1
|
Thiesler H, Hildebrandt H. Polysialic acid-Siglec immune checkpoints of microglia and macrophages: Perspectives for therapeutic intervention. Neural Regen Res 2026; 21:661-662. [PMID: 39688555 DOI: 10.4103/nrr.nrr-d-24-01195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Affiliation(s)
- Hauke Thiesler
- Institute of Clinical Biochemistry, Hannover Medical School and Center for Systems Neuroscience Hannover, Hannover, Germany
| | | |
Collapse
|
2
|
Badina AM, Ceyzériat K, Amossé Q, Tresh A, Abjean L, Guénat L, Vauthey E, Tsartsalis S, Millet P, Tournier BB. Non-linear microglial, inflammatory and oligodendrocyte dynamics across stages of Alzheimer's disease. Neurobiol Dis 2025; 211:106950. [PMID: 40348203 DOI: 10.1016/j.nbd.2025.106950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2025] [Revised: 04/24/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025] Open
Abstract
Alzheimer's disease (AD) is characterized by cognitive decline and neuropathological hallmarks including Aβ plaques and Tau tangles. Emerging evidence indicates oligodendrocyte (OL) dysfunction and demyelination also contribute to disease progression. Here, we analyzed OL markers and inflammatory gene expression in human hippocampal samples at early and late AD stages. In early AD, we observed OL and myelinating pathways downregulation, alongside microglial and astrocytic activation, as well as upregulated chemokine CCL2 and peripheral immune infiltration markers. In late stages, expression of OL-related genes and myelination pathways increase, with a higher NG2/MBP ratio, coinciding with decreased microglial coverage and peripheral immune markers. These findings indicate that early neuroinflammation may impair OL function, while attenuated immune activity in late AD allows partial OL recovery. This study provides insights into stage-specific inflammatory and myelin-related changes in AD, supporting the relevance of understanding oligodendrocyte dynamics and potential regenerative responses for future therapeutic strategies.
Collapse
Affiliation(s)
- Aurélien M Badina
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Quentin Amossé
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Alexandre Tresh
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Laurene Abjean
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Léa Guénat
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Emilie Vauthey
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Stergios Tsartsalis
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Philippe Millet
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland
| | - Benjamin B Tournier
- Department of Psychiatry, Faculty of Medicine, University of Geneva, Geneva, Switzerland; Department of Psychiatry, University Hospitals of Geneva, Geneva, Switzerland.
| |
Collapse
|
3
|
Stadelmann C, Franz J, Nessler S. Recent developments in multiple sclerosis neuropathology. Curr Opin Neurol 2025; 38:173-179. [PMID: 40178490 PMCID: PMC12052066 DOI: 10.1097/wco.0000000000001370] [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: 04/05/2025]
Abstract
PURPOSE OF REVIEW Neuropathological studies in human brain tissue are indispensable for our understanding of disease mechanisms in multiple sclerosis (MS). They inform concepts of lesion evolution, tissue regeneration and disease progression, and ideally reveal new disease mechanisms and therapeutic targets. Here we review recent neuropathological studies that have advanced our knowledge of MS pathogenesis. RECENT FINDINGS Recent cohort studies support the notion that different clinical MS disease phenotypes share underlying pathological features, and that clinical and pathological heterogeneity is derived from a variable combination of innate and adaptive inflammation, demyelinating activity, and neuroaxonal loss. Importantly, emerging technologies for spatial transcriptome analysis enable an unprecedented glimpse into the cellular composition and molecular mechanisms involved in lesion evolution. These promising technologies will help identify the identification of molecular hubs governing tissue damage and regeneration. SUMMARY Recent neuropathological studies helped to identify tissue correlates of disability and disease progression. Substantial progress in molecular brain tissue analysis revealed the complexity of MS-related tissue features. Close collaboration between tissue-based, molecular, bioinformatic, pharmacologic, imaging and clinical experts is needed to continue to advance the field, particularly for the benefit of people with progressive MS.
Collapse
Affiliation(s)
- Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen, Germany
| | | | | |
Collapse
|
4
|
Gorter RP, Arreguin AJ, Oost W, de Jonge JC, Kampinga HH, Amor S, Colognato H, Baron W. Unexpected Remyelination in the Absence of Matrix Metalloproteinase 7. Glia 2025; 73:1288-1306. [PMID: 40059514 PMCID: PMC12012326 DOI: 10.1002/glia.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 04/23/2025]
Abstract
In multiple sclerosis (MS), an influx of immune cells into the central nervous system leads to focal demyelinating lesions in the brain, optic nerve, and spinal cord. As MS progresses, remyelination increasingly fails, leaving neuronal axons vulnerable to degeneration and resulting in permanent neurological disability. In chronic MS lesions, the aberrant accumulation of extracellular matrix (ECM) molecules, including fibronectin and hyaluronan, impairs oligodendrocyte progenitor cell differentiation, contributing to remyelination failure. Removing inhibitory ECM is therefore a therapeutic target to stimulate remyelination in MS. Intriguingly, the expression of the fibronectin-degrading enzyme matrix metalloproteinase 7 (MMP7) is decreased in chronic MS lesions compared to control white matter. Therefore, we examined the role of MMP7 upon cuprizone-induced demyelination, hypothesizing that the lack of MMP7 would lead to impaired breakdown of its ECM substrates, including fibronectin, and diminished remyelination. Unexpectedly, remyelination proceeded efficiently in the absence of MMP7. In the remyelination phase, the lack of MMP7 did not lead to the accumulation of fibronectin or of laminin, another MMP7 substrate. Moreover, in the setting of chronic demyelination, levels of fibronectin were actually lower in MMP7-/- mice, while levels of hyaluronan, which is not a known MMP7 substrate, were also lower. Overall, these results indicate that MMP7 is not essential for remyelination in the cuprizone model and point to an unexpected complexity in how MMP7 deficiency influences fibronectin and hyaluronan levels in chronic demyelination.
Collapse
Affiliation(s)
- Rianne P. Gorter
- Department of Biomedical SciencesUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
- Queen Mary University of LondonMS Center Noord NederlandGroningenthe Netherlands
| | - Andrea J. Arreguin
- Department of Pharmacological SciencesStony Brook UniversityStony BrookNew YorkUSA
| | - Wendy Oost
- Department of Biomedical SciencesUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
- Queen Mary University of LondonMS Center Noord NederlandGroningenthe Netherlands
| | - Jenny C. de Jonge
- Department of Biomedical SciencesUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
- Queen Mary University of LondonMS Center Noord NederlandGroningenthe Netherlands
| | - Harm H. Kampinga
- Department of Biomedical SciencesUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
| | - Sandra Amor
- Department of PathologyAmsterdam UMC‐Location VUmcAmsterdamthe Netherlands
- Blizard Institute, Barts and the London School of Medicine and DentistryDepartment of Neuroscience and TraumaQueen Mary University of LondonLondonUK
- Institute of AnatomyRostock University Medical CenterRostockGermany
| | - Holly Colognato
- Department of Pharmacological SciencesStony Brook UniversityStony BrookNew YorkUSA
| | - Wia Baron
- Department of Biomedical SciencesUniversity of Groningen, University Medical Center GroningenGroningenthe Netherlands
- Queen Mary University of LondonMS Center Noord NederlandGroningenthe Netherlands
| |
Collapse
|
5
|
Nottar Escobar EL, De Silva Mohotti N, Manolescu M, Radadiya A, Dhar P, Hartley MD. Reduced cholesterol alters the biophysical properties of repaired myelin. Biochim Biophys Acta Mol Cell Biol Lipids 2025; 1870:159637. [PMID: 40403837 DOI: 10.1016/j.bbalip.2025.159637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 05/02/2025] [Accepted: 05/19/2025] [Indexed: 05/24/2025]
Abstract
The myelin sheath is a lipid-rich membrane that ensheathes axons and is required for healthy and efficient signal transduction. Myelin is damaged in neurological diseases like multiple sclerosis, but remyelination can occur through the action of oligodendrocyte precursor cells (OPCs), which differentiate into mature oligodendrocytes that wrap axons to form repaired myelin. In this study, a genetic-based mouse model of demyelination was used, which features near-complete demyelination followed by robust remyelination in the brain. Lipid mass spectrometry on isolated myelin from the remyelinated brain revealed a decrease in the percent mole fraction of cholesterol when compared to healthy myelin. Biophysical studies on monomolecular lipid films formed using repaired myelin lipid extracts showed changes in the surface behavior of the lipid films, compared to the healthy myelin lipids. Films formed using the remyelinated lipid extracts resulted in lower surface pressures and lower compressional moduli when compared to healthy controls, suggesting that repaired myelin membranes have lower lateral molecular packing within the lipid film. Synthetically prepared model membranes, based on the major lipid compositions of the healthy and diseased extracts, revealed that changes in cholesterol levels were the primary contributor to the changes in biophysical properties. Supplementation of the diseased lipid extracts with cholesterol led to a robust improvement in membrane surface pressures and compressibility. Together, these results suggest that high cholesterol levels are required for myelin membrane stability and that reduced cholesterol in repaired myelin may have a profound impact on the biophysical properties of the myelin membrane.
Collapse
Affiliation(s)
| | | | - Mara Manolescu
- Department of Chemical Engineering, University of Kansas, Lawrence, KS, USA
| | - Anika Radadiya
- Department of Chemical Engineering, University of Kansas, Lawrence, KS, USA
| | - Prajnaparamita Dhar
- Department of Chemical Engineering, University of Kansas, Lawrence, KS, USA.
| | - Meredith D Hartley
- 2030 Becker Drive, Department of Chemistry, University of Kansas, Lawrence, KS 66047, USA.
| |
Collapse
|
6
|
Murayama R, Cai Y, Nakamura H, Hashimoto K. Demyelination in psychiatric and neurological disorders: Mechanisms, clinical impact, and novel therapeutic strategies. Neurosci Biobehav Rev 2025; 174:106209. [PMID: 40368261 DOI: 10.1016/j.neubiorev.2025.106209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 04/14/2025] [Accepted: 05/10/2025] [Indexed: 05/16/2025]
Abstract
Demyelination, defined as the loss of myelin sheaths around neuronal axons, is increasingly recognized as a key factor in a broad range of psychiatric and neurological disorders, including schizophrenia, major depressive disorder, bipolar disorder, post-traumatic stress disorder, autism spectrum disorder, substance use disorders, Alzheimer's disease, Parkinson's disease, and multiple sclerosis. This review investigates the core mechanisms driving demyelination, its clinical impact, and emerging therapeutic strategies aimed at maintaining or restoring myelin integrity. Disruption of myelin impairs crucial neural communication pathways, resulting in cognitive, motor, and behavioral deficits that substantially reduce quality of life and create significant economic and social challenges. Key contributors to demyelination include genetic predisposition, environmental triggers, immune dysregulation, neuroinflammation, and alterations in the gut-brain axis mediated by the vagus nerve. Promising therapies include sphingosine 1-phosphate receptor modulators and muscarinic acetylcholine receptor antagonists, both of which diminish immune-related myelin damage and may enhance neuroprotection. In addition, the novel antidepressant arketamine appears to boost myelination through transforming growth factor-β1 signaling pathways. Approaches targeting the gut-brain axis, such as noninvasive transcutaneous auricular vagus nerve stimulation and fecal microbiota transplantation, may also help reduce inflammation and support myelin repair. Future research should center on clarifying the precise molecular mechanisms of demyelination, developing targeted therapies, and leveraging advanced neuroimaging for earlier detection and personalized treatment. By combining immunomodulatory and neuroprotective strategies, there is potential to significantly improve outcomes for individuals affected by demyelinating psychiatric and neurological disorders.
Collapse
Affiliation(s)
- Rumi Murayama
- Laboratory of Chemical Pharmacology, Chiba University Graduate School of Pharmaceutical Sciences, Chiba 260-8675, Japan; Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Yi Cai
- Laboratory of Chemical Pharmacology, Chiba University Graduate School of Pharmaceutical Sciences, Chiba 260-8675, Japan; Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Hiroyuki Nakamura
- Laboratory of Chemical Pharmacology, Chiba University Graduate School of Pharmaceutical Sciences, Chiba 260-8675, Japan
| | - Kenji Hashimoto
- Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan.
| |
Collapse
|
7
|
Gluck L, Gerstein B, Kaunzner UW. Repair mechanisms of the central nervous system: From axon sprouting to remyelination. Neurotherapeutics 2025:e00583. [PMID: 40348704 DOI: 10.1016/j.neurot.2025.e00583] [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: 11/30/2024] [Revised: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 05/14/2025] Open
Abstract
The central nervous system (CNS), comprising the brain, spinal cord, and optic nerve, has limited regenerative capacity, posing significant challenges in treating neurological disorders. Recent advances in neuroscience and neurotherapeutics have introduced promising strategies to stimulate CNS repair, particularly in the context of neurodegenerative diseases such as multiple sclerosis. This review explores the complex interplay between inflammation, demyelination, and remyelination possibilities. Glial cells, including oligodendrocyte precursors, oligodendrocytes, astrocytes and microglia play dual roles in injury response, with reactive gliosis promoting repair but also potentially inhibiting recovery through glial scar formation. There is also an emphasis on axonal regeneration, axonal sprouting and stem cell therapies. We highlight the role of neuroplasticity in recovery post-injury and the limited regenerative potential of axons in the CNS due to inhibitory factors such as myelin-associated inhibitors. Moreover, neurotrophic factors support neuronal survival and axonal growth, while stem cell-based approaches offer promise for replacing lost neurons and glial cells. However, challenges such as stem cell survival, integration, and risk of tumor formation remain. Furthermore, we examine the role of neurogenesis in CNS repair and the remodeling of the extracellular matrix, which can facilitate regeneration. Through these diverse mechanisms, ongoing research aims to overcome the intrinsic and extrinsic barriers to CNS repair and advance therapeutic strategies for neurological diseases.
Collapse
Affiliation(s)
- Lauren Gluck
- Montefiore Medical Center, 1250 Waters Place Tower 2, Bronx, NY 10461, USA.
| | - Brittany Gerstein
- Weill-Cornell-Medicine, Department of Neurology, 1305 York Avenue, New York City, 10021, USA.
| | - Ulrike W Kaunzner
- Weill-Cornell-Medicine, Department of Neurology, 1305 York Avenue, New York City, 10021, USA.
| |
Collapse
|
8
|
Bu J, Liu Y, Zhao Y, Liu L, Shen J, Li Y. Paroxetine ameliorates corticosterone-induced myelin damage by promoting the proliferation and differentiation of oligodendrocyte precursor cells. Neuroscience 2025; 573:344-354. [PMID: 40164278 DOI: 10.1016/j.neuroscience.2025.03.065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/22/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Depression is frequently associated with demyelination in the prefrontal cortex (PFC), and promoting remyelination can improve neuronal signaling and alleviate depressive symptoms. Paroxetine, a classic selective serotonin reuptake inhibitor (SSRI), is known to exert its antidepressant effects by increasing serotonin levels. However, its potential to alleviate myelin damage in depression remains unclear. A corticosterone (CORT)-induced mouse model of depression was used in this study. Myelin staining and transmission electron microscopy (TEM) were employed to assess myelin damage in the PFC, while immunofluorescence and western blot were performed to evaluate the expression of myelin-associated proteins. The primary oligodendrocyte precursor cells (OPCs) were cultured in vitro. The results demonstrated that paroxetine significantly alleviated CORT-induced depressive-like behaviors, including increased sucrose preference and spontaneous activity in the open field, while reduced immobility time in the tail suspension and forced swimming tests. Paroxetine also increased myelin thickness and restoring myelin integrity in the PFC. Moreover, paroxetine upregulated the expression of MBP, MAG, and neurofilament light chain protein (NFEL). Immunofluorescence analysis that paroxetine significantly increased the number of OPC (Olig2+/NG2+) and promoted OL differentiation (Olig2+/CC-1+), as well as upregulating the expression of PDGFRα. BrdU assays further confirmed that paroxetine enhanced OPC proliferation. In vitro, paroxetine significantly increased the viability of primary OPCs and promoted their proliferation and differentiation, with the most potent effect observed at 20 nM. These findings suggest that paroxetine alleviates CORT-induced myelin damage and improves depressive-like behaviors by promoting OPC proliferation and differentiation, providing new insights into its antidepressant mechanisms.
Collapse
Affiliation(s)
- Jingjing Bu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao in Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yuan Liu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao in Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yufang Zhao
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao in Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Liming Liu
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao in Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Jiduo Shen
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao in Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, China; College of Pharmacy, Henan University of Chinese Medicine, Zhengzhou 450046, China
| | - Yucheng Li
- Collaborative Innovation Center of Research and Development on the Whole Industry Chain of Yu-Yao in Henan Province, Henan University of Chinese Medicine, Zhengzhou 450046, China; Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou 450046, China.
| |
Collapse
|
9
|
Wang J, Gao Y, Wang B, Zhang C, Yuan Y, Xu R, Ji H, Zhang X. Low-Intensity Pulsed Ultrasound Promotes Oligodendrocyte Maturation and Remyelination by Down-regulating the Interleukin-17A/Notch1 Signaling Pathway in Mice with Ischemic Stroke. RESEARCH (WASHINGTON, D.C.) 2025; 8:0676. [PMID: 40290135 PMCID: PMC12022504 DOI: 10.34133/research.0676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 03/22/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025]
Abstract
Increasing evidence indicates that oligodendrocyte (OL) numbers and myelin as a dynamic cellular compartment perform a key role in the maintenance of neuronal function. Inhibiting white matter (WM) demyelination or promoting remyelination has garnered interest for its potential therapeutic strategy against ischemic stroke. Our previous work has shown that low-intensity pulsed ultrasound (LIPUS) could improve stroke recovery. However, it is unclear whether LIPUS can maintain WM integrity early after stroke or promote late WM repair. This study evaluated the efficacy of LIPUS on WM repair and long-term neurologic recovery after stroke. Male adult C57BL/6 mice underwent a focal cerebral ischemia model and were randomized to receive ultrasound stimulation (30 min once daily for 14 days). The effect of LIPUS on sensorimotor function was assessed by modified neurological severity score, rotarod test, grip strength test, and gait analysis up to 28 days after stroke. We found that ischemic stroke-induced WM damage was severe on day 7 and partially recovered on day 28. LIPUS prevented neuronal and oligodendrocyte progenitor cell (OPC) death during the acute phase of stroke (d7), protected WM integrity, and reduced brain atrophy and tissue damage during the recovery phase (d28). To further confirm the effect of LIPUS on remyelination, we assessed the proliferation and differentiation of OPCs. We found that LIPUS did not increase the number of OPCs (PDGFRα+ or NG2+), but markedly increased the number of newly produced mature OLs (APC+) and myelin protein levels. Mechanistically, LIPUS may promote OL maturation and remyelination by down-regulating the interleukin-17A/Notch1 signaling pathway. In summary, LIPUS can protect OLs and neurons early after stroke and promote long-term WM repair and functional recovery. LIPUS will be a viable strategy for the treatment of ischemic stroke in the future.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Yuxiao Gao
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Bin Wang
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Cong Zhang
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Yi Yuan
- School of Electrical Engineering,
Yanshan University, Qinhuangdao 066004, China
- Key Laboratory of Intelligent Rehabilitation and Neuromodulation of Hebei Province,
Yanshan University, Qinhuangdao 066004, China
| | - Renhao Xu
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Hui Ji
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| | - Xiangjian Zhang
- Department of Neurology,
Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050000, China
- Hebei Key Laboratory of Vascular Homeostasis and Hebei Collaborative Innovation Center for Cardio-cerebrovascular Disease, Shijiazhuang, Hebei 050000, China
- Key Laboratory of Clinical Neurology (Hebei Medical University), Ministry of Education, Shijiazhuang, Hebei 050000, China
| |
Collapse
|
10
|
Yuan J, Tao Y, Wang M, Chen Y, Han X, Wu H, Shi H, Huang F, Wu X. Astragaloside II, a natural saponin, facilitates remyelination in demyelination neurological diseases via p75NTR receptor mediated β-catenin/Id2/MBP signaling axis in oligodendrocyte precursor cells. J Adv Res 2025:S2090-1232(25)00273-5. [PMID: 40258474 DOI: 10.1016/j.jare.2025.04.028] [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: 12/18/2024] [Revised: 04/15/2025] [Accepted: 04/18/2025] [Indexed: 04/23/2025] Open
Abstract
BACKGROUND Demyelination is a hallmark of neurological disorders such as multiple sclerosis and neuromyelitis optica, leading to neurological deficits. Existing therapies primarily modulate immune responses but lack efficacy in directly promoting myelin repair. Enhancing oligodendrocyte precursor cell (OPC) differentiation and oligodendrocytes (OLs) production is crucial for restoring myelin integrity. OBJECTIVES This study investigated the therapeutic potential of astragaloside II (AS-II), a bioactive saponin with neuroprotective and pro-differentiation properties, derived from Astragalus membranaceus, uniquely in promoting OPC differentiation and myelin endogenous repair, distinguishing it from existing immunomodulatory treatments. AS-II directly targets p75 neurotrophin receptor (p75NTR) signaling, a pathway linked to myelin regeneration but underestimated in current remyelination strategies. METHODS We conducted in vitro OPC differentiation assays and in vivo demyelination models, including cuprizone and experimental autoimmune encephalomyelitis. Drug affinity responsive target stability mass spectrometry, cellular thermal shift assay, and surface plasmon resonance assays identified and validated p75NTR as the direct target of AS-II. p75NTR knockout mice and lentiviral transduction were used to confirm its role. RESULTS AS-II improved neurobehavioral outcomes, increased OLs production, and enhanced myelin integrity by suppressing β-catenin/Id2/MBP signaling. Mechanistically, AS-II bound to p75NTR (Pro253, Ser257), stabilizing its structure and promoting remyelination. In p75NTR knockout mice, AS-II failed to restore myelin or neural function, confirming its p75NTR-dependent mechanism. CONCLUSION AS-II represents a novel therapeutic candidate for demyelinating diseases, offering a targeted approach to myelin regeneration through direct p75NTR modulation and addressing gaps in current treatment strategies.
Collapse
Affiliation(s)
- Jinfeng Yuan
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yanlin Tao
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Institute for Translational Brain Research, Fudan University, Shanghai 200433, China
| | - Mengxue Wang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yufeng Chen
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xinyan Han
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Hui Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Hailin Shi
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Fei Huang
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiaojun Wu
- Shanghai Key Laboratory of Compound Chinese Medicines, The Ministry of Education (MOE) Key Laboratory for Standardization of Chinese Medicines, The MOE Innovation Center for Basic Medicine Research on Qi-Blood TCM Theories, Institute of Chinese Materia Medica, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
11
|
Chen Z, Liu L, Guo X, Zhang Y, Zhong M, Xu Y, Peng T, Peng T, Zhang Y, Hou Q, Fan D, Gao T, He L, Tang H, Hu H, Xu K. Upregulating mTOR/S6 K Pathway by CASTOR1 Promotes Astrocyte Proliferation and Myelination in Gpam -/--induced mouse model of cerebral palsy. Mol Neurobiol 2025:10.1007/s12035-025-04901-w. [PMID: 40234290 DOI: 10.1007/s12035-025-04901-w] [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: 04/20/2024] [Accepted: 03/27/2025] [Indexed: 04/17/2025]
Abstract
GPAM, a key enzyme for lipid synthesis, is predominantly expressed in astrocytes (ASTs), where it facilitates lipid supply for myelin formation. Our previous studies identified GPAM as a novel causative gene for cerebral palsy (CP) and led to the development of a CP mouse model with GPAM deficiency (Gpam-/-). The model closely recapitulated the clinical phenotype of children with CP, due to the restricted proliferation of ASTs in the brain, reduced the amount of lipid, thinner brain white matter, and myelin dysplasia. The mammalian target of rapamycin (mTOR) pathway plays an important role in cell proliferation and lipid synthesis. Cytosolic arginine sensor (CASTOR1) interacts with GATOR2 to regulate mTOR complex 1 (mTORC1). Targeted degradation of CASTOR1 can activate the mTOR pathway. However, it remains unclear the involvement of mTOR pathway in neurological diseases such as CP. In this study, we demonstrated that the mTOR pathway was inhibited in Gpam-/- mice. Notably, CASTOR1 could regulate the activity of mTOR/S6K pathway, functioning as a negative upstream regulator. Furthermore, inhibition of CASTOR1 upregulated mTOR/S6K signaling, promoting astrocyte proliferation and myelination, which in turn enhanced motor function in the Gpam-/--induced CP mouse model. Collectively, these findings reveal the role of astrocytic mTOR in the pathogenesis of CP mice, broaden the therapeutic strategies, and provide a promising candidate target for CP treatment.
Collapse
Affiliation(s)
- Zhaofang Chen
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
| | - Liru Liu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
| | - Xiaolin Guo
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, 200438, China
| | - Yage Zhang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
| | - Mengru Zhong
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
| | - Yi Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
- Department of Sports and Health, Guangzhou Sport University, Guangzhou, 510500, China
| | - Tingting Peng
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
| | - Tingting Peng
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
| | - Yuan Zhang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
- Department of Sport Rehabilitation, Shanghai University of Sport, Shanghai, 200438, China
| | - Qingfen Hou
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
- Department of Sports and Health, Guangzhou Sport University, Guangzhou, 510500, China
| | - Danxia Fan
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
- School of Nursing, Guangdong Pharmaceutical University, Guangzhou, 510310, China
| | - Ting Gao
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
| | - Lu He
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
| | - Hongmei Tang
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China
| | - Hao Hu
- Laboratory of Medical Systems Biology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Kaishou Xu
- Department of Rehabilitation, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510120, China.
| |
Collapse
|
12
|
Hill BM, Holloway RK, Forbes LH, Davies CL, Monteiro JK, Brown CM, Rose J, Fudge N, Plant PJ, Mahmood A, Brand-Arzamendi K, Kent SA, Molina-Gonzalez I, Gyoneva S, Ransohoff RM, Wipke B, Priller J, Schneider R, Moore CS, Miron VE. Monocyte-secreted Wnt reduces the efficiency of central nervous system remyelination. PLoS Biol 2025; 23:e3003073. [PMID: 40233100 PMCID: PMC12052099 DOI: 10.1371/journal.pbio.3003073] [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: 01/11/2024] [Revised: 05/05/2025] [Accepted: 02/18/2025] [Indexed: 04/17/2025] Open
Abstract
The regeneration of myelin in the central nervous system (CNS) reinstates nerve health and function, yet its decreased efficiency with aging and progression of neurodegenerative disease contributes to axonal loss and/or degeneration. Although CNS myeloid cells have been implicated in regulating the efficiency of remyelination, the distinct contribution of blood monocytes versus that of resident microglia is unclear. Here, we reveal that monocytes have non-redundant functions compared to microglia in regulating remyelination. Using a transgenic mouse in which classical monocytes have reduced egress from bone marrow (Ccr2-/-), we demonstrate that monocytes drive the timely onset of oligodendrocyte differentiation and myelin protein expression, yet impede myelin production. Ribonucleic acid sequencing revealed a Wnt signature in wild-type mouse lesion monocytes, which was confirmed in monocytes from multiple sclerosis white matter lesions and blood. Genetic or pharmacological inhibition of Wnt release by monocytes increased remyelination. Our findings reveal monocytes to be critical regulators of remyelination and identify monocytic Wnt signaling as a promising therapeutic target to inhibit for increased efficiency of CNS regeneration.
Collapse
Affiliation(s)
- Bianca M. Hill
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Rebecca K. Holloway
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Lindsey H. Forbes
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Claire L. Davies
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Jonathan K. Monteiro
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
| | - Christina M. Brown
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Jamie Rose
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Neva Fudge
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Pamela J. Plant
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Ayisha Mahmood
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Koroboshka Brand-Arzamendi
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Sarah A. Kent
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Irene Molina-Gonzalez
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Stefka Gyoneva
- Previously at Biogen Ltd, Cambridge, Massachusetts, United States of America
| | - Richard M. Ransohoff
- Previously at Biogen Ltd, Cambridge, Massachusetts, United States of America
- Third Rock Ventures, Boston, Massachusetts, United States of America
| | - Brian Wipke
- Previously at Biogen Ltd, Cambridge, Massachusetts, United States of America
- Manifold Bio, Boston, Massachusetts, United States of America
| | - Josef Priller
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Clinical Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Department of Psychiatry and Psychotherapy, Klinikum rechts der Isar, School of Medicine, Technical University of Munich, Munich, Germany
- Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité-Universitätsmedizin Berlin and DZNE, Berlin, Germany
| | - Raphael Schneider
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
| | - Craig S. Moore
- Division of BioMedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John’s, Newfoundland, Canada
| | - Veronique E. Miron
- Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, Toronto, Ontario, Canada
- BARLO Multiple Sclerosis Centre, St. Michael’s Hospital, Toronto, Ontario, Canada
- Department of Immunology, University of Toronto, Toronto, Ontario, Canada
- United Kingdom Dementia Research Institute at The University of Edinburgh, Edinburgh, Scotland, United Kingdom
- Centre for Discovery Brain Sciences, Chancellor’s Building, The University of Edinburgh, Edinburgh, Scotland, United Kingdom
| |
Collapse
|
13
|
Zhang X, Li H, Gu Y, Ping A, Chen J, Zhang Q, Xu Z, Wang J, Tang S, Wang R, Lu J, Lu L, Jin C, Jin Z, Zhang J, Shi L. Repair-associated macrophages increase after early-phase microglia attenuation to promote ischemic stroke recovery. Nat Commun 2025; 16:3089. [PMID: 40164598 PMCID: PMC11958652 DOI: 10.1038/s41467-025-58254-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
Ischemic stroke recovery involves dynamic interactions between the central nervous system and infiltrating immune cells. Peripheral immune cells compete with resident microglia for spatial niches in the brain, but how modulating this balance affects recovery remains unclear. Here, we use PLX5622 to create spatial niches for peripheral immune cells, altering the competition between infiltrating immune cells and resident microglia in male mice following transient middle cerebral artery occlusion (tMCAO). We find that early-phase microglia attenuation promotes long-term functional recovery. This intervention amplifies a subset of monocyte-derived macrophages (RAMf) with reparative properties, characterized by high expression of GPNMB and CD63, enhanced lipid metabolism, and pro-angiogenic activity. Transplantation of RAMf into stroke-affected mice improves white matter integrity and vascular repair. We identify Mafb as the transcription factor regulating the reparative phenotype of RAMf. These findings highlight strategies to optimize immune cell dynamics for post-stroke rehabilitation.
Collapse
Affiliation(s)
- Xiaotao Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Huaming Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Yichen Gu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - An Ping
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jiarui Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Qia Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Zhouhan Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Junjie Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Shenjie Tang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Rui Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jianan Lu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Lingxiao Lu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Chenghao Jin
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Ziyang Jin
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
- Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China.
| | - Ligen Shi
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China.
| |
Collapse
|
14
|
Montilla A, Zabala A, Calvo I, Bosch-Juan M, Tomé-Velasco I, Mata P, Koster M, Sierra A, Kooistra SM, Soria FN, Eggen BJL, Fresnedo O, Fernández JA, Tepavcevic V, Matute C, Domercq M. Microglia regulate myelin clearance and cholesterol metabolism after demyelination via interferon regulatory factor 5. Cell Mol Life Sci 2025; 82:131. [PMID: 40137979 PMCID: PMC11947375 DOI: 10.1007/s00018-025-05648-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 12/16/2024] [Accepted: 01/08/2025] [Indexed: 03/29/2025]
Abstract
Interferon regulatory factor 5 (IRF5) is a transcription factor that plays a role in orchestrating innate immune responses, particularly in response to viral infections. Notably, IRF5 has been identified as a microglia risk gene linked to multiple sclerosis (MS), but its specific role in MS pathogenesis remains unclear. Through the use of Irf5-/- mice, our study uncovers a non-canonical function of IRF5 in MS recovery. Irf5-/- mice exhibited increased damage in an experimental autoimmune encephalomyelitis (EAE) model and demonstrated impaired oligodendrocyte recruitment into the lesion core following lysolecithin-induced demyelination. Transcriptomic and lipidomic analyses revealed that IRF5 has a role in microglia-mediated myelin phagocytosis, lipid metabolism, and cholesterol homeostasis. Indeed, Irf5-/- microglia phagocytose myelin, but myelin debris is not adequately degraded, leading to an accumulation of lipid droplets, cholesterol esters, and cholesterol crystals within demyelinating lesions. This abnormal buildup can hinder remyelination processes. Importantly, treatments that promote cholesterol transport were found to reduce lipid droplet accumulation and mitigate the exacerbated damage in Irf5-/- mice with EAE. Altogether, our study identified the antiviral transcription factor IRF5 as a key transcriptional regulator of lipid degradation and cholesterol homeostasis and suggest that loss of IRF5 function leads to pathogenic lipid accumulation in microglia, thereby obstructing remyelination. These data and the fact that Irf5 polymorphisms are significantly associated with MS, highlight IRF5 as a potential therapeutic target to promote regenerative responses.
Collapse
Affiliation(s)
- Alejandro Montilla
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain.
| | - Alazne Zabala
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Ibai Calvo
- Department of Physical Chemistry, Faculty of Sciences, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Marina Bosch-Juan
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Irene Tomé-Velasco
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Paloma Mata
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Mirjam Koster
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Amanda Sierra
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Ikerbasque Foundation, E-48009, Bilbao, Spain
- Department of Biochemistry and Molecular Biology, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Susanne M Kooistra
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Federico N Soria
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Ikerbasque Foundation, E-48009, Bilbao, Spain
| | - Bart J L Eggen
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Olatz Fresnedo
- Lipids & Liver Research Group, Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - José Andrés Fernández
- Department of Physical Chemistry, Faculty of Sciences, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain
| | - Vanja Tepavcevic
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain
| | - Carlos Matute
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Leioa, Spain.
| | - María Domercq
- Achucarro Basque Center for Neuroscience, E-48940, Leioa, Spain.
- Department of Neuroscience, University of the Basque Country UPV/EHU, E-48940, Leioa, Spain.
| |
Collapse
|
15
|
Huang LX, Sun T, Sun J, Wu ZM, Zhao YB, Li MY, Huo QY, Ling C, Zhang BY, Chen C, Wang H. The Role of Endothelial Cell Glycolysis in Schwann Cells and Peripheral Nerve Injury Repair: A Novel and Important Research Area. Neurochem Res 2025; 50:121. [PMID: 40100469 DOI: 10.1007/s11064-025-04374-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/20/2025]
Abstract
Endothelial cell glycolysis plays a novel and significant role in Schwann cells and peripheral nerve injury repair, which represents an emerging and important area of research. Glycolysis in endothelial cells is a conserved and tightly regulated biological process that provides essential energy (ATP) and intermediates by ultimately converting glucose into lactate. This metabolic pathway is crucial for maintaining the normal function of endothelial cells. During peripheral nerve injury repair, endothelial cell glycolysis influences the function of Schwann cells and the efficiency of nerve regeneration. Beyond glycolysis, endothelial cells also secrete various factors, including growth factors and extracellular vesicles, which further modulate Schwann cell activity and contribute to the repair process. This review will summarize the role of endothelial cell glycolysis in Schwann cell function and peripheral nerve injury repair, aiming to provide new insights for the development of novel strategies for peripheral nerve injury treatment.
Collapse
Affiliation(s)
- Li-Xin Huang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Tao Sun
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Jun Sun
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Zhi-Min Wu
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Yi-Bo Zhao
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Ming-Yang Li
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Qing-Yi Huo
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Cong Ling
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Bao-Yu Zhang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China
| | - Chuan Chen
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| | - Hui Wang
- Department of Neurosurgery, The Third Affiliated Hospital, Sun Yat-Sen University, 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| |
Collapse
|
16
|
Bergner CG, van der Meer F, Franz J, Vakrakou A, Würfel T, Nessler S, Schäfer L, Nau-Gietz C, Winkler A, Lagumersindez-Denis N, Wrzos C, Damkou IA, Sergiou C, Schultz V, Knauer C, Metz I, Bahn E, Garea Rodriguez E, Merkler D, Simons M, Stadelmann C. BCAS1-positive oligodendrocytes enable efficient cortical remyelination in multiple sclerosis. Brain 2025; 148:908-920. [PMID: 39319704 PMCID: PMC11884765 DOI: 10.1093/brain/awae293] [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: 11/04/2023] [Revised: 07/15/2024] [Accepted: 07/27/2024] [Indexed: 09/26/2024] Open
Abstract
Remyelination is a crucial regenerative process in demyelinating diseases, limiting persisting damage to the CNS. It restores saltatory nerve conduction and ensures trophic support of axons. In patients with multiple sclerosis, remyelination has been observed in both white and grey matter and found to be more efficient in the cortex. Brain-enriched myelin-associated protein 1 (BCAS1) identifies oligodendrocyte lineage cells in the stage of active myelin formation in development and regeneration. Other than in the white matter, BCAS1+ oligodendrocytes are maintained at high densities in the cortex throughout life. Here, we investigated cortical lesions in human biopsy and autopsy tissue from patients with multiple sclerosis in direct comparison to demyelinating mouse models and demonstrate that following a demyelinating insult BCAS1+ oligodendrocytes in remyelinating cortical lesions shift from a quiescent to an activated, internode-forming morphology co-expressing myelin-associated glycoprotein (MAG), necessary for axonal contact formation. Of note, activated BCAS1+ oligodendrocytes are found at early time points of experimental demyelination amidst ongoing inflammation. In human tissue, activated BCAS1+ oligodendrocytes correlate with the density of myeloid cells, further supporting their involvement in an immediate regenerative response. Furthermore, studying the microscopically normal appearing non demyelinated cortex in patients with chronic multiple sclerosis, we find a shift from quiescent BCAS1+ oligodendrocytes to mature, myelin-maintaining oligodendrocytes, suggesting oligodendrocyte differentiation and limited replenishment of BCAS1+ oligodendrocytes in long-standing disease. We also demonstrate that part of perineuronal satellite oligodendrocytes are BCAS1+ and contribute to remyelination in human and experimental cortical demyelination. In summary, our results provide evidence from human tissue and experimental models that BCAS1+ cells in the adult cortex represent a population of pre-differentiated oligodendrocytes that rapidly react after a demyelinating insult thus enabling immediate myelin regeneration. In addition, our data suggest that limited replenishment of BCAS1+ oligodendrocytes may contribute to the remyelination failure observed in the cortex in chronic multiple sclerosis.
Collapse
Affiliation(s)
- Caroline Gertrud Bergner
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Department of Neurology, University Hospital Leipzig, Leipzig 04103, Germany
| | - Franziska van der Meer
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Jonas Franz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Aigli Vakrakou
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens 11528, Greece
| | - Thea Würfel
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Stefan Nessler
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Lisa Schäfer
- Department of Neurology, University Hospital Leipzig, Leipzig 04103, Germany
| | - Cora Nau-Gietz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Anne Winkler
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | | | - Claudia Wrzos
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Ioanna Alkmini Damkou
- Institute of Neuronal Cell Biology, Technical University Munich, Munich 81377, Germany
| | - Christina Sergiou
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Verena Schultz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Institute of Infection, Immunity and Inflammation, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, UK
| | - Carolin Knauer
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Imke Metz
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | - Erik Bahn
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
| | | | - Doron Merkler
- Department of Pathology and Immunology, University of Geneva, Geneva 4 1211, Switzerland
- Division of Clinical Pathology, Geneva University Hospital, Geneva 1205, Switzerland
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich 81377, Germany
- German Center for Neurodegenerative Diseases (DZNE), Munich 81377, Germany
- Munich Cluster of Systems Neurology (SyNergy), Munich 81377, Germany
| | - Christine Stadelmann
- Department of Neuropathology, University Medical Center Göttingen, Göttingen 37075, Germany
- Cluster of Excellence “Multiscale Bioimaging: from Molecular Machines to Network of Excitable Cells”(MBExC), University of Göttingen, Göttingen 37073, Germany
| |
Collapse
|
17
|
Cui QL, Mohammadnia A, Yaqubi M, Weng C, Dorion MF, Pernin F, Hall JA, Dudley R, Stratton JA, Kennedy TE, Srour M, Antel JP. Myelination potential and injury susceptibility of grey versus white matter human oligodendrocytes. Brain 2025; 148:921-932. [PMID: 39378316 DOI: 10.1093/brain/awae311] [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: 03/08/2024] [Revised: 08/01/2024] [Accepted: 09/09/2024] [Indexed: 10/10/2024] Open
Abstract
Increasing evidence indicates heterogeneity in functional and molecular properties of oligodendrocyte lineage cells both during development and in pathological conditions. In multiple sclerosis, remyelination of grey matter lesions exceeds that in white matter. Here, we used cells derived from grey matter versus white matter regions of surgically resected human brain tissue samples to compare the capacities of human A2B5-positive progenitor cells and mature oligodendrocytes to ensheath synthetic nanofibres, and we related differences to the molecular profiles of these cells. For both cell types, the percentage of ensheathing cells was greater for grey matter versus white matter cells. For both grey matter and white matter samples, the percentage of cells ensheathing nanofibres was greater for A2B5-positive cells versus mature oligodendrocytes. Grey matter A2B5-positive cells were more susceptible than white matter A2B5-positive cells to injury induced by metabolic insults. Bulk RNA sequencing indicated that separation by cell type (A2B5-positive versus mature oligodendrocytes) is more significant than by region, but segregation for each cell type by region is apparent. Molecular features of grey matter- versus white matter-derived A2B5-positive and mature oligodendrocytes were lower expression of mature oligodendrocyte genes and increased expression of early oligodendrocyte lineage genes. Genes and pathways with increased expression in grey matter-derived cells with relevance for myelination included those related to responses to the external environment, cell-cell communication, cell migration and cell adhesion. Immune- and cell death-related genes were upregulated in grey matter-derived cells. We observed a significant number of upregulated genes shared between the stress/injury and myelination processes, providing a basis for these features. In contrast to oligodendrocyte lineage cells, no functional or molecular heterogeneity was detected in microglia maintained in vitro, probably reflecting the plasticity of these cells ex vivo. The combined functional and molecular data indicate that grey matter human oligodendrocytes have increased intrinsic capacity to myelinate but also increased injury susceptibility, in part reflecting their being at a stage earlier in the oligodendrocyte lineage.
Collapse
Affiliation(s)
- Qiao-Ling Cui
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Abdulshakour Mohammadnia
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Moein Yaqubi
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Chao Weng
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Marie-France Dorion
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Florian Pernin
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Jeffery A Hall
- Department of Neurosurgery, McGill University Health Centre, Montreal, QC H3A 2B4, Canada
- Department of Neurology and Neurosurgery, McGill University Health Centre, Montreal, QC H3A 2B4, Canada
| | - Roy Dudley
- Department of Pediatric Neurosurgery, Montreal Children's Hospital, Montreal, QC H4A 3J1, Canada
| | - Jo Anne Stratton
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Timothy E Kennedy
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Myriam Srour
- Division of Pediatric Neurology, Montreal Children's Hospital, Montreal, QC H4A 3J1, Canada
| | - Jack P Antel
- Neuroimmunology Unit, Montreal Neurological Institute and Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| |
Collapse
|
18
|
Kornberg MD, Calabresi PA. Multiple Sclerosis and Other Acquired Demyelinating Diseases of the Central Nervous System. Cold Spring Harb Perspect Biol 2025; 17:a041374. [PMID: 38806240 PMCID: PMC11875095 DOI: 10.1101/cshperspect.a041374] [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] [Indexed: 05/30/2024]
Abstract
Acquired demyelinating diseases of the central nervous system (CNS) comprise inflammatory conditions, including multiple sclerosis (MS) and related diseases, as well as noninflammatory conditions caused by toxic, metabolic, infectious, traumatic, and neurodegenerative insults. Here, we review the spectrum of diseases producing acquired CNS demyelination before focusing on the prototypical example of MS, exploring the pathologic mechanisms leading to myelin injury in relapsing and progressive MS and summarizing the mechanisms and modulators of remyelination. We highlight the complex interplay between the immune system, oligodendrocytes and oligodendrocyte progenitor cells (OPCs), and other CNS glia cells such as microglia and astrocytes in the pathogenesis and clinical course of MS. Finally, we review emerging therapeutic strategies that exploit our growing understanding of disease mechanisms to limit progression and promote remyelination.
Collapse
Affiliation(s)
- Michael D Kornberg
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University, Baltimore, Maryland 21287, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, Maryland 21205, USA
| |
Collapse
|
19
|
Ahmed MC, Kakunuri T, Peris L, Meffre D, Yilmaz EN, Grewing L, Guerrero González R, Manfroi B, Gout E, Vivès RR, Fitzgerald U, Schneider P, Jafarian-Tehrani M, Kuhlmann T, Huard B. The inflammatory APRIL (a proliferation-inducing ligand) antagonizes chondroitin sulphate proteoglycans to promote axonal growth and myelination. Brain Commun 2025; 7:fcae473. [PMID: 39926615 PMCID: PMC11803424 DOI: 10.1093/braincomms/fcae473] [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: 05/13/2024] [Revised: 11/19/2024] [Accepted: 02/03/2025] [Indexed: 02/11/2025] Open
Abstract
Lesions in the CNS are frequently associated to a detrimental inflammatory reaction. In autoimmune neurodegenerative diseases, a proliferation-inducing ligand (APRIL) produced by CNS-infiltrating inflammatory cells binds to chondroitin sulphate proteoglycans (CSPGs). The latter are well-established obstacles to neural regeneration and remyelination in the CNS by interacting with receptor protein tyrosine phosphatase (RPTP) and Nogo receptor (NgR) families. Here, we are showing that APRIL blocks the interactions of RPTP and NgR with all types of chondroitin sulphate (CS). Functionally, APRIL neutralized the inhibitory effects of CS on mouse and human neuronal process growth. APRIL also blocked the inhibition of CS on mouse and human oligodendrocyte differentiation. Finally, APRIL increased myelination in an ex vivo organotypic model of demyelination in the presence of endogenous CSPG upregulation. Our data demonstrate the potential value for a recombinant form of soluble APRIL to achieve repair in the CNS.
Collapse
Affiliation(s)
- Mashal Claude Ahmed
- Institute for Advanced Biosciences, University Grenoble-Alpes/INSERM U1209/CNRS UMR5209, La Tronche 38700, France
| | - Tejaswini Kakunuri
- T-RAIG, TIMC, University Grenoble-Alpes/CNRS UMR5525, La Tronche 38700, France
| | - Leticia Peris
- Institut des Neurosciences, Université Grenoble Alpes, La Tronche 38700, France
| | - Delphine Meffre
- UMR-S 1124, University Paris-Cité and INSERM, Paris 75006, France
| | - Elif Nur Yilmaz
- Institute of Neuropathology, University Hospital Muenster, Muenster 8149, Germany
| | - Laureen Grewing
- Institute of Neuropathology, University Hospital Muenster, Muenster 8149, Germany
| | | | - Benoit Manfroi
- Institute for Advanced Biosciences, University Grenoble-Alpes/INSERM U1209/CNRS UMR5209, La Tronche 38700, France
| | - Evelyne Gout
- CNRS, CEA, IBS, University of Grenoble Alpes, Grenoble 38000, France
| | - Romain R Vivès
- CNRS, CEA, IBS, University of Grenoble Alpes, Grenoble 38000, France
| | - Una Fitzgerald
- Galway Neuroscience Centre, School of Natural Sciences, National University of Ireland, Galway H91 W2TY, Ireland
| | - Pascal Schneider
- Department of Immunobiology, University of Lausanne, Epalinges 1066, Switzerland
| | | | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Muenster, Muenster 8149, Germany
| | - Bertrand Huard
- T-RAIG, TIMC, University Grenoble-Alpes/CNRS UMR5525, La Tronche 38700, France
| |
Collapse
|
20
|
Liu Z, Wei J, Su H, She Y, Wang J, Liu Y, Mo L, Tan C, Liu X, Chen L. Diabetes may contribute to cognitive impairment in Parkinson's disease via damaging white matter tracts. Neuroreport 2025; 36:71-80. [PMID: 39661531 DOI: 10.1097/wnr.0000000000002123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Parkinson's disease with dementia (PDD) severely affects the quality of life of patients with Parkinson's disease (PD) in the later stages. Recently, PD patients with diabetes were found to have a higher risk of cognitive decline and developing dementia with a faster progression, but the underlying mechanism remains unclear. Diabetes-related white matter damage may partially explain the mechanism by which diabetes participates in PDD. Seventy PD patients were included. PD patients underwent diffusion tensor imaging from The Second Affiliated Hospital of Chongqing Medical University were collected and were divided into four groups: PD with diabetes without dementia, PD with dementia without diabetes, PD without dementia and diabetes. Tract-based spatial statistics analysis and region-of-interest-based analysis were performed. Factorial analysis with diabetes and dementia taken as the main effects was performed, and the differences between the white matter fibers of PD patients from the four groups were also analyzed. The interaction between diabetes and dementia in the damage of white matter in PD patients was also analyzed. We found that both diabetes and dementia were found to be related to the damage in internal capsule, corona radiata, and thalamic radiation of the PD patients. There is an interaction between diabetes and dementia in the white matter damage of PD patients. Both diabetes and dementia were found to be related to the damage in internal capsule, corona radiata, and thalamic radiation of the PD patients. Diabetes may participate in cognitive decline in PD patients via damaging cognition-related white matter tracts.
Collapse
Affiliation(s)
- Zhihui Liu
- Department of Neurology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Lee CJ, Jang SH, Lim J, Park H, Ahn SH, Park SY, Seo H, Song SJ, Shin JA, Choi C, Gee HY, Choi YH. Exosome-based targeted delivery of NF-κB ameliorates age-related neuroinflammation in the aged mouse brain. Exp Mol Med 2025; 57:235-248. [PMID: 39833561 PMCID: PMC11799301 DOI: 10.1038/s12276-024-01388-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/25/2024] [Accepted: 10/29/2024] [Indexed: 01/22/2025] Open
Abstract
Neuroinflammation, a significant contributor to various neurodegenerative diseases, is strongly associated with the aging process; however, to date, no efficacious treatments for neuroinflammation have been developed. In aged mouse brains, the number of infiltrating immune cells increases, and the key transcription factor associated with increased chemokine levels is nuclear factor kappa B (NF-κB). Exosomes are potent therapeutics or drug delivery vehicles for various materials, including proteins and regulatory genes, to target cells. In the present study, we evaluated the therapeutic efficacy of exosomes loaded with a nondegradable form of IκB (Exo-srIκB), which inhibits the nuclear translocation of NF-κB to suppress age-related neuroinflammation. Single-cell RNA sequencing revealed that these anti-inflammatory exosomes targeted macrophages and microglia, reducing the expression of inflammation-related genes. Treatment with Exo-srIκB also suppressed the interactions between macrophages/microglia and T and B cells in the aged brain. We demonstrated that Exo-srIκB successfully alleviates neuroinflammation by primarily targeting activated macrophages and partially modulating the functions of age-related interferon-responsive microglia in the brain. Thus, our findings highlight Exo-srIκB as a potential therapeutic agent for treating age-related neuroinflammation.
Collapse
Affiliation(s)
- Chae-Jeong Lee
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea
| | - Seung Hyun Jang
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Jiwoo Lim
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea
| | - Hyunju Park
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea
| | - So-Hee Ahn
- ILIAS Biologics Inc., Daejeon, 34014, Republic of Korea
| | | | - Hyangmi Seo
- ILIAS Biologics Inc., Daejeon, 34014, Republic of Korea
| | - Soo-Jin Song
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea
| | - Jung-A Shin
- Department of Anatomy, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea
| | - Chulhee Choi
- ILIAS Biologics Inc., Daejeon, 34014, Republic of Korea.
| | - Heon Yung Gee
- Department of Pharmacology, Brain Korea 21 PLUS Project for Medical Sciences, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| | - Youn-Hee Choi
- Department of Physiology, Inflammation-Cancer Microenvironment Research Center, Ewha Womans University College of Medicine, Seoul, 07804, Republic of Korea.
| |
Collapse
|
22
|
McCaffrey D, Weickert CS, Walker AK. Blood IL-1α and IL-6 predict specific breast cancer-induced increases in hippocampal pro-inflammatory cytokines in mice. Cytokine 2025; 186:156826. [PMID: 39667084 DOI: 10.1016/j.cyto.2024.156826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 11/22/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024]
Abstract
Neuroinflammation is a key factor in cognitive and behavioral changes seen in patients with non-CNS cancers, and cytokine levels in the blood are often used as a proxy for brain inflammation. However, this approach has yielded inconsistent results, and a common inflammatory signature remains elusive. To explore whether a blood-to-brain inflammatory signature exists across breast cancer types, we assessed cytokine and glial protein responses in the hippocampus, prefrontal cortex (PFC), and their relationship to serum cytokines in mice bearing three different mammary cancers (n = 40). While cytokine profiles in both serum and brain varied by cancer type, IL-1β and IL-4 were consistently altered across brain regions. In some cases, elevated serum IL-1α and IL-6 correlated with increased hippocampal IL-6. These findings support the use of blood cytokines to identify cancer patients at risk for cognitive and psychiatric comorbidities. However, our data also suggest that relying solely on serum cytokines may lead to under-diagnosis, as some mice exhibited brain cytokine elevations without changes in serum levels. This underscores the need for a broader range of inflammatory markers in blood to better identify at-risk patients. Brain region-specific differences in the cytokine response to mammary cancer highlighted the hippocampus as more vulnerable to cancer-induced inflammation than the PFC. We observed region-specific glial cell reactivity, however, only astrocyte and oligodendrocyte markers were correlated with cytokine changes within the hippocampus. Elevated serum IL-1α and IL-6 were correlated with reduced cortical astrocyte reactivity, suggesting that these cytokines can inform glial cell-specific changes in this region.
Collapse
Affiliation(s)
- Delyse McCaffrey
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, New South Wales, Australia; Discipline of Psychiatry and Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia
| | - Cynthia Shannon Weickert
- Discipline of Psychiatry and Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia; Schizophrenia Research Laboratory, Neuroscience Research Australia, Randwick, New South Wales, Australia; Department of Neuroscience & Physiology, Upstate Medical University, Syracuse, NY, USA
| | - Adam K Walker
- Laboratory of ImmunoPsychiatry, Neuroscience Research Australia, Randwick, New South Wales, Australia; Discipline of Psychiatry and Mental Health, Faculty of Medicine, University of New South Wales, Sydney, Australia; Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Australia.
| |
Collapse
|
23
|
Tang Y, Zhang L, Huang P, She Z, Luo S, Peng H, Chen Y, Luo J, Duan W, Xiao Y, Liu L, Liu L. Understanding the intricacies of cellular mechanisms in remyelination: The role of circadian rhythm. Neurochem Int 2025; 183:105929. [PMID: 39756585 DOI: 10.1016/j.neuint.2025.105929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
The term "circadian rhythm" refers to the 24-h oscillations found in various physiological processes in organisms, responsible for maintaining bodily homeostasis. Many neurological diseases mainly involve the process of demyelination, and remyelination is crucial for the treatment of neurological diseases. Current research mainly focuses on the key role of circadian clocks in the pathophysiological mechanisms of multiple sclerosis. Various studies have shown that the circadian rhythm regulates various cellular molecular mechanisms and signaling pathways involved in remyelination. The process of remyelination is primarily mediated by oligodendrocyte precursor cells (OPCs), oligodendrocytes, microglia, and astrocytes. OPCs are activated, proliferate, migrate, and ultimately differentiate into oligodendrocytes after demyelination, involving many key signaling pathway and regulatory factors. Activated microglia secretes important cytokines and chemokines, promoting OPC proliferation and differentiation, and phagocytoses myelin debris that inhibits remyelination. Astrocytes play a crucial role in supporting remyelination by secreting signals that promote remyelination or facilitate the phagocytosis of myelin debris by microglia. Additionally, cell-to-cell communication via gap junctions allows for intimate contact between astrocytes and oligodendrocytes, providing metabolic support for oligodendrocytes. Therefore, gaining a deeper understanding of the mechanisms and molecular pathways of the circadian rhythm at various stages of remyelination can help elucidate the fundamental characteristics of remyelination and provide insights into treating demyelinating disorders.
Collapse
Affiliation(s)
- Yufen Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Zhou She
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Senlin Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Hong Peng
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Yuqiong Chen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Jinwen Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Wangxin Duan
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Yangyang Xiao
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China
| | - Lingjuan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China.
| | - Liqun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Department of Pediatric Neurology, Children's Medical Center, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China; Clinical Medical Research Center for Child Development and Behavior, Changsha, 410011, Hunan, China.
| |
Collapse
|
24
|
Wang W, Wang Y, Huang X, Wu P, Li L, Zhang Y, Chen Y, Chen Z, Li C, Zhou Y, Zhang J. Pathophysiology-Directed Engineering of a Combination Nanoanalgesic for Neuropathic Pain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405483. [PMID: 39716944 PMCID: PMC11848598 DOI: 10.1002/advs.202405483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 12/08/2024] [Indexed: 12/25/2024]
Abstract
Neuropathic pain, one of the most refractory pain diseases, remains a formidable medical challenge. There is still an unmet demand for effective and safe therapies to address this condition. Herein, a rat model of nerve injury-induced neuropathic pain is first established to explore its pathophysiological characteristics. Recognizing the role of neuroinflammation, an inflammation-resolving amphiphilic conjugate PPT is designed and synthesized by simultaneously conjugating polyethylene glycol, phenylboronic acid pinacol ester, and Tempol onto a cyclic scaffold. PPT can self-assemble into nanomicelles (termed PPTN). Following intravenous injection, PPTN preferentially accumulates in the injured nerve, ameliorates the neuroinflammatory milieu, and promotes nerve regeneration, thereby shortening neuropathic pain duration in rats. Moreover, the Ca2+ channel α2δ1 subunit is identified as a therapeutic target by RNA-sequencing analysis of the injured nerve. Based on this target, a mimicking peptide (AD peptide) is screened as an analgesic. By packaging AD peptide into PPTN, a combination nano-analgesic APTN is developed. Besides potentiated anti-hyperalgesic effects due to site-specific delivery and on-demand release of AD peptide at target sites, APTN simultaneously inhibits neuroinflammation and promotes nerve regeneration by reprogramming macrophages via regulating MAPK/NF-kB signaling pathways and NLRP3 inflammasome activation, thus affording synergistic efficacies in treating nerve injury-induced neuropathic pain.
Collapse
Affiliation(s)
- Wenkai Wang
- Department of OrthopedicsXinqiao HospitalThird Military Medical University (Army Medical University)Chongqing400037P. R. China
- Department of OrthopedicsGeneral Hospital of PLA Xizang Military Area CommandLhasa850007P. R. China
| | - Yan Wang
- Department of PharmaceuticsCollege of PharmacyThird Military Medical University (Army Medical University)Chongqing400038P. R. China
- War Trauma Medical CenterState key Laboratory of TraumaBurns and Combined injuryArmy Medical CenterDaping HospitalThird Military Medical University (Army Medical University)Chongqing400038P. R. China
| | - Xinle Huang
- Department of OrthopedicsXinqiao HospitalThird Military Medical University (Army Medical University)Chongqing400037P. R. China
- Department of OrthopedicsThe Second Naval Hospital of Southern Theater CommandSanya572000P. R. China
| | - Peng Wu
- Department of PharmaceuticsCollege of PharmacyThird Military Medical University (Army Medical University)Chongqing400038P. R. China
- School of PharmacyHanzhong Vocational and Technical CollegeHanzhong723002P. R. China
| | - Lanlan Li
- Department of PharmaceuticsCollege of PharmacyThird Military Medical University (Army Medical University)Chongqing400038P. R. China
| | - Yang Zhang
- Department of OrthopedicsXinqiao HospitalThird Military Medical University (Army Medical University)Chongqing400037P. R. China
| | - Yihui Chen
- Department of General SurgeryXinqiao HospitalThird Military Medical University (Army Medical University)Chongqing400037P. R. China
| | - Zhiyu Chen
- Department of OrthopedicsThe First Affiliated HospitalChongqing Medical UniversityChongqing400016P. R. China
| | - Changqing Li
- Department of OrthopedicsXinqiao HospitalThird Military Medical University (Army Medical University)Chongqing400037P. R. China
| | - Yue Zhou
- Department of OrthopedicsXinqiao HospitalThird Military Medical University (Army Medical University)Chongqing400037P. R. China
| | - Jianxiang Zhang
- Department of PharmaceuticsCollege of PharmacyThird Military Medical University (Army Medical University)Chongqing400038P. R. China
- State Key Laboratory of Trauma and Chemical PoisoningThird Military Medical University (Army Medical University)Chongqing400038P. R. China
- Yu‐Yue Pathology Scientific Research Center313 Gaoteng Avenue, Jiulongpo DistrictChongqing400039P. R. China
| |
Collapse
|
25
|
Shin HG, Kim W, Lee JH, Lee HS, Nam Y, Kim J, Li X, van Zijl PCM, Calabresi PA, Lee J, Jang J. Association of iron deposition in MS lesion with remyelination capacity using susceptibility source separation MRI. Neuroimage Clin 2025; 45:103748. [PMID: 39904206 PMCID: PMC11847087 DOI: 10.1016/j.nicl.2025.103748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/21/2025] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
OBJECTIVES Susceptibility source-separation (χ-separation) MRI provides in-vivo proxy of myelin (diamagnetic susceptibility, χdia) and iron concentrations (paramagnetic susceptibility, χpara) in the central nervous system, potentially uncovering myelin- and iron-related pathology in multiple sclerosis (MS) lesions (e.g., demyelination, remyelination, and iron-laden microglia/macrophages formation). This study aims to monitor longitudinal changes in χpara and χdia signals within MS lesions using χ-separation and evaluate the association between lesional iron and remyelination capability. METHODS Fifty participants with MS (pwMS) were followed annually over a mean period of 3.3 years (SD = 1.8 years) with MRI, including χ-separation, and clinical assessments. To monitor lesions from their early stage (lesion age < 1 year), we identified newly-noted lesions (NNLs) and contrast-enhancing lesions (CELs), and tracked their longitudinal changes in χpara and χdia signals. RESULTS Twenty-three pwMS were detected with NNLs and/or CELs (38 NNLs, 31 CELs;7 overlapped). Among these lesions (62 lesions in total), 27 exhibited χpara hyperintensity, termed hyper-paramagnetic sign (HPS), indicating iron deposition "throughout" the lesion (not confined to rim sign). Early-stage HPS correlated with future remyelination failure detected by χdia myelin signals (P < 0.001). After adjustment, lesions with early HPS demonstrated an annual loss in myelin signal (-1.94 ppb/year), whereas those without early HPS exhibited annual recovery (+0.66 ppb/year). Participants with confirmed disability improvement (CDI) had fewer HPS-positive lesions at baseline than those without CDI (P < 0.001). CONCLUSION The presence of HPS is associated with impaired remyelination capacity and a lack of disease improvement in pwMS. Identifying HPS may help demarcate lesions more amenable to myelin repair therapies.
Collapse
Affiliation(s)
- Hyeong-Geol Shin
- Department of Radiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21218, United States; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, United States
| | - Woojun Kim
- Department of Neurology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jung Hwan Lee
- Department of Neurology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Hyun-Soo Lee
- MR Research Collaboration, Siemens Healthineers, Seoul 06620, Republic of Korea
| | - Yoonho Nam
- Division of Biomedical Engineering, Hankuk University of Foreign Studies, Yongin 17035, South Korea
| | - Jiwoong Kim
- Department of Mathematics and Statistics, University of South Florida, Tampa, FL 33620, United States
| | - Xu Li
- Department of Radiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21218, United States; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, United States
| | - Peter C M van Zijl
- Department of Radiology, School of Medicine, Johns Hopkins University, Baltimore, MD 21218, United States; F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, United States
| | - Peter A Calabresi
- Department of Neurology and Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21218, United States
| | - Jongho Lee
- Department of Electrical and Computer Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jinhee Jang
- Department of Radiology, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea; Institute for Precision Health, University of California, Irvine, Irvine, CA 92697, United States.
| |
Collapse
|
26
|
Zhu Z, Meng M, Mo S, Wang X, Qiao L. M2 Microglia-Derived Exosomal miR-144-5p Attenuates White Matter Injury in Preterm Infants by Regulating the PTEN/AKT Pathway Through KLF12. Mol Biotechnol 2025:10.1007/s12033-025-01364-1. [PMID: 39841378 DOI: 10.1007/s12033-025-01364-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 12/27/2024] [Indexed: 01/23/2025]
Abstract
Perinatal white matter injury (WMI), which is prevalent in premature infants, involves M2 microglia affecting oligodendrocyte precursor cells (OPCs) through exosomes, promoting OPC growth and reducing WMI. The molecular mechanism of WMI remains unclear, and this study explored the role of M2 microglia-derived exosomes in WMI. A tMCAO rat model was constructed to simulate WMI characteristics in vivo. Cresyl violet staining, neurobehavioral tests, rotarod tests, immunofluorescence and immunochemistry were used to assess the role of exos-derived miR-144-5p in pathological and neurological changes in rats. OGD/R cellular models were constructed to mimic WMI characteristics in vitro. CCK-8, TUNEL, Western blotting and immunofluorescence were used to assess the role of exos-derived miR-144-5p in OPC phenotypes. Rescue assays were used to assess the role of the PTEN/AKT pathway in miR-144-5p-mediated OPC phenotypes. Bioinformatics and mechanistic experiments were used to assess the association of PTEN or KLF12 with miR-144-5p in OPCs. M2-Exos suppressed cerebral injury and facilitated demyelination repair in rats post WMI. M2-Exos suppressed OGD/R-stimulated OPC apoptosis and facilitated OGD/R-stimulated OPC differentiation. M2-Exo-derived miR-144-5p suppressed OGD/R-stimulated OPC apoptosis and facilitated OGD/R-stimulated OPC differentiation. M2-Exo-derived miR-144-5p suppressed cerebral injury and facilitated demyelination repair in rats post WMI. MiR-144-5p suppressed OGD/R-stimulated OPC apoptosis and facilitated OGD/R-stimulated OPC differentiation through PTEN downregulation. MiR-144-5p targeted the KLF12 3'UTR to repress PTEN transcription in OPCs. M2 microglia secrete miR-144-5p to reduce WMI by targeting KLF12 in OPCs, inhibiting PTEN/AKT pathway activity, and offering potential targeted therapeutic insights for WMI.
Collapse
Affiliation(s)
- Zhaokui Zhu
- Department of Pediatrics, Zhongda Hospital, The School of Medicine, Southeast University, No. 87 Dingjiaqiao, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Meng Meng
- The School of Medicine, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Sisi Mo
- Department of Pediatrics, Zhongda Hospital, The School of Medicine, Southeast University, No. 87 Dingjiaqiao, Hunan Road, Nanjing, 210009, Jiangsu, China
| | - Xinyu Wang
- The Hospital of Yangzhou University, Yangzhou, 210033, Jiangsu, China
| | - Lixing Qiao
- Department of Pediatrics, Zhongda Hospital, The School of Medicine, Southeast University, No. 87 Dingjiaqiao, Hunan Road, Nanjing, 210009, Jiangsu, China.
| |
Collapse
|
27
|
Gaitsch H, Assinck P, Dimas P, Zhao C, Morcom L, Rowitch DH, Reich DS, Franklin RJM. Significant oligodendrocyte progenitor and microglial cell death is a feature of remyelination following toxin-induced experimental demyelination. Brain Commun 2025; 7:fcae386. [PMID: 39830424 PMCID: PMC11739797 DOI: 10.1093/braincomms/fcae386] [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: 05/03/2024] [Revised: 10/01/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025] Open
Abstract
The extent to which glial cell turnover features in successful remyelination is unclear. In this study, the rat caudal cerebellar peduncle-ethidium bromide lesion model was used to profile oligodendroglial and microglial/macrophage cell death and proliferation dynamics over the course of repair. Lesioned and control tissue was co-labelled with antibody markers for cell identity, proliferation, and apoptosis (TUNEL assay), then imaged at full thickness using confocal microscopy and quantified using custom CellProfiler pipelines. Early remyelination time points were marked by an increased density of total proliferating cells, including oligodendrocyte progenitor cells. Late remyelination time points featured increased TUNEL+ oligodendrocyte progenitor cells: however, most TUNEL+ cells within remyelinating lesions were Iba1+ microglia/macrophages. These results indicate that repairing lesions are characterized by a high degree of glial cell death and suggest that monitoring cell death-related by-products might have clinical value in the setting of remyelination.
Collapse
Affiliation(s)
- Hallie Gaitsch
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Peggy Assinck
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
- Altos Labs, Cambridge Institute of Science, Cambridge CB21 6GQ, UK
| | - Penelope Dimas
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
- Altos Labs, Cambridge Institute of Science, Cambridge CB21 6GQ, UK
| | - Chao Zhao
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
- Altos Labs, Cambridge Institute of Science, Cambridge CB21 6GQ, UK
| | - Laura Morcom
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
| | - David H Rowitch
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Robin J M Franklin
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0AW, UK
- Altos Labs, Cambridge Institute of Science, Cambridge CB21 6GQ, UK
| |
Collapse
|
28
|
Chen JQA, Wever DD, McNamara NB, Bourik M, Smolders J, Hamann J, Huitinga I. Inflammatory microglia correlate with impaired oligodendrocyte maturation in multiple sclerosis. Front Immunol 2025; 15:1522381. [PMID: 39877374 PMCID: PMC11772157 DOI: 10.3389/fimmu.2024.1522381] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/19/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction Remyelination of demyelinated axons can occur as an endogenous repair mechanism in multiple sclerosis (MS), but its efficacy varies between both MS individuals and lesions. The molecular and cellular mechanisms that drive remyelination remain poorly understood. Here, we studied the relation between microglia activation and remyelination activity in MS. Methods We correlated regenerative (CD163+) and inflammatory (iNOS+) microglia with BCAS1+ oligodendrocytes, subdivided into early-stage (<3 processes) and late-stage (≥3 processes) cells in brain donors with high or low remyelinating potential in remyelinated lesions and active lesions with ramified/amoeboid (non-foamy) or foamy microglia. A cohort of MS donors categorized as efficiently remyelinating donors (ERDs; n=25) or poorly remyelinating donors (PRDs; n=17) was included, based on their proportion of remyelinated lesions at autopsy. Results and discussion We hypothesized more CD163+ microglia and BCAS1+ oligodendrocytes in remyelinated and active non-foamy lesions from ERDs and more iNOS+ microglia with fewer BCAS1+ oligodendrocytes in active foamy lesions from PRDs. For CD163+ microglia, however, no differences were observed between MS lesions and MS donor groups. In line with our hypothesis, we found that INOS+ microglia were significantly increased in PRDs compared to ERDs within remyelinated lesions. MS lesions, more late-stage BCAS1+ oligodendrocytes were detected in active lesions with non-foamy or foamy microglia in comparison with remyelinated lesions. Although no differences were found for early-stage BCAS1+ oligodendrocytes between MS lesions, we did find significantly more early-stage BCAS1+ oligodendrocytes in PRDs vs ERDs in remyelinated lesions. Interestingly, a positive correlation was identified between iNOS+ microglia and the presence of early-stage BCAS1+ oligodendrocytes. These findings suggest that impaired maturation of early-stage BCAS1+ oligodendrocytes, encountering inflammatory microglia, may underlie remyelination deficits and unsuccessful lesion repair in MS.
Collapse
Affiliation(s)
- J. Q. Alida Chen
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Dennis D. Wever
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Niamh B. McNamara
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Morjana Bourik
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
| | - Joost Smolders
- Departments of Neurology and Immunology, MS Center ErasMS, Erasmus MC, University Medical Center, Rotterdam, Netherlands
| | - Jörg Hamann
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
- Department of Experimental Immunology, Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Inge Huitinga
- Neuroimmunology Research Group, Netherlands Institute for Neuroscience, Amsterdam, Netherlands
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
29
|
Groh J, Simons M. White matter aging and its impact on brain function. Neuron 2025; 113:127-139. [PMID: 39541972 DOI: 10.1016/j.neuron.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/16/2024] [Accepted: 10/18/2024] [Indexed: 11/17/2024]
Abstract
Aging has a detrimental impact on white matter, resulting in reduced volume, compromised structural integrity of myelinated axons, and an increase in white matter hyperintensities. These changes are closely linked to cognitive decline and neurological disabilities. The deterioration of myelin and its diminished ability to regenerate as we age further contribute to the progression of neurodegenerative disorders. Understanding these changes is crucial for devising effective disease prevention strategies. Here, we will discuss the structural alterations in white matter that occur with aging and examine the cellular and molecular mechanisms driving these aging-related transformations. We highlight how the progressive disruption of white matter may initiate a self-perpetuating cycle of inflammation and neural damage.
Collapse
Affiliation(s)
- Janos Groh
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany.
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, Munich, Germany.
| |
Collapse
|
30
|
Gharibani P, Abramson E, Shanmukha S, Smith MD, Godfrey WH, Lee JJ, Hu J, Baydyuk M, Dorion MF, Deng X, Guo Y, Calle AJ, A Hwang S, Huang JK, Calabresi PA, Kornberg MD, Kim PM. The protein kinase C modulator bryostatin-1 therapeutically targets microglia to attenuate neuroinflammation and promote remyelination. Sci Transl Med 2025; 17:eadk3434. [PMID: 39772770 DOI: 10.1126/scitranslmed.adk3434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 09/19/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025]
Abstract
In multiple sclerosis (MS), microglia and macrophages within the central nervous system (CNS) play an important role in determining the balance among demyelination, neurodegeneration, and myelin repair. Phagocytic and regenerative functions of these CNS innate immune cells support remyelination, whereas chronic and maladaptive inflammatory activation promotes lesion expansion and disability, particularly in the progressive forms of MS. No currently approved drugs convincingly target microglia and macrophages within the CNS, contributing to the lack of therapies aimed at promoting remyelination and slowing disease progression for individuals with MS. Here, we found that the protein kinase C (PKC)-modulating drug bryostatin-1 (bryo-1), a CNS-penetrant compound with an established human safety profile, shifts the transcriptional programs of microglia and CNS-associated macrophages from a proinflammatory phenotype to a regenerative phenotype in vitro and in vivo. Treatment of microglia with bryo-1 stimulated scavenger pathways, phagocytosis, and secretion of factors that prevented the activation of neuroinflammatory reactive astrocytes while also promoting neuroaxonal health and oligodendrocyte differentiation. In line with these findings, systemic treatment of mice with bryo-1 augmented remyelination after a focal demyelinating injury. Our results demonstrate the potential of bryo-1 and possibly a wider class of PKC modulators as myelin-regenerative and supportive agents in MS and other neurologic diseases.
Collapse
Affiliation(s)
- Payam Gharibani
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Efrat Abramson
- Interdepartmental Neuroscience Program, Yale University School of Medicine, Yale University, New Haven, CT 06510, USA
| | - Shruthi Shanmukha
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Matthew D Smith
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Wesley H Godfrey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Judy J Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jingwen Hu
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Maryna Baydyuk
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Marie-France Dorion
- Neuroimmunology Unit, Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Montreal, QC H3A 2B4, Canada
| | - Xiaojing Deng
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Yu Guo
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Andrew J Calle
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Soonmyung A Hwang
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Jeffrey K Huang
- Department of Biology, Georgetown University, Washington, DC 20057, USA
| | - Peter A Calabresi
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Michael D Kornberg
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Paul M Kim
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
31
|
Wang W, Guo M, Tu X, Jiang M, Zhang CJ. Neuronal STING activation mediates inflammation-induced neurodegeneration via ferroptosis pathways in multiple sclerosis. BIOPHYSICS REPORTS 2024; 10:416-417. [PMID: 39758424 PMCID: PMC11693497 DOI: 10.52601/bpr.2024.240908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 07/15/2024] [Indexed: 01/07/2025] Open
Affiliation(s)
- Weiyan Wang
- Department of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Mengdi Guo
- Department of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Xiao Tu
- Department of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Meiling Jiang
- Department of Science and Technology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Cun-Jin Zhang
- Department of Neurology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu 611731, China
| |
Collapse
|
32
|
Cruz-Chamorro I, Álvarez-López AI, Santos-Sánchez G, Álvarez-Sánchez N, Pedroche J, Millán-Linares MDC, Lardone PJ, Carrillo-Vico A. A Lupin ( Lupinus angustifolius) Protein Hydrolysate Decreases the Severity of Experimental Autoimmune Encephalomyelitis: A Preliminary Study. Int J Mol Sci 2024; 26:32. [PMID: 39795896 PMCID: PMC11720533 DOI: 10.3390/ijms26010032] [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: 11/25/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Multiple sclerosis (MS) is a neurodegenerative disease, with inflammation and oxidative stress in the central nervous system being the main triggers. There are many drugs that reduce the clinical signs of MS, but none of them cure the disease. Food proteins have been shown to contain encrypted peptides that can be released after hydrolysis and exert numerous biological activities. Recently, we have demonstrated the anti-inflammatory and antioxidant activities of a lupin protein hydrolysate (LPH) both in vitro and in vivo. Therefore, the aim of this study was to evaluate whether LPH is capable of reducing the clinical signs of experimental autoimmune encephalomyelitis (EAE), a mouse model of MS. EAE was induced in female C57BL/6N mice and they were treated intragastrically with LPH (100 mg/kg) or vehicle (control group) from day 0 (prophylactic approach) or from the onset of the disease (day 12 post-induction; therapeutic approach) and the clinical score of each mouse was recorded daily. Prophylactic treatment with LPH reduced the clinical score of the mice compared to the control group, as well as the maximum and cumulative scores, without changing the day of onset of the symptoms while the therapeutic intervention did not significantly improve the severity of the disease. For the first time, we demonstrated that prophylactic administration of LPH reduces the severity of MS, suggesting a potential nutraceutical or new functional foods in neuroinflammation. However, further studies are needed to confirm this nutritional effect in a clinical context.
Collapse
Affiliation(s)
- Ivan Cruz-Chamorro
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain; (A.I.Á.-L.); (G.S.-S.); (P.J.L.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain;
| | - Ana Isabel Álvarez-López
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain; (A.I.Á.-L.); (G.S.-S.); (P.J.L.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain;
| | - Guillermo Santos-Sánchez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain; (A.I.Á.-L.); (G.S.-S.); (P.J.L.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain;
| | - Nuria Álvarez-Sánchez
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain;
| | - Justo Pedroche
- Department of Food & Health, Instituto de la Grasa, CSIC, Ctra, Utrera Km 1, 41013 Seville, Spain; (J.P.); (M.d.C.M.-L.)
| | | | - Patricia Judith Lardone
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain; (A.I.Á.-L.); (G.S.-S.); (P.J.L.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain;
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, 41013 Seville, Spain; (A.I.Á.-L.); (G.S.-S.); (P.J.L.)
- Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, 41009 Seville, Spain;
| |
Collapse
|
33
|
Wang Q, Yang S, Zhang X, Zhang S, Chen L, Wang W, Chen N, Yan J. Inflammasomes in neurodegenerative diseases. Transl Neurodegener 2024; 13:65. [PMID: 39710713 PMCID: PMC11665095 DOI: 10.1186/s40035-024-00459-0] [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: 08/23/2024] [Accepted: 11/27/2024] [Indexed: 12/24/2024] Open
Abstract
Inflammasomes represent a crucial component of the innate immune system, which respond to threats by recognizing different molecules. These are known as pathogen-associated molecular patterns (PAMPs) or host-derived damage-associated molecular patterns (DAMPs). In neurodegenerative diseases and neuroinflammation, the accumulation of misfolded proteins, such as beta-amyloid and alpha-synuclein, can lead to inflammasome activation, resulting in the release of interleukin (IL)-1β and IL-18. This activation also induces pyroptosis, the release of inflammatory mediators, and exacerbates neuroinflammation. Increasing evidence suggests that inflammasomes play a pivotal role in neurodegenerative diseases. Therefore, elucidating and investigating the activation and regulation of inflammasomes in these diseases is of paramount importance. This review is primarily focused on evidence indicating that inflammasomes are activated through the canonical pathway in these diseases. Inflammasomes as potential targets for treating neurodegenerative diseases are also discussed.
Collapse
Affiliation(s)
- Qianchen Wang
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Songwei Yang
- Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, School of Pharmacy, Hunan University of Chinese Medicine, Changsha, 410208, China
| | - Xuan Zhang
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shanshan Zhang
- China Three Gorges University College of Medicine and Health Sciences, Yichang, 443002, China
| | - Liping Chen
- Department of Pharmacy, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Wanxue Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Naihong Chen
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica and Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, China
| | - Jiaqing Yan
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
34
|
Pérez-Osorio IN, Espinosa-Cerón JA, Álvarez-Gutiérrez C, Gonzalez-Flores R, Besedovsky H, Fragoso G, Torres-Ramos MA, Sciutto E. Combined Use of Intranasal Methylprednisolone and Allopregnanolone: Revisiting Anti-inflammatory and Remyelinating Treatment in a Murine Model of Multiple Sclerosis. FRONT BIOSCI-LANDMRK 2024; 29:420. [PMID: 39735995 DOI: 10.31083/j.fbl2912420] [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: 08/20/2024] [Revised: 10/30/2024] [Accepted: 11/06/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND Multiple sclerosis (MS) is a demyelinating, neuroinflammatory, progressive disease that severely affects human health of young adults. Neuroinflammation (NI) and demyelination, as well as their interactions, are key therapeutic targets to halt or slow disease progression. Potent steroidal anti-inflammatory drugs such as methylprednisolone (MP) and remyelinating neurosteroids such as allopregnanolone (ALLO) could be co-administered intranasally to enhance their efficacy by providing direct access to the central nervous system (CNS). METHODS The individual and combined effects of MP and ALLO to control the clinical score of murine experimental autoimmune encephalitis (EAE), to preserve spinal cord tissue integrity, modulate cellular infiltration and gliosis, promote remyelination, and modify the expression of Aryl hydrocarbon receptor (AhR) were evaluated. In silico studies, to deep insight into the mechanisms involved for the treatments, were also conducted. RESULTS MP was the only treatment that significantly reduced the EAE severity, infiltration of inflammatory cells and ionized calcium-binding adapter molecule 1 (Iba-1) expression respect to those EAE non-treated mice but with no-significant differences between the three treatments. MP, ALLO and MP+ALLO significantly reduced tissue damage, AhR expression, and promoted remyelination. Overall, these results suggest that MP, with or without the co-administration with ALLO is an effective and safe strategy to reduce the inflammatory status and the progression of EAE. Despite the expectations of the use of ALLO to reduce the inflammation in EAE, its effect in the dose-scheme used herein is limited only to improve myelination, an effect that supports its usefulness in demyelinating diseases. These results indicate the interest in exploring different doses of ALLO to recommend its use. CONCLUSIONS ALLO treatment mainly maintain the integrity of the spinal cord tissue and the presence of myelin without affecting NI and the clinical outcome. AhR could be involved in the effect observed in both, MP and ALLO treatments. These results will help in the development of a more efficient therapy for MS patients.
Collapse
Affiliation(s)
- Iván Nicolás Pérez-Osorio
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| | - José Alejandro Espinosa-Cerón
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| | - Camila Álvarez-Gutiérrez
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| | - Rodrigo Gonzalez-Flores
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| | - Hugo Besedovsky
- Research Group Immunophysiology, Division of Neurophysiology, Institute of Physiology and Pathophysiology, Philipps Universität, 35037 Marburg, Germany
| | - Gladis Fragoso
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| | - Mónica A Torres-Ramos
- Research Directorate, National Institute of Neurology and Neurosurgery Manuel Velasco Suárez, Tlalpan, 14269 Mexico City, Mexico
- Laboratory 4 Translational Sciences, Center for Research on Aging, CINVESTAV South Headquarters, 14330, Mexico City, Mexico
| | - Edda Sciutto
- Department of Immunology, Institute of Biomedical Research Universidad Nacional Autónoma de México, UNAM, 04510 Mexico City, Mexico
| |
Collapse
|
35
|
Medina R, Derias AM, Lakdawala M, Speakman S, Lucke-Wold B. Overview of emerging therapies for demyelinating diseases. World J Clin Cases 2024; 12:6361-6373. [PMID: 39464332 PMCID: PMC11438674 DOI: 10.12998/wjcc.v12.i30.6361] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/04/2024] Open
Abstract
This paper provides an overview of autoimmune disorders of the central nervous system, specifically those caused by demyelination. We explore new research regarding potential therapeutic interventions, particularly those aimed at inducing remyelination. Remyelination is a detailed process, involving many cell types-oligodendrocyte precursor cells (OPCs), astrocytes, and microglia-and both the innate and adaptive immune systems. Our discussion of this process includes the differentiation potential of neural stem cells, the function of adult OPCs, and the impact of molecular mediators on myelin repair. Emerging therapies are also explored, with mechanisms of action including the induction of OPC differentiation, the transplantation of mesenchymal stem cells, and the use of molecular mediators. Further, we discuss current medical advancements in relation to many myelin-related disorders, including multiple sclerosis, optic neuritis, neuromyelitis optica spectrum disorder, myelin oligodendrocyte glycoprotein antibody-associated disease, transverse myelitis, and acute disseminated encephalomyelitis. Beyond these emerging systemic therapies, we also introduce the dimethyl fumarate/silk fibroin nerve conduit and its potential role in the treatment of peripheral nerve injuries. Despite these aforementioned scientific advancements, this paper maintains the need for ongoing research to deepen our understanding of demyelinating diseases and advance therapeutic strategies that enhance affected patients' quality of life.
Collapse
Affiliation(s)
- Robert Medina
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Ann-Marie Derias
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Maria Lakdawala
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Skye Speakman
- University of Florida College of Medicine, University of Florida, Gainesville, Fl 32610, United States
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, United States
| |
Collapse
|
36
|
Raj S, Sarangi P, Goyal D, Kumar H. The Hidden Hand in White Matter: Pericytes and the Puzzle of Demyelination. ACS Pharmacol Transl Sci 2024; 7:2912-2923. [PMID: 39421660 PMCID: PMC11480894 DOI: 10.1021/acsptsci.4c00192] [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: 04/03/2024] [Revised: 08/01/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024]
Abstract
Disruption of myelin, the fatty sheath-insulating nerve fibers in the white matter, blocks or slows the rapid transmission of electrical signals along nerve cells and contributes to several neurodegenerative diseases such as multiple sclerosis. Traditionally, research has focused on neuronal dysfunction as the primary factor, including autoimmunity, infections, inflammation, and genetic disorders causing demyelination. However, recent insights emphasize the critical role of pericytes, non-neuronal cells that regulate blood flow and maintain the health of blood vessels within white matter. This Perspective explores the principal mechanisms through which pericyte dysfunction contributes to damage and demyelination, including impaired communication with neurons (neurovascular uncoupling), excessive formation of scar tissue (fibrosis), and the infiltration of detrimental substances from the bloodstream. Understanding these mechanisms of pericyte-driven demyelination may lead to the creation of new therapeutic strategies for tackling a range of neurodegenerative conditions.
Collapse
Affiliation(s)
- Siddharth Raj
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Priyabrata Sarangi
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Divya Goyal
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| | - Hemant Kumar
- Department of Pharmacology
and Toxicology, National Institute of Pharmaceutical
Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat, India, 382355
| |
Collapse
|
37
|
Comi G, Dalla Costa G, Stankoff B, Hartung HP, Soelberg Sørensen P, Vermersch P, Leocani L. Assessing disease progression and treatment response in progressive multiple sclerosis. Nat Rev Neurol 2024; 20:573-586. [PMID: 39251843 DOI: 10.1038/s41582-024-01006-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/30/2024] [Indexed: 09/11/2024]
Abstract
Progressive multiple sclerosis poses a considerable challenge in the evaluation of disease progression and treatment response owing to its multifaceted pathophysiology. Traditional clinical measures such as the Expanded Disability Status Scale are limited in capturing the full scope of disease and treatment effects. Advanced imaging techniques, including MRI and PET scans, have emerged as valuable tools for the assessment of neurodegenerative processes, including the respective role of adaptive and innate immunity, detailed insights into brain and spinal cord atrophy, lesion dynamics and grey matter damage. The potential of cerebrospinal fluid and blood biomarkers is increasingly recognized, with neurofilament light chain levels being a notable indicator of neuro-axonal damage. Moreover, patient-reported outcomes are crucial for reflecting the subjective experience of disease progression and treatment efficacy, covering aspects such as fatigue, cognitive function and overall quality of life. The future incorporation of digital technologies and wearable devices in research and clinical practice promises to enhance our understanding of functional impairments and disease progression. This Review offers a comprehensive examination of these diverse evaluation tools, highlighting their combined use in accurately assessing disease progression and treatment efficacy in progressive multiple sclerosis, thereby guiding more effective therapeutic strategies.
Collapse
Affiliation(s)
- Giancarlo Comi
- Department of Neurorehabilitation Sciences, Casa di Cura Igea, Milan, Italy.
| | | | - Bruno Stankoff
- Sorbonne Université, Paris Brain Institute, Institut du Cerveau et de la Moelle Épinière, Centre National de la Recherche Scientifique, Inserm, Paris, France
| | - Hans-Peter Hartung
- Brain and Mind Center, University of Sydney, Sydney, Australia
- Department of Neurology, Palacky University Olomouc, Olomouc, Czech Republic
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Per Soelberg Sørensen
- Department of Neurology, Danish Multiple Sclerosis Center, University of Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Patrick Vermersch
- University of Lille, Inserm U1172, Lille Neuroscience & Cognition, Centre Hospitalier Universitaire de Lille, Fédération Hospitalo-Universitaire Precision Medicine in Psychiatry, Lille, France
| | - Letizia Leocani
- Vita-Salute San Raffaele University, Milan, Italy
- Multiple Sclerosis Center, Casa di Cura Igea, Milan, Italy
| |
Collapse
|
38
|
Du J, Yin Y, Wu D, Diao C, Zhao T, Peng F, Li N, Wang D, Shi J, Wang L, Kong L, Zhou W, Hao A. SIRT6 modulates lesion microenvironment in LPC induced demyelination by targeting astrocytic CHI3L1. J Neuroinflammation 2024; 21:243. [PMID: 39342313 PMCID: PMC11438192 DOI: 10.1186/s12974-024-03241-1] [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: 06/17/2024] [Accepted: 09/23/2024] [Indexed: 10/01/2024] Open
Abstract
Demyelination occurs widely in the central nervous system (CNS) neurodegenerative diseases, especially the multiple sclerosis (MS), which with a complex and inflammatory lesion microenvironment inhibiting remyelination. Sirtuin6 (SIRT6), a histone/protein deacetylase is of interest for its promising effect in transcriptional regulation, cell cycling, inflammation, metabolism and longevity. Here we show that SIRT6 participates in the remyelination process in mice subjected to LPC-induced demyelination. Using pharmacological SIRT6 inhibitor or activator, we found that SIRT6 modulated LPC-induced damage in motor or cognitive function. Inhibition of SIRT6 impaired myelin regeneration, exacerbated neurological deficits, and decreased oligodendrocyte precursor cells (OPCs) proliferation and differentiation, whereas activation of SIRT6 reversed behavioral performance in mice, demonstrating a beneficial effect of SIRT6. Importantly, based on RNA sequencing analysis of the corpus callosum tissues, it was further revealed that SIRT6 took charge in regulation of glial activation during remyelination, and significant alterations in CHI3L1 were obtained, a glycoprotein specifically secreted by astrocytes. Impaired proliferation and differentiation of OPCs could be induced in vitro using supernatants from reactive astrocyte, especially when SIRT6 was inhibited. Mechanistically, SIRT6 regulates the secretion of CHI3L1 from reactive astrocytes by histone-H3-lysine-9 acetylation (H3K9Ac). Adeno-associated virus-overexpression of SIRT6 (AAV-SIRT6-OE) in astrocytes improved remyelination and functional recovery after LPC-induced demyelination, whereas together with AAV-CHI3L1-OE inhibits this therapeutic effect. Collectively, our data elucidate the role of SIRT6 in remyelination and further reveal astrocytic SIRT6/CHI3L1 as the key regulator for improving the remyelination environment, which may be a potential target for MS therapy.
Collapse
Affiliation(s)
- Jingyi Du
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Yue Yin
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Dong Wu
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Can Diao
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Tiantian Zhao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Fan Peng
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Naigang Li
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Dongshuang Wang
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Jiaming Shi
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China
| | - Liyan Wang
- School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong, 250012, China
| | - Liang Kong
- Department of Clinical Laboratory, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wenjuan Zhou
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China.
| | - Aijun Hao
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Mental Disorders and Intelligent Control, Department of Anatomy and Histoembryology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, 44#, Wenhua Xi Road, Jinan, Shandong, 250012, China.
| |
Collapse
|
39
|
Kråkenes T, Sandvik CE, Ytterdal M, Gavasso S, Evjenth EC, Bø L, Kvistad CE. The Therapeutic Potential of Exosomes from Mesenchymal Stem Cells in Multiple Sclerosis. Int J Mol Sci 2024; 25:10292. [PMID: 39408622 PMCID: PMC11477223 DOI: 10.3390/ijms251910292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/17/2024] [Accepted: 09/23/2024] [Indexed: 10/20/2024] Open
Abstract
Although treatment for multiple sclerosis (MS) has undergone a revolution in the last decades, at least two important barriers remain: alleviation of innate inflammation driving disease progression and promotion of remyelination and neural regeneration. Mesenchymal stem cells (MSCs) possess immunomodulatory properties and promote remyelination in murine MS models. The main therapeutic mechanism has, however, been attributed to their potent paracrine capacity, and not to in vivo tissue implantation. Studies have demonstrated that exosomes released as part of the cells' secretome effectively encapsulate the beneficial properties of MSCs. These membrane-enclosed nanoparticles contain a variety of proteins and nucleic acids and serve as mediators of intercellular communication. In vitro studies have demonstrated that exosomes from MSCs modulate activated microglia from an inflammatory to an anti-inflammatory phenotype and thereby dampen the innate inflammation. Rodent studies have also demonstrated potent immunomodulation and remyelination with improved outcomes following exosome administration. Thus, exosomes from MSCs may represent a potential cell-free treatment modality to prevent disease progression and promote remyelination in MS. In this narrative review, we summarize the current knowledge of exosomes from MSCs as a potential treatment for MS and discuss the remaining issues before successful translation into clinical trials.
Collapse
Affiliation(s)
- Torbjørn Kråkenes
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| | - Casper Eugen Sandvik
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
| | - Marie Ytterdal
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| | - Sonia Gavasso
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
| | - Elisabeth Claire Evjenth
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| | - Lars Bø
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
- Department of Clinical Medicine, Faculty of Medicine, University of Bergen, 5021 Bergen, Norway
| | - Christopher Elnan Kvistad
- Neuro-SysMed, Department of Neurology, Haukeland University Hospital, 5021 Bergen, Norway (L.B.); (C.E.K.)
| |
Collapse
|
40
|
Tiwari V, Prajapati B, Asare Y, Damkou A, Ji H, Liu L, Naser N, Gouna G, Leszczyńska KB, Mieczkowski J, Dichgans M, Wang Q, Kawaguchi R, Shi Z, Swarup V, Geschwind DH, Prinz M, Gokce O, Simons M. Innate immune training restores pro-reparative myeloid functions to promote remyelination in the aged central nervous system. Immunity 2024; 57:2173-2190.e8. [PMID: 39053462 DOI: 10.1016/j.immuni.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 11/21/2023] [Accepted: 07/01/2024] [Indexed: 07/27/2024]
Abstract
The reduced ability of the central nervous system to regenerate with increasing age limits functional recovery following demyelinating injury. Previous work has shown that myelin debris can overwhelm the metabolic capacity of microglia, thereby impeding tissue regeneration in aging, but the underlying mechanisms are unknown. In a model of demyelination, we found that a substantial number of genes that were not effectively activated in aged myeloid cells displayed epigenetic modifications associated with restricted chromatin accessibility. Ablation of two class I histone deacetylases in microglia was sufficient to restore the capacity of aged mice to remyelinate lesioned tissue. We used Bacillus Calmette-Guerin (BCG), a live-attenuated vaccine, to train the innate immune system and detected epigenetic reprogramming of brain-resident myeloid cells and functional restoration of myelin debris clearance and lesion recovery. Our results provide insight into aging-associated decline in myeloid function and how this decay can be prevented by innate immune reprogramming.
Collapse
Affiliation(s)
- Vini Tiwari
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Bharat Prajapati
- Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden
| | - Yaw Asare
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Alkmini Damkou
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Hao Ji
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Lu Liu
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Nawraa Naser
- Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Garyfallia Gouna
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Katarzyna B Leszczyńska
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02093 Warsaw, Poland
| | - Jakub Mieczkowski
- Laboratory of Molecular Neurobiology, Nencki Institute of Experimental Biology of the Polish Academy of Sciences, 02093 Warsaw, Poland; 3P-Medicine Laboratory, Medical University of Gdańsk, 80211 Gdańsk, Poland
| | - Martin Dichgans
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany
| | - Qing Wang
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Riki Kawaguchi
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zechuan Shi
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Vivek Swarup
- Department of Neurobiology and Behavior, University of California, Irvine, CA, USA
| | - Daniel H Geschwind
- Departments of Neurology and Human Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marco Prinz
- Institute of Neuropathology, Faculty of Medicine, University of Freiburg, 79085 Freiburg, Germany; Signalling Research Centres BIOSS and CIBSS, University of Freiburg, 79104 Freiburg, Germany
| | - Ozgun Gokce
- Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany; Department of Neurodegenerative Diseases and Geriatric Psychiatry, University Hospital Bonn, 53127 Bonn, Germany
| | - Mikael Simons
- Institute of Neuronal Cell Biology, Technical University Munich, 81377 Munich, Germany; German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany; Munich Cluster of Systems Neurology (SyNergy), 81377 Munich, Germany; Institute for Stroke and Dementia Research, University Hospital of Munich, LMU Munich, 81377 Munich, Germany.
| |
Collapse
|
41
|
Theophanous S, Sargiannidou I, Kleopa KA. Glial Cells as Key Regulators in Neuroinflammatory Mechanisms Associated with Multiple Sclerosis. Int J Mol Sci 2024; 25:9588. [PMID: 39273535 PMCID: PMC11395575 DOI: 10.3390/ijms25179588] [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: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Even though several highly effective treatments have been developed for multiple sclerosis (MS), the underlying pathological mechanisms and drivers of the disease have not been fully elucidated. In recent years, there has been a growing interest in studying neuroinflammation in the context of glial cell involvement as there is increasing evidence of their central role in disease progression. Although glial cell communication and proper function underlies brain homeostasis and maintenance, their multiple effects in an MS brain remain complex and controversial. In this review, we aim to provide an overview of the contribution of glial cells, oligodendrocytes, astrocytes, and microglia in the pathology of MS during both the activation and orchestration of inflammatory mechanisms, as well as of their synergistic effects during the repair and restoration of function. Additionally, we discuss how the understanding of glial cell involvement in MS may provide new therapeutic targets either to limit disease progression or to facilitate repair.
Collapse
Affiliation(s)
- Styliani Theophanous
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
- Center for Multiple Sclerosis and Related Disorders, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| |
Collapse
|
42
|
Armstrong RC, Sullivan GM, Perl DP, Rosarda JD, Radomski KL. White matter damage and degeneration in traumatic brain injury. Trends Neurosci 2024; 47:677-692. [PMID: 39127568 DOI: 10.1016/j.tins.2024.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/17/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024]
Abstract
Traumatic brain injury (TBI) is a complex condition that can resolve over time but all too often leads to persistent symptoms, and the risk of poor patient outcomes increases with aging. TBI damages neurons and long axons within white matter tracts that are critical for communication between brain regions; this causes slowed information processing and neuronal circuit dysfunction. This review focuses on white matter injury after TBI and the multifactorial processes that underlie white matter damage, potential for recovery, and progression of degeneration. A multiscale perspective across clinical and preclinical advances is presented to encourage interdisciplinary insights from whole-brain neuroimaging of white matter tracts down to cellular and molecular responses of axons, myelin, and glial cells within white matter tissue.
Collapse
Affiliation(s)
- Regina C Armstrong
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Military Traumatic Brain Injury Initiative (MTBI(2)), Bethesda, MD, USA.
| | - Genevieve M Sullivan
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Military Traumatic Brain Injury Initiative (MTBI(2)), Bethesda, MD, USA; Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| | - Daniel P Perl
- Pathology, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA; Department of Defense - Uniformed Services University Brain Tissue Repository, Bethesda, MD, USA
| | - Jessica D Rosarda
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Kryslaine L Radomski
- Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| |
Collapse
|
43
|
Jansen MI, Mahmood Y, Lee J, Broome ST, Waschek JA, Castorina A. Targeting the PAC1 receptor mitigates degradation of myelin and synaptic markers and diminishes locomotor deficits in the cuprizone demyelination model. J Neurochem 2024; 168:3250-3267. [PMID: 39115025 DOI: 10.1111/jnc.16199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/19/2024] [Accepted: 07/25/2024] [Indexed: 10/04/2024]
Abstract
Multiple sclerosis (MS) is a demyelinating disease of the central nervous system with a strong neuroinflammatory component. Current treatments principally target the immune system but fail to preserve long-term myelin health and do not prevent neurological decline. Studies over the past two decades have shown that the structurally related neuropeptides VIP and PACAP (vasoactive intestinal peptide and pituitary adenylate cyclase-activating polypeptide, respectively) exhibit pronounced anti-inflammatory activities and reduce clinical symptoms in MS disease models, largely via actions on their bivalent VIP receptor type 1 and 2. Here, using the cuprizone demyelination model, we demonstrate that PACAP and VIP, and strikingly the PACAP-selective receptor PAC1 agonist maxadilan, prevented locomotor deficits in the horizontal ladder and open field tests. Moreover, only PACAP and maxadilan were able to prevent myelin deterioration, as assessed by a reduction in the expression of the myelin markers proteolipid protein 1, oligodendrocyte transcription factor 2, quaking-7 (APC) and Luxol Fast Blue staining. Furthermore, PACAP and maxadilan (but not VIP), prevented striatal synaptic loss and diminished astrocyte and microglial activation in the corpus callosum of cuprizone-fed mice. In vitro, PACAP or maxadilan prevented lipopolysaccharide (LPS)-induced polarisation of primary astrocytes at 12-24 h, an effect that was not seen with maxadilan in LPS-stimulated microglia. Taken together, our data demonstrates for the first time that PAC1 agonists provide distinctive protective effects against white matter deterioration, neuroinflammation and consequent locomotor dysfunctions in the cuprizone model. The results indicate that targeting the PAC1 receptor may provide a path to treat myelin-related diseases in humans.
Collapse
Affiliation(s)
- Margo I Jansen
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Yasir Mahmood
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Jordan Lee
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Sarah Thomas Broome
- Semel Institute for Neuroscience and Human Behavior/Neuropsychiatric Institute, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, California, USA
| | - James A Waschek
- Semel Institute for Neuroscience and Human Behavior/Neuropsychiatric Institute, Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, Los Angeles, California, USA
| | - Alessandro Castorina
- Laboratory of Cellular and Molecular Neuroscience, School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
44
|
Krsek A, Jagodic A, Baticic L. Nanomedicine in Neuroprotection, Neuroregeneration, and Blood-Brain Barrier Modulation: A Narrative Review. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1384. [PMID: 39336425 PMCID: PMC11433843 DOI: 10.3390/medicina60091384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/19/2024] [Accepted: 08/22/2024] [Indexed: 09/30/2024]
Abstract
Nanomedicine is a newer, promising approach to promote neuroprotection, neuroregeneration, and modulation of the blood-brain barrier. This review includes the integration of various nanomaterials in neurological disorders. In addition, gelatin-based hydrogels, which have huge potential due to biocompatibility, maintenance of porosity, and enhanced neural process outgrowth, are reviewed. Chemical modification of these hydrogels, especially with guanidine moieties, has shown improved neuron viability and underscores tailored biomaterial design in neural applications. This review further discusses strategies to modulate the blood-brain barrier-a factor critically associated with the effective delivery of drugs to the central nervous system. These advances bring supportive solutions to the solving of neurological conditions and innovative therapies for their treatment. Nanomedicine, as applied to neuroscience, presents a significant leap forward in new therapeutic strategies that might help raise the treatment and management of neurological disorders to much better levels. Our aim was to summarize the current state-of-knowledge in this field.
Collapse
Affiliation(s)
- Antea Krsek
- Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia;
| | - Ana Jagodic
- Department of Family Medicine, Community Health Center Krapina, 49000 Krapina, Croatia;
| | - Lara Baticic
- Department of Medical Chemistry, Biochemistry and Clinical Chemistry, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
45
|
Jörg LM, Schlötzer-Schrehardt U, Lefebvre V, Sock E, Wegner M. Transcription Factors Sox8 and Sox10 Contribute with Different Importance to the Maintenance of Mature Oligodendrocytes. Int J Mol Sci 2024; 25:8754. [PMID: 39201442 PMCID: PMC11354551 DOI: 10.3390/ijms25168754] [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: 07/17/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 09/02/2024] Open
Abstract
Myelin-forming oligodendrocytes in the vertebrate nervous system co-express the transcription factor Sox10 and its paralog Sox8. While Sox10 plays crucial roles throughout all stages of oligodendrocyte development, including terminal differentiation, the loss of Sox8 results in only mild and transient perturbations. Here, we aimed to elucidate the roles and interrelationships of these transcription factors in fully differentiated oligodendrocytes and myelin maintenance in adults. For that purpose, we conducted targeted deletions of Sox10, Sox8, or both in the brains of two-month-old mice. Three weeks post-deletion, none of the resulting mouse mutants exhibited significant alterations in oligodendrocyte numbers, myelin sheath counts, myelin ultrastructure, or myelin protein levels in the corpus callosum, despite efficient gene inactivation. However, differences were observed in the myelin gene expression in mice with Sox10 or combined Sox8/Sox10 deletion. RNA-sequencing analysis on dissected corpus callosum confirmed substantial alterations in the oligodendrocyte expression profile in mice with combined deletion and more subtle changes in mice with Sox10 deletion alone. Notably, Sox8 deletion did not affect any aspects of the expression profile related to the differentiated state of oligodendrocytes or myelin integrity. These findings extend our understanding of the roles of Sox8 and Sox10 in oligodendrocytes into adulthood and have important implications for the functional relationship between the paralogs and the underlying molecular mechanisms.
Collapse
Affiliation(s)
- Lisa Mirja Jörg
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D91054 Erlangen, Germany; (L.M.J.); (E.S.)
| | | | - Véronique Lefebvre
- Department of Surgery, Division of Orthopaedic Surgery, Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA;
| | - Elisabeth Sock
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D91054 Erlangen, Germany; (L.M.J.); (E.S.)
| | - Michael Wegner
- Institut für Biochemie, Friedrich-Alexander-Universität Erlangen-Nürnberg, D91054 Erlangen, Germany; (L.M.J.); (E.S.)
| |
Collapse
|
46
|
Al Jaf AIA, Peria S, Fabiano T, Ragnini-Wilson A. Remyelinating Drugs at a Crossroad: How to Improve Clinical Efficacy and Drug Screenings. Cells 2024; 13:1326. [PMID: 39195216 PMCID: PMC11352944 DOI: 10.3390/cells13161326] [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: 08/01/2024] [Accepted: 08/06/2024] [Indexed: 08/29/2024] Open
Abstract
Axons wrapped around the myelin sheath enable fast transmission of neuronal signals in the Central Nervous System (CNS). Unfortunately, myelin can be damaged by injury, viral infection, and inflammatory and neurodegenerative diseases. Remyelination is a spontaneous process that can restore nerve conductivity and thus movement and cognition after a demyelination event. Cumulative evidence indicates that remyelination can be pharmacologically stimulated, either by targeting natural inhibitors of Oligodendrocyte Precursor Cells (OPCs) differentiation or by reactivating quiescent Neural Stem Cells (qNSCs) proliferation and differentiation in myelinating Oligodendrocytes (OLs). Although promising results were obtained in animal models for demyelination diseases, none of the compounds identified have passed all the clinical stages. The significant number of patients who could benefit from remyelination therapies reinforces the urgent need to reassess drug selection approaches and develop strategies that effectively promote remyelination. Integrating Artificial Intelligence (AI)-driven technologies with patient-derived cell-based assays and organoid models is expected to lead to novel strategies and drug screening pipelines to achieve this goal. In this review, we explore the current literature on these technologies and their potential to enhance the identification of more effective drugs for clinical use in CNS remyelination therapies.
Collapse
Affiliation(s)
- Aland Ibrahim Ahmed Al Jaf
- Department of Biology, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Simone Peria
- Department of Biology, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| | - Tommaso Fabiano
- Department of Biology, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy
- Department of Life Sciences, University of Trieste, 34127 Trieste, Italy
| | - Antonella Ragnini-Wilson
- Department of Biology, University of Rome “Tor Vergata”, Via della Ricerca Scientifica 1, 00133 Rome, Italy
| |
Collapse
|
47
|
Yang C, Ma Y, Lu Q, Qu Y, Li Y, Cheng S, Xiao C, Chen J, Wang C, Wang F, Xiang AP, Huang W, Tang X, Zheng H. 2-Bromo-1,4-Naphthalenedione promotes CD8 + T cell expansion and limits Th1/Th17 to mitigate experimental autoimmune encephalomyelitis. J Neuroinflammation 2024; 21:181. [PMID: 39068463 PMCID: PMC11283727 DOI: 10.1186/s12974-024-03172-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 07/11/2024] [Indexed: 07/30/2024] Open
Abstract
Treating Multiple sclerosis (MS), a well-known immune-mediated disease characterized by axonal demyelination, is challenging due to its complex causes. Naphthalenedione, present in numerous plants, is being explored as a potential medicine for MS due to its immunomodulatory properties. However, its effects on lymphocytes can vary depending on factors such as the specific compound, concentration, and experimental conditions. In this study, we aim to explore the therapeutic potential of 2-bromo-1,4-naphthalenedione (BrQ), a derivative of naphthalenedione, in experimental autoimmune encephalomyelitis (EAE), an animal model of MS, and to elucidate its underlying mechanisms. We observed that mice treated with BrQ exhibited reduced severity of EAE symptoms, including lower clinical scores, decreased leukocyte infiltration, and less extensive demyelination in central nervous system. Furthermore, it was noted that BrQ does not directly affect the remyelination process. Through cell-chat analysis based on bulk RNA-seq data, coupled with validation of flow analysis, we discovered that BrQ significantly promotes the expansion of CD8+ T cells and their interactions with other immune cells in peripheral immune system in EAE mice. Subsequent CD8+ T cell depletion experiments confirmed that BrQ alleviates EAE in a CD8+ T cell-dependent manner. Mechanistically, expanded CD8+ cells were found to selectively reduce antigen-specific CD4+ cells and subsequently inhibit Th1 and Th17 cell development in vivo, ultimately leading to relief from EAE. In summary, our findings highlight the crucial role of BrQ in modulating the pathogenesis of MS, suggesting its potential as a novel drug candidate for treating MS and other autoimmune diseases.
Collapse
Affiliation(s)
- Cuixia Yang
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, China
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Yuanchen Ma
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Qiying Lu
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Yuliang Qu
- Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-sen University, Ministry of Education, Guangzhou, China
| | - Yuantao Li
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-Sen University, Ministry of Education, Guangzhou, China
| | - Shimei Cheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Chongjun Xiao
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Jinshuo Chen
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Chuangjia Wang
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China
| | - Feng Wang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Andy Peng Xiang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-Sen University, Ministry of Education, Guangzhou, China
- Department of Biochemistry, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Weijun Huang
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Sun Yat-Sen University, Ministry of Education, Guangzhou, China.
| | - Xiaorong Tang
- Central Laboratory, Chaozhou Central Hospital Affiliated to Southern Medical University, Chaozhou, Guangdong Province, China.
| | - Haiqing Zheng
- Department of Rehabilitation Medicine, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, China.
| |
Collapse
|
48
|
Zveik O, Rechtman A, Ganz T, Vaknin-Dembinsky A. The interplay of inflammation and remyelination: rethinking MS treatment with a focus on oligodendrocyte progenitor cells. Mol Neurodegener 2024; 19:53. [PMID: 38997755 PMCID: PMC11245841 DOI: 10.1186/s13024-024-00742-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 07/01/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Multiple sclerosis (MS) therapeutic goals have traditionally been dichotomized into two distinct avenues: immune-modulatory-centric interventions and pro-regenerative strategies. Oligodendrocyte progenitor cells (OPCs) were regarded for many years solely in concern to their potential to generate oligodendrocytes and myelin in the central nervous system (CNS). However, accumulating data elucidate the multifaceted roles of OPCs, including their immunomodulatory functions, positioning them as cardinal constituents of the CNS's immune landscape. MAIN BODY In this review, we will discuss how the two therapeutic approaches converge. We present a model by which (1) an inflammation is required for the appropriate pro-myelinating immune function of OPCs in the chronically inflamed CNS, and (2) the immune function of OPCs is crucial for their ability to differentiate and promote remyelination. This model highlights the reciprocal interactions between OPCs' pro-myelinating and immune-modulating functions. Additionally, we review the specific effects of anti- and pro-inflammatory interventions on OPCs, suggesting that immunosuppression adversely affects OPCs' differentiation and immune functions. CONCLUSION We suggest a multi-systemic therapeutic approach, which necessitates not a unidimensional focus but a harmonious balance between OPCs' pro-myelinating and immune-modulatory functions.
Collapse
Affiliation(s)
- Omri Zveik
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Ariel Rechtman
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Tal Ganz
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel
| | - Adi Vaknin-Dembinsky
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, 91120, Israel.
- The Department of Neurology and Laboratory of Neuroimmunology, The Agnes-Ginges Center for Human Neurogenetics, Hadassah-Hebrew University Medical Center, Ein-Kerem P.O.B. 12000, Jerusalem, 91120, Israel.
| |
Collapse
|
49
|
Li C, Luo Y, Li S. The roles of neural stem cells in myelin regeneration and repair therapy after spinal cord injury. Stem Cell Res Ther 2024; 15:204. [PMID: 38978125 PMCID: PMC11232222 DOI: 10.1186/s13287-024-03825-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/02/2024] [Indexed: 07/10/2024] Open
Abstract
Spinal cord injury (SCI) is a complex tissue injury that results in a wide range of physical deficits, including permanent or progressive disabilities of sensory, motor and autonomic functions. To date, limitations in current clinical treatment options can leave SCI patients with lifelong disabilities. There is an urgent need to develop new therapies for reconstructing the damaged spinal cord neuron-glia network and restoring connectivity with the supraspinal pathways. Neural stem cells (NSCs) possess the ability to self-renew and differentiate into neurons and neuroglia, including oligodendrocytes, which are cells responsible for the formation and maintenance of the myelin sheath and the regeneration of demyelinated axons. For these properties, NSCs are considered to be a promising cell source for rebuilding damaged neural circuits and promoting myelin regeneration. Over the past decade, transplantation of NSCs has been extensively tested in a variety of preclinical models of SCI. This review aims to highlight the pathophysiology of SCI and promote the understanding of the role of NSCs in SCI repair therapy and the current advances in pathological mechanism, pre-clinical studies, as well as clinical trials of SCI via NSC transplantation therapeutic strategy. Understanding and mastering these frontier updates will pave the way for establishing novel therapeutic strategies to improve the quality of recovery from SCI.
Collapse
Affiliation(s)
- Chun Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, Tongji University School of Medicine, Shanghai, 200092, China
| | - Yuping Luo
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Siguang Li
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Department of Neurology, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| |
Collapse
|
50
|
Zhou Y, Wang X, Yin W, Li Y, Guo Y, Chen C, Boltze J, Liesz A, Sparwasser T, Wen D, Yu W, Li P. Perioperative stroke deteriorates white matter integrity by enhancing cytotoxic CD8 + T-cell activation. CNS Neurosci Ther 2024; 30:e14747. [PMID: 38973085 PMCID: PMC11227991 DOI: 10.1111/cns.14747] [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: 02/04/2024] [Revised: 03/28/2024] [Accepted: 04/17/2024] [Indexed: 07/09/2024] Open
Abstract
AIM To explore the regulatory mechanisms of microglia-mediated cytotoxic CD8+ T-cell infiltration in the white matter injury of perioperative stroke (PIS). METHODS Adult male C57BL/6 mice were subjected to ileocolic bowel resection (ICR) 24 h prior to permanent distant middle cerebral artery occlusion (dMCAO) to establish model PIS. White matter injury, functional outcomes, peripheral immune cell infiltration, and microglia phenotype were assessed up to 28 days after dMCAO using behavioral phenotyping, immunofluorescence staining, transmission electron microscopy, western blot, and FACS analysis. RESULTS We found surgery aggravated white matter injury and deteriorated sensorimotor deficits up to 28 days following PIS. The PIS mice exhibited significantly increased activation of peripheral and central CD8+ T cells, while significantly reduced numbers of mature oligodendrocytes compared to IS mice. Neutralizing CD8+ T cells partly reversed the aggravated demyelination following PIS. Pharmacological blockage or genetic deletion of receptor-interacting protein kinase 1 (RIPK1) activity could alleviate CD8+ T-cell infiltration and demyelination in PIS mice. CONCLUSION Surgery exacerbates demyelination and worsens neurological function by promoting infiltration of CD8+ T cells and microglia necroptosis, suggesting that modulating interactions of CD8+ T cells and microglia could be a novel therapeutic target of long-term neurological deficits of PIS.
Collapse
Affiliation(s)
- Yuxi Zhou
- Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghaiChina
| | - Xin Wang
- Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghaiChina
| | - Wen Yin
- Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghaiChina
| | - Yan Li
- Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghaiChina
| | - Yunlu Guo
- Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghaiChina
| | - Chen Chen
- Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghaiChina
| | | | - Arthur Liesz
- Institute for Stroke and Dementia Research (ISD), University HospitalLMU MunichMunichGermany
- Munich Cluster for Systems Neurology (SyNergy)MunichGermany
| | - Tim Sparwasser
- Institute of Medical Microbiology and HygieneUniversity Medical Center of the Johannes Gutenberg‐University MainzMainzGermany
- Research Center for Immunotherapy (FZI)University Medical Center, Johannes Gutenberg‐University MainzMainzGermany
| | - Daxiang Wen
- Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghaiChina
| | - Weifeng Yu
- Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghaiChina
| | - Peiying Li
- Department of Anesthesiology, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Key Laboratory of Anesthesiology (Shanghai Jiao Tong University)Ministry of EducationShanghaiChina
- Clinical Research Center, Renji HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Outcomes Research ConsortiumClevelandOhioUSA
| |
Collapse
|