelination therapy. It also underlines the importance of continued research to develop more effective treatment regimens, taking into account the complexities of MS pathology and the different factors that influence disease progression and treatment.
Collapse
Affiliation(s)
- Tarun Sharma
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Aarti Tiwari
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | - Zuber Khan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, India
| | | | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC 27516, USA
| |
Collapse
|
24
|
Padmanabhan P, Lu J, Ng KC, Srinivasan DK, Sundramurthy K, Nilewski LG, Sikkema WKA, Tour JM, Kent TA, Gulyás B, Carlstedt-Duke J. Neuroprotective Effects of Functionalized Hydrophilic Carbon Clusters: Targeted Therapy of Traumatic Brain Injury in an Open Blast Rat Model. Biomedicines 2024; 12:2832. [PMID: 39767738 PMCID: PMC11673356 DOI: 10.3390/biomedicines12122832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/28/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Traumatic brain injury (TBI) causes multiple cerebrovascular disruptions and oxidative stress. These pathological mechanisms are often accompanied by serious impairment of cerebral blood flow autoregulation and neuronal and glial degeneration. BACKGROUND/OBJECTIVES Multiple biochemical cascades are triggered by brain damage, resulting in reactive oxygen species production alongside blood loss and hypoxia. However, most currently available early antioxidant therapies lack capacity and hence sufficient efficacy against TBI. The aim of this study was to test a novel catalytic antioxidant nanoparticle to alleviate the damage occurring in blast TBI. METHODS TBI was elicited in an open blast rat model, in which the rats were exposed to the effects of an explosive blast. Key events of the post-traumatic chain in the brain parenchyma were studied using immunohistochemistry. The application of a newly developed biologically compatible catalytic superoxide dismutase mimetic carbon-based nanocluster, a poly-ethylene-glycol-functionalized hydrophilic carbon cluster (PEG-HCC), was tested post-blast to modulate the components of the TBI process. RESULTS The PEG-HCC was shown to significantly ameliorate neuronal loss in the brain cortex, the dentate gyrus, and hippocampus when administered shortly after the blast. There was also a significant increase in endothelial activity to repair blood-brain barrier damage as well as the modulation of microglial and astrocyte activity and an increase in inducible NO synthase in the cortex. CONCLUSIONS We have demonstrated qualitatively and quantitatively that the previously demonstrated antioxidant properties of PEG-HCCs have a neuroprotective effect after traumatic brain injury following an explosive blast, acting at multiple levels of the pathological chain of events elicited by TBI.
Collapse
Affiliation(s)
- Parasuraman Padmanabhan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (J.L.); (K.S.)
| | - Jia Lu
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (J.L.); (K.S.)
- Defence Science Organisation National Laboratories, Singapore 117510, Singapore;
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Singapore, Singapore 119228, Singapore;
| | - Kian Chye Ng
- Defence Science Organisation National Laboratories, Singapore 117510, Singapore;
| | - Dinesh Kumar Srinivasan
- Department of Anatomy, Yong Loo Lin School of Medicine, National University Singapore, Singapore 119228, Singapore;
| | - Kumar Sundramurthy
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (J.L.); (K.S.)
| | - Lizanne Greer Nilewski
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (L.G.N.); (W.K.A.S.); (J.M.T.); (T.A.K.)
| | - William K. A. Sikkema
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (L.G.N.); (W.K.A.S.); (J.M.T.); (T.A.K.)
| | - James M. Tour
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (L.G.N.); (W.K.A.S.); (J.M.T.); (T.A.K.)
| | - Thomas A. Kent
- Department of Chemistry, Rice University, Houston, TX 77005, USA; (L.G.N.); (W.K.A.S.); (J.M.T.); (T.A.K.)
- Institute of Biosciences and Technology, Texas A&M University, Houston, TX 77030, USA
| | - Balázs Gulyás
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 636921, Singapore; (J.L.); (K.S.)
| | - Jan Carlstedt-Duke
- President’s Office (Retired), Nanyang Technological University, Singapore 639798, Singapore
| |
Collapse
|
25
|
Ramirez AM, Bertholim-Nasciben L, Moura S, Coombs LE, Rajabli F, DeRosa BA, Whitehead PG, Adams LD, Starks TD, Mena P, Illannes-Manrique M, Tejada SJ, Byrd GS, Caban-Holt A, Cuccaro M, McInerney K, Cornejo-Olivas M, Feliciano-Astacio B, Wang L, Robayo MC, Xu W, Jin F, Pericak-Vance MA, Griswold AJ, Dykxhoorn DM, Young JI, Vance JM. Ancestral Genomic Functional Differences in Oligodendroglia: Implications for Alzheimer's Disease. RESEARCH SQUARE 2024:rs.3.rs-5338140. [PMID: 39678342 PMCID: PMC11643296 DOI: 10.21203/rs.3.rs-5338140/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Background This study aims to elucidate ancestry-specific changes to the genomic regulatory architecture in induced pluripotent stem cell (iPSC)-derived oligodendroglia, focusing on their implications for Alzheimer's disease (AD). This work addresses the lack of diversity in previous iPSC studies by including ancestries that contribute to African American (European/African) and Hispanic/Latino populations (Amerindian/African/European). Methods We generated 12 iPSC lines-four African, four Amerindian, and four European- from both AD patients and non-cognitively impaired individuals, with varying APOE genotypes (APOE3/3 and APOE4/4). These lines were differentiated into neural spheroids containing oligodendrocyte lineage cells. Single-nuclei RNA sequencing and ATAC sequencing were employed to analyze transcriptional and chromatin accessibility profiles, respectively. Differential gene expression, chromatin accessibility, and Hi-C analyses were conducted, followed by pathway analysis to interpret the results. Results We identified ancestry-specific differences in gene expression and chromatin accessibility. Notably, numerous AD GWAS-associated genes were differentially expressed across ancestries. The largest number of differentially expressed genes (DEGs) were found in European vs. Amerindian and African vs. Amerindian iPSC-derived oligodendrocyte progenitor cells (OPCs). Pathway analysis of APOE4/4 carriers vs APOE3/3 carriers exhibited upregulation of a large number of disease and metabolic pathways in APOE4/4 individuals of all ancestries. Of particular interest was that APOE4/4 carriers had significantly upregulated cholesterol biosynthesis genes relative to APOE3/3 individuals across all ancestries, strongest in iOPCs. Comparison of iOPC and iOL transcriptome data with corresponding human frontal cortex data demonstrated a high correlation (R2 > 0.85). Conclusions This research emphasizes the importance of including diverse ancestries in AD research to uncover critical gene expression differences between populations and ancestries that may influence disease susceptibility and therapeutic interventions. The upregulation of cholesterol biosynthesis genes in APOE4/4 carriers of all three ancestries supports the concept that APOE4 may produce disease effects early in life, which could have therapeutic implications as we move forward towards specific therapy for APOE4 carriers. These findings and the high correlation between brain and iPSC-derived OPC and OL transcriptomes support the relevance of this approach as a model for disease study.
Collapse
Affiliation(s)
- Aura M Ramirez
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | | | - Sofia Moura
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Lauren E Coombs
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Farid Rajabli
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Brooke A DeRosa
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Patrice G Whitehead
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Larry D Adams
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Takiyah D Starks
- Wake Forest School of Medicine: Wake Forest University School of Medicine
| | - Pedro Mena
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | | | - Sergio J Tejada
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Goldie S Byrd
- Wake Forest School of Medicine: Wake Forest University School of Medicine
| | - Allison Caban-Holt
- Wake Forest School of Medicine: Wake Forest University School of Medicine
| | - Michael Cuccaro
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Katalina McInerney
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Mario Cornejo-Olivas
- Universidad Científica del Sur Facultad de Ciencias de la Salud: Universidad Cientifica del Sur Facultad de Ciencias de la Salud
| | | | - Liyong Wang
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Maria C Robayo
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Wanying Xu
- Case Western Reserve University School of Medicine
| | - Fulai Jin
- Case Western Reserve University School of Medicine
| | | | - Anthony J Griswold
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Derek M Dykxhoorn
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Juan I Young
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| | - Jeffery M Vance
- University of Miami Miller School of Medicine: University of Miami School of Medicine
| |
Collapse
|
26
|
Smith S, Swan ER, Furber KL. Establishing validated RT-qPCR workflow for the analysis of oligodendrocyte gene expression in the developing murine brain. Biochem Cell Biol 2024; 102:492-505. [PMID: 39116457 DOI: 10.1139/bcb-2024-0088] [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: 08/10/2024] Open
Abstract
Myelination is essential for the proper conduction of impulses across neuronal networks. Mature, myelinating glia differentiate from progenitor cells through distinct stages that correspond to oligodendrocyte-specific gene expression markers. Reverse transcription quantiatative PCR (RT-qPCR) is a common technique used to quantify gene expression across cell development; however, a lack of standardization and transparency in methodology may lead to irreproducible data. Here, we have designed and validated RT-qPCR assays for oligodendrocyte genes and reference genes in the developing C57BL6/J mouse brain that align with the MIQE guidelines, including quality controls for primer specificity, temperature dependence, and efficiency. A panel of eight commonly used reference genes was ranked using a series of reference gene stability methods that consistently identified Gapdh, Sdha, Hmbs, Hprt1, and Pgk1 as the top candidates for normalization across brain regions. In the cerebrum, myelin genes peaked in expression at postnatal day 21, which corresponds to the peak of developmental myelination. The gene expression patterns from the brain homogenate were in agreement with previously reported RNA-seq and microarray profiles from oligodendrocyte lineage cells. The validated RT-qPCR assays begin to build a framework for future investigation into the molecular mechanisms that regulate myelination in mouse models of brain development, aging, and disease.
Collapse
Affiliation(s)
- Samantha Smith
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, BC, Canada
| | - Emma R Swan
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, BC, Canada
| | - Kendra L Furber
- Northern Medical Program, Division of Medical Sciences, University of Northern British Columbia, Prince George, BC, Canada
| |
Collapse
|
27
|
Fujiwara Y, Eitoku S, Sakae N, Izumi T, Kumazoe H, Kitajima M. Single-point macromolecular proton fraction mapping using a 0.3 T permanent magnet MRI system: phantom and healthy volunteer study. Radiol Phys Technol 2024; 17:869-877. [PMID: 39251498 DOI: 10.1007/s12194-024-00843-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 08/12/2024] [Accepted: 09/02/2024] [Indexed: 09/11/2024]
Abstract
In a 0.3 T permanent-magnet magnetic resonance imaging (MRI) system, quantifying myelin content is challenging owing to long imaging times and low signal-to-noise ratio. macromolecular proton fraction (MPF) offers a quantitative assessment of myelin in the nervous system. We aimed to demonstrate the practical feasibility of MPF mapping in the brain using a 0.3 T MRI. Both 0.3 T and 3.0 T MRI systems were used. The MPF-mapping protocol used a standard 3D fast spoiled gradient-echo sequence based on the single-point reference method. Proton density, T1, and magnetization transfer-weighted images were obtained from a protein phantom at 0.3 T and 3.0 T to calculate MPF maps. MPF was measured in all phantom sections to assess its relationship to protein concentration. We acquired MPF maps for 16 and 8 healthy individuals at 0.3 T and 3.0 T, respectively, measuring MPF in nine brain tissues. Differences in MPF between 0.3 T and 3.0 T, and between 0.3 T and previously reported MPF at 0.5 T, were investigated. Pearson's correlation coefficient between protein concentration and MPF at 0.3 T and 3.0 T was 0.92 and 0.90, respectively. The 0.3 T MPF of brain tissue strongly correlated with 3.0 T MPF and literature values measured at 0.5 T. The absolute mean differences in MPF between 0.3 T and 0.5 T were 0.42% and 1.70% in white and gray matter, respectively. Single-point MPF mapping using 0.3 T permanent-magnet MRI can effectively assess myelin content in neural tissue.
Collapse
Affiliation(s)
- Yasuhiro Fujiwara
- Department of Medical Imaging Technology, Faculty of Life Sciences, Kumamoto University, 4-24-1, Kuhonji, Chuo-Ku, Kumamoto, 862-0976, Japan.
| | - Shoma Eitoku
- Department of Radiology, Hospital of the University of Occupational and Environmental Health, 1-1, Iseigaoka, Yahatanishi-Ku, Kitakyushu, 807-8556, Japan
| | - Nobutaka Sakae
- Department of Neurology, National Hospital Organization Omuta National Hospital, 1044-1, Tachibana, Omuta, 837-0911, Japan
| | - Takahisa Izumi
- Department of Radiology, National Hospital Organization Kumamoto Saishun Medical Center, 2659 Suya, Koshi, Kumamoto, 861-1196, Japan
| | - Hiroyuki Kumazoe
- Department of Radiology, National Hospital Organization Omuta National Hospital, 1044-1, Tachibana, Omuta, 837-0911, Japan
| | - Mika Kitajima
- Department of Diagnostic Imaging Technology, Faculty of Life Sciences, Kumamoto University, 4-24-1, Kuhonji, Chuo-Ku, Kumamoto, 862-0976, Japan
| |
Collapse
|
28
|
Wang Q, Wang X, Shang Z, Zhao L. Mechanism and prospects of mitochondrial transplantation for spinal cord injury treatment. Stem Cell Res Ther 2024; 15:457. [PMID: 39609871 PMCID: PMC11606159 DOI: 10.1186/s13287-024-04077-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 11/22/2024] [Indexed: 11/30/2024] Open
Abstract
Spinal cord injury (SCI) involves a continuous and dynamic cascade of complex reactions, with mitochondrial damage and dysfunction-induced energy metabolism disorders playing a central role throughout the process. These disorders not only determine the severity of secondary injuries but also influence the potential for axonal regeneration. Given the critical role of energy metabolism disturbances in the pathology of SCI, strategies such as enhancing mitochondrial transport within axons to alleviate local energy deficits, or transplanting autologous or allogeneic mitochondria to restore energy supply to damaged tissues, have emerged as potential approaches for SCI repair. These strategies also aim to modulate local inflammatory responses and apoptosis. Preclinical studies have initially demonstrated that mitochondrial transplantation (MT) significantly reduces neuronal death and promotes axonal regeneration following spinal cord injury. MT achieves this by regulating signaling pathways such as MAPK/ERK and PI3K/Akt, promoting the expression of growth-associated protein-43 (GAP-43) in neurons, and inhibiting the expression of apoptosis-related proteins like Grp78, Chop, and P-Akt, thereby enhancing the survival and regeneration of damaged neurons. Additionally, MT plays a role in promoting the expression of vascular endothelial growth factor, facilitating tissue repair, and reducing the secretion of pro-inflammatory cytokines such as TNF-α, IL-1β, and IL-6. Furthermore, MT modulates neuronal apoptosis and inflammatory responses by decreasing the expression of p-JNK, a member of the MAPK family. In summary, by reviewing the detailed mechanisms underlying the cascade of pathological processes in SCI, we emphasize the changes in endogenous mitochondria post-SCI and the potential of exogenous MT in SCI repair. This review aims to provide insights and a basis for developing more effective clinical treatments for SCI.
Collapse
Affiliation(s)
- Qin Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
| | - Xin Wang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China.
- Department of Orthopedics, The First Hospital of Lanzhou University, Lanzhou, 730000, China.
| | - Zhizhong Shang
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
- The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510000, China
| | - Long Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou, 730000, China
- Department of Orthopedics, The First Hospital of Lanzhou University, Lanzhou, 730000, China
| |
Collapse
|
29
|
Ishii A, Pathoulas JA, MoustafaFathy Omar O, Ge Y, Yao AY, Pantalena T, Singh N, Zhou J, He W, Murphy P, Yan R, Hu X. Contribution of amyloid deposition from oligodendrocytes in a mouse model of Alzheimer's disease. Mol Neurodegener 2024; 19:83. [PMID: 39548583 PMCID: PMC11568619 DOI: 10.1186/s13024-024-00759-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: 07/26/2024] [Accepted: 10/01/2024] [Indexed: 11/18/2024] Open
Abstract
BACKGROUND The accumulation of β-amyloid (Aβ) peptides into insoluble plaques is an early pathological feature of Alzheimer's disease (AD). BACE1 is the sole β-secretase for Aβ generation, making it an attractive therapeutic target for AD therapy. While BACE1 inhibitors have been shown to reduce Aβ levels in people with AD, clinical trials targeting BACE1 have failed due to unwanted synaptic deficits. Understanding the physiological role of BACE1 in individual cell types is essential for developing effective BACE inhibitors for the treatment of AD. Recent single-cell RNA transcriptomic assays revealed that oligodendrocytes are enriched with genes required for generating Aβ. However, the contribution of oligodendrocytes to amyloid plaque burden in AD and the side effects of oligodendrocyte-specific Bace1 deletion remain to be explored. METHODS We generated an oligodendrocyte-specific Bace1 knockout model (Bace1fl/fl;Olig2-Cre) to monitor potential disruptions in myelination using standard electron microscopy. Long-term potentiation (LTP) was monitored to measure synaptic integrity. We crossed the Bace1fl/fl;Olig2-Cre model with heterozygous AppNL-G-F/wt knock-in AD mice to generate AD mice lacking oligodendrocyte Bace1 (Bace1fl/fl;Olig2-Cre; AppNL-G-F/wt) and examined amyloid plaque number and insoluble Aβ levels and gliosis in these animals. Single nuclei RNA sequencing experiments were conducted to examine molecular changes in response to Bace1 deficiency in oligodendrocytes in the wild type or APP knock-in background. RESULTS Bace1 deletion in oligodendrocytes caused no change in myelin thickness in the corpus callosum but a marginal reduction in myelin sheath thickness of the optic nerve. Synaptic strength measured by LTP was not different between Bace1fl/fl;Olig2-Cre and age-matched Bace1fl/fl control animals, suggesting no major effect on synaptic plasticity. Intriguingly, deletion of Bace1 in 12-month-old heterozygous AD knock-in mice (Bace1fl/fl;Olig2-Cre; AppNL-G-F/wt mice) caused a significant reduction of amyloid plaques by ~ 33% in the hippocampus and ~ 29% in the cortex compared to age-matched AD mice (Bace1fl/fl;AppNL-G-F/wt). Insoluble Aβ1-40 and Aβ1-42 levels were reduced comparably while more astrocytes and microglia were observed in surrounding amyloid plaques. Unbiased single-nuclei RNA sequencing results revealed that deletion of oligodendrocyte Bace1 in APPNL-G-F/wt knock-in mice increased expression of genes associated with Aβ generation and clearance such as ADAM10, Ano4, ApoE, Il33, and Sort1. CONCLUSION Our results provide compelling evidence that the amyloidogenic pathway in oligodendrocytes contributes to Aβ plaque formation in the AD brain. While specifically targeting BACE1 inhibition in oligodendrocytes for reducing Aβ pathology in AD is likely challenging, this is a potentially explorable strategy in future studies.
Collapse
Affiliation(s)
- Akihiro Ishii
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - Joseph A Pathoulas
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - Omar MoustafaFathy Omar
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - Yingying Ge
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - Annie Y Yao
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - Tressa Pantalena
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - Neeraj Singh
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - John Zhou
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - Wanxia He
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - Patrick Murphy
- Department of Cell Biology and Vascular Biology Center, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA.
| | - Xiangyou Hu
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA.
- Department of Neuroscience, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT, 06030-3401, USA.
| |
Collapse
|
30
|
Wu S, Lin W. The physiological role of the unfolded protein response in the nervous system. Neural Regen Res 2024; 19:2411-2420. [PMID: 38526277 PMCID: PMC11090440 DOI: 10.4103/1673-5374.393105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Accepted: 12/12/2023] [Indexed: 03/26/2024] Open
Abstract
The unfolded protein response (UPR) is a cellular stress response pathway activated when the endoplasmic reticulum, a crucial organelle for protein folding and modification, encounters an accumulation of unfolded or misfolded proteins. The UPR aims to restore endoplasmic reticulum homeostasis by enhancing protein folding capacity, reducing protein biosynthesis, and promoting protein degradation. It also plays a pivotal role in coordinating signaling cascades to determine cell fate and function in response to endoplasmic reticulum stress. Recent research has highlighted the significance of the UPR not only in maintaining endoplasmic reticulum homeostasis but also in influencing various physiological processes in the nervous system. Here, we provide an overview of recent findings that underscore the UPR's involvement in preserving the function and viability of neuronal and myelinating cells under physiological conditions, and highlight the critical role of the UPR in brain development, memory storage, retinal cone development, myelination, and maintenance of myelin thickness.
Collapse
Affiliation(s)
- Shuangchan Wu
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| | - Wensheng Lin
- Department of Neuroscience, University of Minnesota, Minneapolis, MN, USA
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
31
|
Tiwari S, Phoolmala, Goyal S, Yadav RK, Chaturvedi RK. Bisphenol-F and Bisphenol-S (BPF and BPS) Impair the Stemness of Neural Stem Cells and Neuronal Fate Decision in the Hippocampus Leading to Cognitive Dysfunctions. Mol Neurobiol 2024; 61:9347-9368. [PMID: 38635025 DOI: 10.1007/s12035-024-04160-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 04/01/2024] [Indexed: 04/19/2024]
Abstract
Neurogenesis occurs throughout life in the hippocampus of the brain, and many environmental toxicants inhibit neural stem cell (NSC) function and neuronal generation. Bisphenol-A (BPA), an endocrine disrupter used for surface coating of plastic products causes injury in the developing and adult brain; thus, many countries have banned its usage in plastic consumer products. BPA analogs/alternatives such as bisphenol-F (BPF) and bisphenol-S (BPS) may also cause neurotoxicity; however, their effects on neurogenesis are still not known. We studied the effects of BPF and BPS exposure from gestational day 6 to postnatal day 21 on neurogenesis. We found that exposure to non-cytotoxic concentrations of BPF and BPS significantly decreased the number/size of neurospheres, BrdU+ (proliferating NSC marker) and MAP-2+ (neuronal marker) cells and GFAP+ astrocytes in the hippocampus NSC culture, suggesting reduced NSC stemness and self-renewal and neuronal differentiation and increased gliogenesis. These analogs also reduced the number of BrdU/Sox-2+, BrdU/Dcx+, and BrdU/NeuN+ co-labeled cells in the hippocampus of the rat brain, suggesting decreased NSC proliferation and impaired maturation of newborn neurons. BPF and BPS treatment increases BrdU/cleaved caspase-3+ cells and Bax-2 and cleaved caspase protein levels, leading to increased apoptosis in hippocampal NSCs. Transmission electron microscopy studies suggest that BPF and BPS also caused degeneration of neuronal myelin sheath, altered mitochondrial morphology, and reduced number of synapses in the hippocampus leading to altered cognitive functions. These results suggest that BPF and BPS exposure decreased the NSC pool, inhibited neurogenesis, induced apoptosis of NSCs, caused myelin degeneration/synapse degeneration, and impaired learning and memory in rats.
Collapse
Affiliation(s)
- Saurabh Tiwari
- Molecular Neurotoxicology and Cell Integrity Laboratory, Systems Toxicology and Health Risk Assessment Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh (U.P.), India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Phoolmala
- Molecular Neurotoxicology and Cell Integrity Laboratory, Systems Toxicology and Health Risk Assessment Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh (U.P.), India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Shweta Goyal
- Molecular Neurotoxicology and Cell Integrity Laboratory, Systems Toxicology and Health Risk Assessment Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh (U.P.), India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ranjeet Kumar Yadav
- Molecular Neurotoxicology and Cell Integrity Laboratory, Systems Toxicology and Health Risk Assessment Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh (U.P.), India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rajnish Kumar Chaturvedi
- Molecular Neurotoxicology and Cell Integrity Laboratory, Systems Toxicology and Health Risk Assessment Group, FEST Division, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, 226001, Uttar Pradesh (U.P.), India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
32
|
Aguilar-Delgadillo A, Cruz-Mendoza F, Luquin-de Andais teh S, Ruvalcaba-Delgadillo Y, Jáuregui-Huerta F. Stress-induced c-fos expression in the medial prefrontal cortex differentially affects the main residing cell phenotypes. Heliyon 2024; 10:e39325. [PMID: 39498004 PMCID: PMC11532284 DOI: 10.1016/j.heliyon.2024.e39325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 11/07/2024] Open
Abstract
Stress poses a challenge to the body's equilibrium and triggers a series of responses that enable organisms to adapt to stressful stimuli. The medial prefrontal cortex (mPFC), particularly in acute stress conditions, undergoes significant physiological changes to cope with the demands associated with cellular activation. The proto-oncogene c-fos and its protein product c-Fos have long been utilized to investigate the effects of external factors on the central nervous system (CNS). While c-Fos expression has traditionally been attributed to neurons, emerging evidence suggests its potential expression in glial cells. In this study, our main objective was to explore the expression of c-Fos in glial cells and examine how acute stress influences these activity patterns. We conducted our experiments on male Wistar rats, subjecting them to acute stress and sacrificing them 2 h after the stressor initiation. Using double-labelling fluorescent immunohistochemistry targeting c-Fos, along with markers such as GFAP, Iba-1, Olig2, NG2, and NeuN, we analyzed 35 μm brain slices obtained from the mPFC. Our findings compellingly demonstrate that c-Fos expression extends beyond neurons and is present in astrocytes, oligodendrocytes, microglia, and NG2 cells-the diverse population of glial cells. Moreover, we observed distinct regulation of c-Fos expression in response to stress across different subregions of the mPFC. These results emphasize the importance of considering glial cells and their perspective in studies investigating brain activity, highlighting c-Fos as a response marker in glial cells. By shedding light on the differential regulation of c-Fos expression in response to stress, our study contributes to the understanding of glial cell involvement in stress-related processes. This underscores the significance of including glial cells in investigations of brain activity and expands our knowledge of c-Fos as a potential marker for glial cell responses.
Collapse
Affiliation(s)
| | - Fernando Cruz-Mendoza
- Neurosciences Department, Health sciences center, University of Guadalajara, Guadalajara, Mexico
| | | | | | - Fernando Jáuregui-Huerta
- Neurosciences Department, Health sciences center, University of Guadalajara, Guadalajara, Mexico
- Department of Physiology, School of Medicine, National Autonomous University of Mexico, Mexico City, Mexico
| |
Collapse
|
33
|
Sutley-Koury SN, Anderson A, Taitano-Johnson C, Ajayi M, Kulinich AO, Contreras K, Regalado J, Tiwari-Woodruff SK, Ethell IM. Astrocytic Ephrin-B1 Regulates Oligodendrocyte Development and Myelination. ASN Neuro 2024; 16:2401753. [PMID: 39437409 PMCID: PMC11792131 DOI: 10.1080/17590914.2024.2401753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2024] Open
Abstract
Astrocytes have been implicated in oligodendrocyte development and myelination, however, the mechanisms by which astrocytes regulate oligodendrocytes remain unclear. Our findings suggest a new mechanism that regulates astrocyte-mediated oligodendrocyte development through ephrin-B1 signaling in astrocytes. Using a mouse model, we examined the role of astrocytic ephrin-B1 signaling in oligodendrocyte development by deleting ephrin-B1 specifically in astrocytes during the postnatal days (P)14-P28 period and used mRNA analysis, immunohistochemistry, and mouse behaviors to study its effects on oligodendrocytes and myelination. We found that deletion of astrocytic ephrin-B1 downregulated many genes associated with oligodendrocyte development, myelination, and lipid metabolism in the hippocampus and the corpus callosum. Additionally, we observed a reduced number of oligodendrocytes and impaired myelination in the corpus callosum of astrocyte-specific ephrin-B1 KO mice. Finally, our data show reduced motor strength in these mice exhibiting clasping phenotype and impaired performance in the rotarod test most likely due to impaired myelination. Our studies provide new evidence that astrocytic ephrin-B1 positively regulates oligodendrocyte development and myelination, potentially through astrocyte-oligodendrocyte interactions.
Collapse
Affiliation(s)
- Samantha N. Sutley-Koury
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Alyssa Anderson
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Christopher Taitano-Johnson
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
- Neuroscience Graduate Program, University of California Riverside, Riverside, California, USA
| | - Moyinoluwa Ajayi
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Anna O. Kulinich
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Kimberly Contreras
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Jasmin Regalado
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
| | - Seema K. Tiwari-Woodruff
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
- Neuroscience Graduate Program, University of California Riverside, Riverside, California, USA
| | - Iryna M. Ethell
- Division of Biomedical Sciences and Biomedical Sciences Graduate Program, School of Medicine, University of California Riverside, Riverside, California, USA
- Neuroscience Graduate Program, University of California Riverside, Riverside, California, USA
| |
Collapse
|
34
|
Liu P, Zhang K, Tong C, Liu T, Zheng J. Progesterone alleviates esketamine-induced hypomyelination via PI3K/Akt signaling pathway in the developing rat brain. Biotechnol Genet Eng Rev 2024; 40:1202-1217. [PMID: 36946765 DOI: 10.1080/02648725.2023.2193058] [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: 02/10/2023] [Accepted: 03/14/2023] [Indexed: 03/23/2023]
Abstract
The neurodevelopmental toxicity of anesthetics has been confirmed repeatedly, and esketamine is now widely used in pediatric surgeries. Oligodendrocyte precursor cells (OPCs) evolved into mature oligodendrocytes (OLs) and formed myeline sheath during the early brain development. In this study, we investigated whether esketamine exposure interrupted development of OPCs and induced hypomyelination in rats. Further we explored the roles of PI3K/Akt phosphorylation in OPCs development and myelination. Sprague Dawley rats with different ages (postnatal day (P) 1, 3, 7 and 12) were exposed to 40mg/kg esketamine. Progesterone treatment was given (16 mg/kg per day for 3 days) 24 h after esketamine exposure via the intraperitoneal route. Corpus callosum tissues were collected at P8 or P14 for western blot and immunofluorescence analyses. Esketamine exposure at P7 and P12 significantly reduced myelin basic protein (MBP) expression and CC1+ OLs number in corpus callosum. Esketamine exposure at P7 not only aggravated the mature OLs apoptosis, also decreased the OPCs proliferation and differentiation, which was related with dephosphorylation of PI3K/Akt. Progesterone was able to promote OPCs differentiation and ameliorate esketamine-induced hypomyelination by enhancing PI3K/Akt phosphorylation. Stage-dependent abnormality of OPCs/OLs after esketamine leads to the esketamine-induced hypomyelination. Esketamine interrupted OPCs evolution via PI3K/Akt signaling pathway, which can be ameliorated by progesterone.
Collapse
Affiliation(s)
- Peiwen Liu
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Kan Zhang
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chaoyang Tong
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Ting Liu
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jijian Zheng
- Department of Anesthesiology, Shanghai Children's Medical Center & National Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
35
|
Yamaguchi H, Nishimura Y, Matsuse D, Sekiya H, Masaki K, Tanaka T, Saiga T, Harada M, Kira YI, Dickson DW, Fujishima K, Matsuo E, Tanaka KF, Yamasaki R, Isobe N, Kira JI. A rapidly progressive multiple system atrophy-cerebellar variant model presenting marked glial reactions with inflammation and spreading of α-synuclein oligomers and phosphorylated α-synuclein aggregates. Brain Behav Immun 2024; 121:122-141. [PMID: 38986725 DOI: 10.1016/j.bbi.2024.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 04/30/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024] Open
Abstract
Multiple system atrophy (MSA) is a severe α-synucleinopathy facilitated by glial reactions; the cerebellar variant (MSA-C) preferentially involves olivopontocerebellar fibres with conspicuous demyelination. A lack of aggressive models that preferentially involve olivopontocerebellar tracts in adulthood has hindered our understanding of the mechanisms of demyelination and neuroaxonal loss, and thus the development of effective treatments for MSA. We therefore aimed to develop a rapidly progressive mouse model that recaptures MSA-C pathology. We crossed Plp1-tTA and tetO-SNCA*A53T mice to generate Plp1-tTA::tetO-SNCA*A53T bi-transgenic mice, in which human A53T α-synuclein-a mutant protein with enhanced aggregability-was specifically produced in the oligodendrocytes of adult mice using Tet-Off regulation. These bi-transgenic mice expressed mutant α-synuclein from 8 weeks of age, when doxycycline was removed from the diet. All bi-transgenic mice presented rapidly progressive motor deterioration, with wide-based ataxic gait around 22 weeks of age and death around 30 weeks of age. They also had prominent demyelination in the brainstem/cerebellum. Double immunostaining demonstrated that myelin basic protein was markedly decreased in areas in which SM132, an axonal marker, was relatively preserved. Demyelinating lesions exhibited marked ionised calcium-binding adaptor molecule 1-, arginase-1-, and toll-like receptor 2-positive microglial reactivity and glial fibrillary acidic protein-positive astrocytic reactivity. Microarray analysis revealed a strong inflammatory response and cytokine/chemokine production in bi-transgenic mice. Neuronal nuclei-positive neuronal loss and patchy microtubule-associated protein 2-positive dendritic loss became prominent at 30 weeks of age. However, a perceived decrease in tyrosine hydroxylase-positive neurons in the substantia nigra pars compacta in bi-transgenic mice compared with wild-type mice was not significant, even at 30 weeks of age. Wild-type, Plp1-tTA, and tetO-SNCA*A53T mice developed neither motor deficits nor demyelination. In bi-transgenic mice, double immunostaining revealed human α-synuclein accumulation in neurite outgrowth inhibitor A (Nogo-A)-positive oligodendrocytes beginning at 9 weeks of age; its expression was further increased at 10 to 12 weeks, and these increased levels were maintained at 12, 24, and 30 weeks. In an α-synuclein-proximity ligation assay, α-synuclein oligomers first appeared in brainstem oligodendrocytes as early as 9 weeks of age; they then spread to astrocytes, neuropil, and neurons at 12 and 16 weeks of age. α-Synuclein oligomers in the brainstem neuropil were most abundant at 16 weeks of age and decreased thereafter; however, those in Purkinje cells successively increased until 30 weeks of age. Double immunostaining revealed the presence of phosphorylated α-synuclein in Nogo-A-positive oligodendrocytes in the brainstem/cerebellum as early as 9 weeks of age. In quantitative assessments, phosphorylated α-synuclein gradually and successively accumulated at 12, 24, and 30 weeks in bi-transgenic mice. By contrast, no phosphorylated α-synuclein was detected in wild-type, tetO-SNCA*A53T, or Plp1-tTA mice at any age examined. Pronounced demyelination and tubulin polymerisation, promoting protein-positive oligodendrocytic loss, was closely associated with phosphorylated α-synuclein aggregates at 24 and 30 weeks of age. Early inhibition of mutant α-synuclein expression by doxycycline diet at 23 weeks led to fully recovered demyelination; inhibition at 27 weeks led to persistent demyelination with glial reactions, despite resolving phosphorylated α-synuclein aggregates. In conclusion, our bi-transgenic mice exhibited progressively increasing demyelination and neuroaxonal loss in the brainstem/cerebellum, with rapidly progressive motor deterioration in adulthood. These mice showed marked microglial and astrocytic reactions with inflammation that was closely associated with phosphorylated α-synuclein aggregates. These features closely mimic human MSA-C pathology. Notably, our model is the first to suggest that α-synuclein oligomers may spread from oligodendrocytes to neurons in transgenic mice with human α-synuclein expression in oligodendrocytes. This model of MSA is therefore particularly useful for elucidating the in vivo mechanisms of α-synuclein spreading from glia to neurons, and for developing therapies that target glial reactions and/or α-synuclein oligomer spreading and aggregate formation in MSA.
Collapse
Affiliation(s)
- Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; School of Physical Therapy, Faculty of Rehabilitation, Reiwa Health Sciences University, Fukuoka, Japan.
| | - Yuji Nishimura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Dai Matsuse
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Hiroaki Sekiya
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Tatsunori Tanaka
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Sumitomo Pharma Co., Ltd., Osaka, Japan.
| | - Toru Saiga
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Masaya Harada
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Yuu-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | - Kei Fujishima
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Eriko Matsuo
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Kenji F Tanaka
- Division of Brain Sciences, Institute for Advanced Medical Research, Keio University School of Medicine, Tokyo, Japan.
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan; Translational Neuroscience Research Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, Fukuoka, Japan; Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, International University of Health and Welfare, Fukuoka, Japan.
| |
Collapse
|
36
|
Fukushima N, Miyamoto Y, Yamauchi J. CRISPR/CasRx-Mediated Knockdown of Rab7B Restores Incomplete Cell Shape Induced by Pelizaeus-Merzbacher Disease-Associated PLP1 p.Ala243Val. Neurosci Insights 2024; 19:26331055241276873. [PMID: 39280331 PMCID: PMC11402064 DOI: 10.1177/26331055241276873] [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: 05/28/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024] Open
Abstract
Pelizaeus-Merzbacher disease (PMD, currently known as hypomyelinating leukodystrophy type 1 [HLD1]) is a hereditary hypomyelinating and/or demyelinating disease associated with the proteolipid protein 1 (plp1) gene in the central nervous system (CNS). One of the major causes of this condition is incomplete or defective oligodendroglial cell myelin sheath formation triggered by endoplasmic reticulum (ER) stress and subsequent unfolded protein response (UPR). The HLD1-associated Ala-243-to-Val mutation (p.Ala243Val) of PLP1 is widely recognized to trigger defective oligodendroglial cell morphological differentiation, primarily due to ER stress. We have previously reported that knockdown of Rab7B (also known as Rab42), a small GTP/GDP-binding protein involved in intracellular vesicle trafficking around the lysosome, can recover chemical ER stress-induced incomplete cell shapes in the FBD-102b cell line, a model of oligodendroglial cell morphological differentiation. Here, we present findings indicating that incomplete cell shapes induced by PLP1 p.Ala243Val can be restored by knockdown of Rab7B using the clustered regularly interspaced short palindromic repeats (CRISPR) and CasRx (also known as Cas13d) system. Also, the knockdown promoted the trafficking of PLP1 p.Ala243Val to lysosome-associated membrane protein 1 (LAMP1)-positive organelles. These results highlight the unique role of Rab7B knockdown in modulating oligodendroglial cell morphological changes and potentially facilitating the transport of mutated PLP1 to LAMP1-positive organelles, suggesting its potential as a therapeutic target for alleviating HLD1 phenotypes, at least in part, at the molecular and cellular levels.
Collapse
Affiliation(s)
- Nana Fukushima
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| |
Collapse
|
37
|
Adonias GL, Siljak H, Balasubramaniam S, Barros MT. In silico modelling of neuron signal impact of cytokine storm-induced demyelination. Open Biol 2024; 14:240138. [PMID: 39226928 PMCID: PMC11371429 DOI: 10.1098/rsob.240138] [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: 05/22/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 09/05/2024] Open
Abstract
In this study, we develop an in silico model of a neuron's behaviour under demyelination caused by a cytokine storm to investigate the effects of viral infections in the brain. We use a comprehensive model to measure how cytokine-induced demyelination affects the propagation of action potential (AP) signals within a neuron. We analysed the effects of neuron-neuron communications by applying information and communication theory at different levels of demyelination. Our simulations demonstrate that virus-induced degeneration can play a role in the signal power and spiking rate, which compromise the propagation and processing of information between neurons. We propose a transfer function to model the weakening effects on the AP. Our results show that demyelination induced by a cytokine storm not only degrades the signal but also impairs its propagation within the axon. Our proposed in silico model can analyse virus-induced neurodegeneration and enhance our understanding of virus-induced demyelination.
Collapse
Affiliation(s)
- Geoflly L. Adonias
- Walton Institute for Information and Communication Systems Science, South East Technology University, Waterford, Ireland
| | - Harun Siljak
- Department of Electronic and Electrical Engineering, Trinity College Dublin, Dublin, Ireland
| | | | - Michael Taynnan Barros
- School of Computer Science and Electronic Engineering, University of Essex, Colchester, UK
| |
Collapse
|
38
|
Liu H, Yi J, Zhang C, Li Y, Wang Q, Wang S, Dai S, Zheng Z, Jiang T, Gao P, Xue A, Huang Z, Kong F, Wang Y, He B, Guo X, Li Q, Chen J, Yin G, Zhao S. Macrophage GIT1 promotes oligodendrocyte precursor cell differentiation and remyelination after spinal cord injury. Glia 2024; 72:1674-1692. [PMID: 38899731 DOI: 10.1002/glia.24577] [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: 08/29/2023] [Revised: 05/02/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
Spinal cord injury (SCI) can result in severe motor and sensory deficits, for which currently no effective cure exists. The pathological process underlying this injury is extremely complex and involves many cell types in the central nervous system. In this study, we have uncovered a novel function for macrophage G protein-coupled receptor kinase-interactor 1 (GIT1) in promoting remyelination and functional repair after SCI. Using GIT1flox/flox Lyz2-Cre (GIT1 CKO) mice, we identified that GIT1 deficiency in macrophages led to an increased generation of tumor necrosis factor-alpha (TNFα), reduced proportion of mature oligodendrocytes (mOLs), impaired remyelination, and compromised functional recovery in vivo. These effects in GIT1 CKO mice were reversed with the administration of soluble TNF inhibitor. Moreover, bone marrow transplantation from GIT1 CWT mice reversed adverse outcomes in GIT1 CKO mice, further indicating the role of macrophage GIT1 in modulating spinal cord injury repair. Our in vitro experiments showed that macrophage GIT1 plays a critical role in secreting TNFα and influences the differentiation of oligodendrocyte precursor cells (OPCs) after stimulation with myelin debris. Collectively, our data uncovered a new role of macrophage GIT1 in regulating the transformation of OPCs into mOLs, essential for functional remyelination after SCI, suggesting that macrophage GIT1 could be a promising treatment target of SCI.
Collapse
Affiliation(s)
- Hao Liu
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiang Yi
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Orthopedics, Yancheng Third People's Hospital, Yancheng, Jiangsu, China
| | - Chenxi Zhang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yin Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Qian Wang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shenyu Wang
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Siming Dai
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ziyang Zheng
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Tao Jiang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Peng Gao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ao Xue
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhenfei Huang
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Fanqi Kong
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yongxiang Wang
- Department of Orthopedics, Clinical Medical College, Yangzhou University, Yangzhou, China
- Northern Jiangsu People's Hospital, Yangzhou, China
| | - Baorong He
- Department of Spine Surgery, Honghui-hospital, Xi'an Jiaotong Uinversity, School of Medicine, Xi'an, China
| | - Xiaodong Guo
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingqing Li
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jian Chen
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guoyong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shujie Zhao
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Jiangsu Institute of Functional Reconstruction and Rehabilitation, Nanjing, Jiangsu, China
- Spinal Cord Disease Research Center, Nanjing Medical University, Nanjing, Jiangsu, China
- Nash Family Department of Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
39
|
Tang Q, Ma Y, Cheng Q, Wu Y, Chen J, Du J, Lu P, Chang EY. Longitudinal Imaging of Injured Spinal Cord Myelin and White Matter with 3D Ultrashort Echo Time Magnetization Transfer (UTE-MT) and Diffusion MRI. J Imaging 2024; 10:213. [PMID: 39330433 PMCID: PMC11433189 DOI: 10.3390/jimaging10090213] [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: 07/12/2024] [Revised: 08/06/2024] [Accepted: 08/27/2024] [Indexed: 09/28/2024] Open
Abstract
Quantitative MRI techniques could be helpful to noninvasively and longitudinally monitor dynamic changes in spinal cord white matter following injury, but imaging and postprocessing techniques in small animals remain lacking. Unilateral C5 hemisection lesions were created in a rat model, and ultrashort echo time magnetization transfer (UTE-MT) and diffusion-weighted sequences were used for imaging following injury. Magnetization transfer ratio (MTR) measurements and preferential diffusion along the longitudinal axis of the spinal cord were calculated as fractional anisotropy or an apparent diffusion coefficient ratio over transverse directions. The area of myelinated white matter was obtained by thresholding the spinal cord using mean MTR or diffusion ratio values from the contralesional side of the spinal cord. A decrease in white matter areas was observed on the ipsilesional side caudal to the lesions, which is consistent with known myelin and axonal changes following spinal cord injury. The myelinated white matter area obtained through the UTE-MT technique and the white matter area obtained through diffusion imaging techniques showed better performance to distinguish evolution after injury (AUCs > 0.94, p < 0.001) than the mean MTR (AUC = 0.74, p = 0.01) or ADC ratio (AUC = 0.68, p = 0.05) values themselves. Immunostaining for myelin basic protein (MBP) and neurofilament protein NF200 (NF200) showed atrophy and axonal degeneration, confirming the MRI results. These compositional and microstructural MRI techniques may be used to detect demyelination or remyelination in the spinal cord after spinal cord injury.
Collapse
Affiliation(s)
- Qingbo Tang
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; (Q.T.); (Q.C.); (Y.W.); (J.C.); (J.D.); (P.L.)
- Department of Radiology, University of California, San Diego, CA 92093, USA;
| | - Yajun Ma
- Department of Radiology, University of California, San Diego, CA 92093, USA;
| | - Qun Cheng
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; (Q.T.); (Q.C.); (Y.W.); (J.C.); (J.D.); (P.L.)
- Department of Neuroscience, University of California, San Diego, CA 92093, USA
| | - Yuanshan Wu
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; (Q.T.); (Q.C.); (Y.W.); (J.C.); (J.D.); (P.L.)
- Department of Bioengineering, University of California, San Diego, CA 92093, USA
| | - Junyuan Chen
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; (Q.T.); (Q.C.); (Y.W.); (J.C.); (J.D.); (P.L.)
- Department of Radiology, University of California, San Diego, CA 92093, USA;
- Department of Bone and Joint Surgery, The First Affiliated Hospital, Jinan University, Guangzhou 510632, China
| | - Jiang Du
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; (Q.T.); (Q.C.); (Y.W.); (J.C.); (J.D.); (P.L.)
- Department of Radiology, University of California, San Diego, CA 92093, USA;
| | - Pengzhe Lu
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; (Q.T.); (Q.C.); (Y.W.); (J.C.); (J.D.); (P.L.)
- Department of Neuroscience, University of California, San Diego, CA 92093, USA
| | - Eric Y. Chang
- Research Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA; (Q.T.); (Q.C.); (Y.W.); (J.C.); (J.D.); (P.L.)
- Radiology Service, Veterans Affairs San Diego Healthcare System, San Diego, CA 92161, USA
| |
Collapse
|
40
|
Fukushima N, Miyamoto Y, Yamauchi J. Knockdown of Rab9 Recovers Defective Morphological Differentiation Induced by Chemical ER Stress Inducer or PMD-Associated PLP1 Mutant Protein in FBD-102b Cells. PATHOPHYSIOLOGY 2024; 31:420-435. [PMID: 39311306 PMCID: PMC11417737 DOI: 10.3390/pathophysiology31030032] [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: 05/29/2024] [Revised: 08/21/2024] [Accepted: 08/25/2024] [Indexed: 09/26/2024] Open
Abstract
Small GTP-binding proteins of the Rab family regulate intracellular vesicle trafficking across many aspects of the transport system. Among these, Rab9 is recognized for its role in controlling the transport system not only around the trans-Golgi network but also around the late endosome. However, the specific functions across different cell types and tissues remain unclear. Here, for the first time, we report that Rab9 negatively regulates morphological changes in the FBD-102b cell line, an oligodendroglial precursor cell line undergoing morphological differentiation. The knockdown of Rab9 led to an increase in cell shape alterations characterized by widespread membrane extensions. These changes were accompanied by increased expression levels of oligodendroglial cell differentiation and myelination marker proteins. Notably, the knockdown of Rab9 was capable of recovering defective cell morphological changes induced by tunicamycin, an inducer of endoplasmic reticulum (ER) stress, which is one of the major causes of oligodendroglial cell diseases such as Pelizaeus-Merzbacher disease (PMD, currently known as hypomyelinating leukodystrophy type 1 [HLD1]). In addition, Rab9 knockdown recovered levels of ER stress marker proteins and differentiation markers. Similar results were obtained in the cases of dithiothreitol (DTT), another chemical ER stress inducer, as well as HLD1-associated proteolipid protein 1 (PLP1) mutant protein. These results indicate a unique role for Rab9 in oligodendroglial cell morphological changes, suggesting its potential as a therapeutic target for mitigating diseases such as HLD1 at the molecular and cellular levels.
Collapse
Affiliation(s)
- Nana Fukushima
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (N.F.); (Y.M.)
| | - Yuki Miyamoto
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (N.F.); (Y.M.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neuroscience and Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan; (N.F.); (Y.M.)
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
41
|
Schepers J, Löser T, Behl C. Lipids and α-Synuclein: adding further variables to the equation. Front Mol Biosci 2024; 11:1455817. [PMID: 39188788 PMCID: PMC11345258 DOI: 10.3389/fmolb.2024.1455817] [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: 06/27/2024] [Accepted: 07/26/2024] [Indexed: 08/28/2024] Open
Abstract
Aggregation of alpha-Synuclein (αSyn) has been connected to several neurodegenerative diseases, such as Parkinson's disease (PD), dementia with Lewy Bodies (DLB), and multiple system atrophy (MSA), that are collected under the umbrella term synucleinopathies. The membrane binding abilities of αSyn to negatively charged phospholipids have been well described and are connected to putative physiological functions of αSyn. Consequently, αSyn-related neurodegeneration has been increasingly connected to changes in lipid metabolism and membrane lipid composition. Indeed, αSyn aggregation has been shown to be triggered by the presence of membranes in vitro, and some genetic risk factors for PD and DLB are associated with genes coding for proteins directly involved in lipid metabolism. At the same time, αSyn aggregation itself can cause alterations of cellular lipid composition and brain samples of patients also show altered lipid compositions. Thus, it is likely that there is a reciprocal influence between cellular lipid composition and αSyn aggregation, which can be further affected by environmental or genetic factors and ageing. Little is known about lipid changes during physiological ageing and regional differences of the lipid composition of the aged brain. In this review, we aim to summarise our current understanding of lipid changes in connection to αSyn and discuss open questions that need to be answered to further our knowledge of αSyn related neurodegeneration.
Collapse
Affiliation(s)
| | | | - Christian Behl
- The Autophagy Lab, Institute of Pathobiochemistry, University Medical Center of the Johannes Gutenberg University Mainz, Mainz, Germany
| |
Collapse
|
42
|
Volmer J, Cerajewski U, Alfes M, Bender J, Abert J, Schmidt C, Ott M, Hinderberger D. Aqueous Ionic Liquid Mixtures as Minimal Models of Lipid Bilayer Membranes. ACS Biomater Sci Eng 2024; 10:4802-4811. [PMID: 39066733 PMCID: PMC11322907 DOI: 10.1021/acsbiomaterials.4c00740] [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: 04/20/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 07/30/2024]
Abstract
We introduce aqueous ionic liquid (IL) mixtures, specifically mixtures of 1-butyl-3-imidazoliumtetrafluoroborate (BMImBF4), with water as a minimal model of lipid bilayer membranes. Imidazolium-based ILs are known to form clustered nanoscale structures in which local inhomogeneities, micellar or lamellar structures, are formed to shield hydrophobic parts of the cation from the polar cosolvent (water). To investigate these nanostructures, dynamic light scattering (DLS) on samples with different mixing ratios of water and BMImBF4 was performed. At mixing ratios of 50% and 45% (v/v), small and homogeneous nanostructures can indeed be detected. To test whether, in particular, these stable nanostructures in aqueous mixtures may mimic the effects of phospholipid bilayer membranes, we further investigated their interaction with myelin basic protein (MBP), a peripheral, intrinsically disordered membrane protein of the myelin sheath. Using dynamic light scattering (DLS), continuous wave (CW) and pulse electron paramagnetic resonance (EPR), and small-angle X-ray scattering (SAXS) on recombinantly produced, "healthy" charge variants rmC1WT and double cysteine variant C1S17CH85C, we find that the size and the shape of the determined nanostructures in an optimum mixture offer model membranes in which the protein exhibits native behavior. SAXS measurements illuminate the size and shape of the nanostructures and indicate IL-rich "beads" clipped together by functional MBP, one of the in vivo roles of the protein in the myelin sheath. All the gathered data combined indicate that the 50% and 45% aqueous IL mixtures can be described as offering minimal models of a lipid mono- or bilayer that allow native processing and potential study of at least peripheral membrane proteins like MBP.
Collapse
Affiliation(s)
- Jonas Volmer
- Martin
Luther University Halle-Wittenberg, Institute of Chemistry, Physical
Chemistry − Complex Self-Organizing Systems, Von-Danckelmann-Platz 4, 06120 Halle (Saale), Germany
| | - Ulrike Cerajewski
- Martin
Luther University Halle-Wittenberg, Institute of Chemistry, Physical
Chemistry − Complex Self-Organizing Systems, Von-Danckelmann-Platz 4, 06120 Halle (Saale), Germany
| | - Marie Alfes
- Interdisciplinary
Research Centre HALOmem, Institute of Biochemistry and Biotechnology,
Charles Tanford Protein Centre, Martin Luther
University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany
| | - Julian Bender
- Interdisciplinary
Research Centre HALOmem, Institute of Biochemistry and Biotechnology,
Charles Tanford Protein Centre, Martin Luther
University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany
| | - Josefin Abert
- Martin
Luther University Halle-Wittenberg, Institute of Chemistry, Physical
Chemistry − Complex Self-Organizing Systems, Von-Danckelmann-Platz 4, 06120 Halle (Saale), Germany
| | - Carla Schmidt
- Interdisciplinary
Research Centre HALOmem, Institute of Biochemistry and Biotechnology,
Charles Tanford Protein Centre, Martin Luther
University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany
- Department
of Chemistry − Biochemistry, Johannes Gutenberg University
Mainz, Biocenter II, Hanns-Dieter-Hüsch-Weg 17, 55128 Mainz, Germany
| | - Maria Ott
- Martin
Luther University Halle-Wittenberg, Institute of Biochemistry and
Biotechnology, Protein Biochemistry, Kurt-Mothes-Str. 3, 06120 Halle (Saale), Germany
| | - Dariush Hinderberger
- Martin
Luther University Halle-Wittenberg, Institute of Chemistry, Physical
Chemistry − Complex Self-Organizing Systems, Von-Danckelmann-Platz 4, 06120 Halle (Saale), Germany
- Interdisciplinary
Research Centre HALOmem, Institute of Biochemistry and Biotechnology,
Charles Tanford Protein Centre, Martin Luther
University Halle-Wittenberg, Kurt-Mothes-Str. 3a, 06120 Halle, Germany
| |
Collapse
|
43
|
Qi W, Guan W. GPR56: A potential therapeutic target for neurological and psychiatric disorders. Biochem Pharmacol 2024; 226:116395. [PMID: 38942087 DOI: 10.1016/j.bcp.2024.116395] [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: 03/21/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 06/30/2024]
Abstract
GPR56, also known as GPR56/ADGRG1, is a member of the ADGRG subgroup belonging to adhesion G protein-coupled receptors (aGPCRs). aGPCRs are the second largest subfamily of the GPCR superfamily, which is the largest family of membrane protein receptors in the human genome. Studies in recent years have demonstrated that GPR56 is integral to the normal development of the brain and functions as an important player in cortical development, suggesting that GPR56 is involved in many physiological processes. Indeed, aberrant expression of GPR56 has been implicated in multiple neurological and psychiatric disorders, including bilateral frontoparietal polymicrogyria (BFPP), depression and epilepsy. In a recent study, it was found that upregulated expression of GPR56 reduced depressive-like behaviours in an animal model of depression, indicating that GPR56 plays an important role in the antidepressant response. Given the link of GPR56 with the antidepressant response, the function of GPR56 has become a focus of research. Although GPR56 may be a potential target for the development of antidepressants, the underlying molecular mechanisms remain largely unknown. Therefore, in this review, we will summarize the latest findings of GPR56 function in neurological and psychiatric disorders (depression, epilepsy, autism, and BFPP) and emphasize the mechanisms of GPR56 in activation and signalling in those conditions. After reviewing several studies, attributing to its significant biological functions and exceptionally long extracellular N-terminus that interacts with multiple ligands, we draw a conclusion that GPR56 may serve as an important drug target for neuropsychological diseases.
Collapse
Affiliation(s)
- Wang Qi
- Department of Pharmacology, The First People's Hospital of Yancheng, China
| | - Wei Guan
- Department of Pharmacology, Pharmacy College, Nantong University, China.
| |
Collapse
|
44
|
Venati SR, Uversky VN. Exploring Intrinsic Disorder in Human Synucleins and Associated Proteins. Int J Mol Sci 2024; 25:8399. [PMID: 39125972 PMCID: PMC11313516 DOI: 10.3390/ijms25158399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 07/27/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
In this work, we explored the intrinsic disorder status of the three members of the synuclein family of proteins-α-, β-, and γ-synucleins-and showed that although all three human synucleins are highly disordered, the highest levels of disorder are observed in γ-synuclein. Our analysis of the peculiarities of the amino acid sequences and modeled 3D structures of the human synuclein family members revealed that the pathological mutations A30P, E46K, H50Q, A53T, and A53E associated with the early onset of Parkinson's disease caused some increase in the local disorder propensity of human α-synuclein. A comparative sequence-based analysis of the synuclein proteins from various evolutionary distant species and evaluation of their levels of intrinsic disorder using a set of commonly used bioinformatics tools revealed that, irrespective of their origin, all members of the synuclein family analyzed in this study were predicted to be highly disordered proteins, indicating that their intrinsically disordered nature represents an evolutionary conserved and therefore functionally important feature. A detailed functional disorder analysis of the proteins in the interactomes of the human synuclein family members utilizing a set of commonly used disorder analysis tools showed that the human α-synuclein interactome has relatively higher levels of intrinsic disorder as compared with the interactomes of human β- and γ- synucleins and revealed that, relative to the β- and γ-synuclein interactomes, α-synuclein interactors are involved in a much broader spectrum of highly diversified functional pathways. Although proteins interacting with three human synucleins were characterized by highly diversified functionalities, this analysis also revealed that the interactors of three human synucleins were involved in three common functional pathways, such as the synaptic vesicle cycle, serotonergic synapse, and retrograde endocannabinoid signaling. Taken together, these observations highlight the critical importance of the intrinsic disorder of human synucleins and their interactors in various neuronal processes.
Collapse
Affiliation(s)
- Sriya Reddy Venati
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
| | - Vladimir N. Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA;
- USF Health Byrd Alzheimer’s Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA
| |
Collapse
|
45
|
Xiao J, Zhu H, Kong W, Jiang X, Wu C, Chen JG, Li X. Stabilizing axin leads to optic nerve hypoplasia in a mouse model of autism. Exp Eye Res 2024; 245:109988. [PMID: 38964496 DOI: 10.1016/j.exer.2024.109988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/06/2024]
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopment disorders characterized by deficits in social interaction and communication, and repetitive or stereotyped behavior. Autistic children are more likely to have vision problems, and ASD is unusually common among blind people. However, the mechanisms behind the vision disorders in autism are unclear. Stabilizing WNT-targeted scaffold protein Axin2 by XAV939 during embryonic development causes overproduction of cortical neurons and leads to autistic-like behaviors in mice. In this study, we investigated the relationship between vision abnormality and autism using an XAV939-induced mouse model of autism. We found that the mice receiving XAV939 had decreased amplitude of bright light-adaptive ERG. The amplitudes and latency of flash visual evoked potential recorded from XAV939-treated mice were lower and longer, respectively than in the control mice, suggesting that XAV939 inhibits visual signal processing and conductance. Anatomically, the diameters of RGC axons were reduced when Axin2 was stabilized during the development, and the optic fibers had defective myelin sheaths and reduced oligodendrocytes. The results suggest that the WNT signaling pathway is crucial for optic nerve development. This study provides experimental evidence that conditions interfering with brain development may also lead to visual problems, which in turn might exaggerate the autistic features in humans.
Collapse
Affiliation(s)
- Jian Xiao
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Hao Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China
| | - Weixi Kong
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xuefeng Jiang
- The Third Hospital of Nanchang, Nanchang, 330000, China
| | - Chunping Wu
- The Third Hospital of Nanchang, Nanchang, 330000, China
| | - Jie-Guang Chen
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xue Li
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
46
|
Sawaguchi S, Ishida M, Miyamoto Y, Yamauchi J. Hypomyelination Leukodystrophy 16 (HLD16)-Associated Mutation p.Asp252Asn of TMEM106B Blunts Cell Morphological Differentiation. Curr Issues Mol Biol 2024; 46:8088-8103. [PMID: 39194695 DOI: 10.3390/cimb46080478] [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: 05/22/2024] [Revised: 07/18/2024] [Accepted: 07/23/2024] [Indexed: 08/29/2024] Open
Abstract
Transmembrane protein 106B (TMEM106B), which is a type II transmembrane protein, is believed to be involved in intracellular dynamics and morphogenesis in the lysosome. TMEM106B is known to be a risk factor for frontotemporal lobar degeneration and has been recently identified as the receptor needed for the entry of SARS-CoV-2, independently of angiotensin-converting enzyme 2 (ACE2). A missense mutation, p.Asp252Asn, of TMEM106B is associated with hypomyelinating leukodystrophy 16 (HLD16), which is an oligodendroglial cell-related white matter disorder causing thin myelin sheaths or myelin deficiency in the central nervous system (CNS). However, it remains to be elucidated how the mutated TMEM106B affects oligodendroglial cells. Here, we show that the TMEM106B mutant protein fails to exhibit lysosome distribution in the FBD-102b cell line, an oligodendroglial precursor cell line undergoing differentiation. In contrast, wild-type TMEM106B was indeed localized in the lysosome. Cells harboring wild-type TMEM106B differentiated into ones with widespread membranes, whereas cells harboring mutated TMEM106B failed to differentiate. It is of note that the output of signaling through the lysosome-resident mechanistic target of rapamycin (mTOR) was greatly decreased in cells harboring mutated TMEM106B. Furthermore, treatment with hesperetin, a citrus flavonoid known as an activator of mTOR signaling, restored the molecular and cellular phenotypes induced by the TMEM106B mutant protein. These findings suggest the potential pathological mechanisms underlying HLD16 and their amelioration.
Collapse
Affiliation(s)
- Sui Sawaguchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Miki Ishida
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
| | - Yuki Miyamoto
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
| | - Junji Yamauchi
- Laboratory of Molecular Neurology, Tokyo University of Pharmacy and Life Sciences, Tokyo 192-0392, Japan
- Laboratory of Molecular Pharmacology, National Research Institute for Child Health and Development, Tokyo 157-8535, Japan
- Diabetic Neuropathy Project, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-8506, Japan
| |
Collapse
|
47
|
Craig GA, Ryan L, Thapar J, McNamara NB, Hoffmann A, Page D, Rose J, Cox SR, Miron VE. Reflective imaging of myelin integrity in the human and mouse central nervous systems. Front Cell Neurosci 2024; 18:1408182. [PMID: 39049821 PMCID: PMC11266064 DOI: 10.3389/fncel.2024.1408182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/13/2024] [Indexed: 07/27/2024] Open
Abstract
The structural integrity of myelin sheaths in the central nervous system (CNS) is crucial for the maintenance of its function. Electron microscopy (EM) is the gold standard for visualizing individual myelin sheaths. However, the tissue processing involved can induce artifacts such as shearing of myelin, which can be difficult to distinguish from true myelin abnormalities. Spectral confocal reflectance (SCoRe) microscopy is an imaging technique that leverages the differential refractive indices of compacted CNS myelin in comparison to surrounding parenchyma to detect individual compact myelin internodes with reflected light, positioning SCoRe as a possible complementary method to EM to assess myelin integrity. Whether SCoRe is sensitive enough to detect losses in myelin compaction when myelin quantity is otherwise unaffected has not yet been directly tested. Here, we assess the capacity of SCoRe to detect differences in myelin compaction in two mouse models that exhibit a loss of myelin compaction without demyelination: microglia-deficient mice (Csf1r-FIRE Δ/Δ) and wild-type mice fed with the CSF1R inhibitor PLX5622. In addition, we compare the ability to detect compact myelin sheaths using SCoRe in fixed-frozen versus paraffin-embedded mouse tissue. Finally, we show that SCoRe can successfully detect individual sheaths in aged human paraffin-embedded samples of deep white matter regions. As such, we find SCoRe to be an attractive technique to investigate myelin integrity, with sufficient sensitivity to detect myelin ultrastructural abnormalities and the ability to perform equally well in tissue preserved using different methods.
Collapse
Affiliation(s)
- Georgina A. Craig
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- United Kingdom Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Lucy Ryan
- United Kingdom Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jessica Thapar
- Department of Immunology, The University of Toronto, Toronto, ON, Canada
| | - Niamh B. McNamara
- United Kingdom Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Alana Hoffmann
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- United Kingdom Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Danielle Page
- Lothian Birth Cohorts, The University of Edinburgh, Edinburgh, United Kingdom
| | - Jamie Rose
- United Kingdom Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Simon R. Cox
- Lothian Birth Cohorts, The University of Edinburgh, Edinburgh, United Kingdom
| | - Veronique E. Miron
- Keenan Research Centre for Biomedical Science, St. Michael’s Hospital, Unity Health Toronto, Toronto, ON, Canada
- United Kingdom Dementia Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
- Department of Immunology, The University of Toronto, Toronto, ON, Canada
| |
Collapse
|
48
|
Isasi E, Wajner M, Duarte JA, Olivera-Bravo S. Cerebral White Matter Alterations Associated With Oligodendrocyte Vulnerability in Organic Acidurias: Insights in Glutaric Aciduria Type I. Neurotox Res 2024; 42:33. [PMID: 38963434 DOI: 10.1007/s12640-024-00710-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 04/27/2024] [Accepted: 06/14/2024] [Indexed: 07/05/2024]
Abstract
The white matter is an important constituent of the central nervous system, containing axons, oligodendrocytes, and its progenitor cells, astrocytes, and microglial cells. Oligodendrocytes are central for myelin synthesis, the insulating envelope that protects axons and allows normal neural conduction. Both, oligodendrocytes and myelin, are highly vulnerable to toxic factors in many neurodevelopmental and neurodegenerative disorders associated with disturbances of myelination. Here we review the main alterations in oligodendrocytes and myelin observed in some organic acidurias/acidemias, which correspond to inherited neurometabolic disorders biochemically characterized by accumulation of potentially neurotoxic organic acids and their derivatives. The yet incompletely understood mechanisms underlying the high vulnerability of OLs and/or myelin in glutaric acidemia type I, the most prototypical cerebral organic aciduria, are particularly discussed.
Collapse
Affiliation(s)
- Eugenia Isasi
- Laboratorio de Neurobiología Celular y Molecular, Unidad Académica de Histología y Embriología, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
- Departamento de Neurobiología y Neuropatología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay
| | - Moacir Wajner
- Department of Biochemistry, Instituto de Ciencias Básicas da Saude, Universidade Federal de Río Grande do Sul, Porto Alegre, Brazil
- Medical Genetics Service, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Juliana Avila Duarte
- Departamento de Medicina Interna, Serviço de Radiología, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| | - Silvia Olivera-Bravo
- Departamento de Neurobiología y Neuropatología, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay.
| |
Collapse
|
49
|
Papadopoulou M, Tzanetakos D, Moschovos C, Korona A, Vartzelis G, Voudris K, Fanouraki S, Dimitriadou EM, Papadimas G, Tzartos JS, Giannopoulos S, Tsivgoulis G. Combined Central and Peripheral Demyelination (CCPD) Associated with MOG Antibodies: Report of Four New Cases and Narrative Review of the Literature. J Clin Med 2024; 13:3604. [PMID: 38930142 PMCID: PMC11204739 DOI: 10.3390/jcm13123604] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024] Open
Abstract
Background/Objectives: Myelin oligodendrocyte glycoprotein (MOG) is exclusively expressed in the central nervous system (CNS) and is found on the outer surface of oligodendrocytes. Antibodies to MOG are associated with CNS demyelination, whereas peripheral nervous system (PNS) demyelination is seldom reported to be related to MOG-IgG. Methods: The database of patients seen in our neurological academic center was searched for MOG-IgG seropositivity and concomitant demyelinating polyneuropathy. For the purpose of the review, in March 2024, we searched for case reports and case series in the following databases: PubMed, Scopus, Cochrane, and ScienceDirect. Inclusion criteria were MOG-IgG seropositivity and demyelinating polyneuropathy. Exclusion criteria were type of publication other than case reports and case series, unconfirmed diagnosis of demyelinating polyneuropathy, and other diseases causing demyelination in either the CNS or PNS. Critical appraisal of the selected case reports and case series was realized by JBI. Results: Four new cases were identified with MOG-IgG and confirmed demyelinating polyneuropathy. This review identified 22 cases that have been published since 2018. Clinical, imaging, neurophysiological, and immunological characteristics, as well as treatment options and outcomes are presented and compared to those of other cases with combined central and peripheral demyelination (CCPD). Conclusions: The pathogenetic mechanism is unclear; thus, different hypotheses are discussed. New case reporting and large cohort studies will help further the exploration of the underlying mechanism and guide more effective therapeutic interventions.
Collapse
Affiliation(s)
- Marianna Papadopoulou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (M.P.); (D.T.); (C.M.); (S.F.); (E.-M.D.); (J.S.T.); (G.T.)
- Department of Physiotherapy, University of West Attica, Ag. Spyridonos Str., 12243 Athens, Greece
| | - Dimitrios Tzanetakos
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (M.P.); (D.T.); (C.M.); (S.F.); (E.-M.D.); (J.S.T.); (G.T.)
| | - Christos Moschovos
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (M.P.); (D.T.); (C.M.); (S.F.); (E.-M.D.); (J.S.T.); (G.T.)
| | - Anastasia Korona
- Department of Neurology, Children’s Hospital of Athens “P. & A. Kyriakou”, 11527 Athens, Greece; (A.K.); (K.V.)
| | - George Vartzelis
- Second Department of Pediatrics, Children’s Hospital ‘P. & A. Kyriakou’, School of Medicine, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Konstantinos Voudris
- Department of Neurology, Children’s Hospital of Athens “P. & A. Kyriakou”, 11527 Athens, Greece; (A.K.); (K.V.)
| | - Stella Fanouraki
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (M.P.); (D.T.); (C.M.); (S.F.); (E.-M.D.); (J.S.T.); (G.T.)
| | - Evangelia-Makrina Dimitriadou
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (M.P.); (D.T.); (C.M.); (S.F.); (E.-M.D.); (J.S.T.); (G.T.)
| | - Georgios Papadimas
- First Department of Neurology, National and Kapodistrian University of Athens, Eginition University Hospital School of Medicine, 11528 Athens, Greece;
| | - John S. Tzartos
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (M.P.); (D.T.); (C.M.); (S.F.); (E.-M.D.); (J.S.T.); (G.T.)
| | - Sotirios Giannopoulos
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (M.P.); (D.T.); (C.M.); (S.F.); (E.-M.D.); (J.S.T.); (G.T.)
| | - Georgios Tsivgoulis
- Second Department of Neurology, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (M.P.); (D.T.); (C.M.); (S.F.); (E.-M.D.); (J.S.T.); (G.T.)
- Department of Neurology, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
50
|
Cohen Z, Lau L, Ahmed M, Jack CR, Liu C. Quantitative susceptibility mapping in the brain reflects spatial expression of genes involved in iron homeostasis and myelination. Hum Brain Mapp 2024; 45:e26688. [PMID: 38896001 PMCID: PMC11187871 DOI: 10.1002/hbm.26688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/02/2024] [Accepted: 04/05/2024] [Indexed: 06/21/2024] Open
Abstract
Quantitative susceptibility mapping (QSM) is an MRI modality used to non-invasively measure iron content in the brain. Iron exhibits a specific anatomically varying pattern of accumulation in the brain across individuals. The highest regions of accumulation are the deep grey nuclei, where iron is stored in paramagnetic molecule ferritin. This form of iron is considered to be what largely contributes to the signal measured by QSM in the deep grey nuclei. It is also known that QSM is affected by diamagnetic myelin contents. Here, we investigate spatial gene expression of iron and myelin related genes, as measured by the Allen Human Brain Atlas, in relation to QSM images of age-matched subjects. We performed multiple linear regressions between gene expression and the average QSM signal within 34 distinct deep grey nuclei regions. Our results show a positive correlation (p < .05, corrected) between expression of ferritin and the QSM signal in deep grey nuclei regions. We repeated the analysis for other genes that encode proteins thought to be involved in the transport and storage of iron in the brain, as well as myelination. In addition to ferritin, our findings demonstrate a positive correlation (p < .05, corrected) between the expression of ferroportin, transferrin, divalent metal transporter 1, several gene markers of myelinating oligodendrocytes, and the QSM signal in deep grey nuclei regions. Our results suggest that the QSM signal reflects both the storage and active transport of iron in the deep grey nuclei regions of the brain.
Collapse
Affiliation(s)
- Zoe Cohen
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Laurance Lau
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Maruf Ahmed
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Clifford R. Jack
- Mayo Foundation for Medical Education and ResearchRochesterMinnesotaUSA
| | - Chunlei Liu
- Department of Electrical Engineering and Computer SciencesUniversity of California, BerkeleyBerkeleyCaliforniaUSA
- Helen Wills Neuroscience InstituteUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| |
Collapse
|