1
|
Yang B, Li Z, Li P, Liu Y, Ding X, Feng E. Piezo1 in microglial cells: Implications for neuroinflammation and tumorigenesis. Channels (Austin) 2025; 19:2492161. [PMID: 40223276 PMCID: PMC12005408 DOI: 10.1080/19336950.2025.2492161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 04/04/2025] [Accepted: 04/07/2025] [Indexed: 04/15/2025] Open
Abstract
Microglia, the central nervous system (CNS) resident immune cells, are pivotal in regulating neurodevelopment, maintaining neural homeostasis, and mediating neuroinflammatory responses. Recent research has highlighted the importance of mechanotransduction, the process by which cells convert mechanical stimuli into biochemical signals, in regulating microglial activity. Among the various mechanosensitive channels, Piezo1 has emerged as a key player in microglia, influencing their behavior under both physiological and pathological conditions. This review focuses on the expression and role of Piezo1 in microglial cells, particularly in the context of neuroinflammation and tumorigenesis. We explore how Piezo1 mediates microglial responses to mechanical changes within the CNS, such as alterations in tissue stiffness and fluid shear stress, which are common in conditions like multiple sclerosis, Alzheimer's disease, cerebral ischemia, and gliomas. The review also discusses the potential of targeting Piezo1 for therapeutic intervention, given its involvement in the modulation of microglial activity and its impact on disease progression. This review integrates findings from recent studies to provide a comprehensive overview of Piezo1's mechanistic pathways in microglial function. These insights illuminate new possibilities for developing targeted therapies addressing CNS disorders with neuroinflammation and pathological tissue mechanics.
Collapse
Affiliation(s)
- Bo Yang
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Zhenyu Li
- Department of Neonatology, Children’s Medical Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Peiliang Li
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Yuhan Liu
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Xinghuan Ding
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| | - Enshan Feng
- Department of Neurosurgery, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
2
|
Malcangio M, Sideris-Lampretsas G. How microglia contribute to the induction and maintenance of neuropathic pain. Nat Rev Neurosci 2025; 26:263-275. [PMID: 40128335 DOI: 10.1038/s41583-025-00914-5] [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: 02/21/2025] [Indexed: 03/26/2025]
Abstract
Neuropathic pain is a debilitating condition caused by damage to the nervous system that results in changes along the pain pathway that lead to persistence of the pain sensation. Unremitting pain conditions are associated with maladaptive plasticity, disruption of neuronal activity that favours excitation over inhibition, and engagement of immune cells. The substantial progress made over the last two decades in the neuroimmune interaction research area points to a mechanistic role of spinal cord microglia, which are resident immune cells of the CNS. Microglia respond to and modulate neuronal activity during establishment and persistence of neuropathic pain states, and microglia-neuron pathways provide targets that can be exploited to attenuate abnormal neuronal activity and provide pain relief.
Collapse
Affiliation(s)
- Marzia Malcangio
- Wolfson Sensory, Pain and Regeneration Centre, King's College London, London, UK.
| | | |
Collapse
|
3
|
Xiao J, Meng Z, Lu Y, Nie Z, Liu Y, Yao Z, Zhang Y, Li L. Targeting microglia-Th17 feed-forward loop to suppress autoimmune neuroinflammation. J Neuroinflammation 2025; 22:118. [PMID: 40275354 DOI: 10.1186/s12974-025-03427-1] [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: 01/31/2025] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
Microglia and Th17 cells are the major immunopathogenic cells in multiple sclerosis and its animal model of immune aspects, experimental autoimmune encephalomyelitis (EAE). While studies have highlighted the distinct roles of microglia and Th17 cells in EAE, it remains unclear whether microglia, as potential professional antigen-presenting cells, activate and stabilize the effector program of EAE-pathogenic Th17 cells in vivo; and if so, whether the Th17 could in turn reinforce the active state of the microglia. Our data demonstrate in an array of mouse models, including active/passive-EAE and transgenic mice, a microglia-Th17 feed-forward activation loop drives EAE disease progression through a mechanism dependent on both MHC-II, proinflammatory cytokines, inflammatory chemokines as well as STING→NF-κB pathway in the microglia and effector cytokines produced by the pathogenic Th17 cells. We also captured and identified the molecular properties of the feed-forward loop, which are two-cell entities of microglia-Th17, and proved them as the functional units of antigen presentation and bi-directional activation between the two cell types. Moreover, ACT001, an orphan drug to treat glioblastoma, disrupts this feed-forward activation loop by inhibiting the STING→NF-κB pathway in microglia, thereby alleviating EAE. These findings emphasize the importance of interactions and bi-directional activations between microglia and Th17 in the autoimmune neuroinflammation, and provide rationale for further investigation on ACT001 as therapeutic option for autoimmune inflammatory diseases driven by similar mechanisms.
Collapse
MESH Headings
- Animals
- Microglia/drug effects
- Microglia/immunology
- Microglia/metabolism
- Th17 Cells/drug effects
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Mice
- Mice, Transgenic
- Mice, Inbred C57BL
- Neuroinflammatory Diseases/immunology
- Female
Collapse
Affiliation(s)
- Jun Xiao
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin, 300070, China
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Zihan Meng
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin, 300070, China
| | - Yao Lu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin, 300070, China
| | - Zongchang Nie
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin, 300070, China
| | - Yujie Liu
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin, 300070, China
| | - Zhi Yao
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin, 300070, China.
| | - Yingchi Zhang
- State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Institute of Hematology and Blood Diseases Hospital, National Clinical Research Center for Blood Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China.
| | - Long Li
- Tianjin Institute of Immunology, Key Laboratory of Immune Microenvironment and Disease (Ministry of Education), State Key Laboratory of Experimental Hematology, Department of Immunology, Tianjin Medical University, Tianjin, 300070, China.
| |
Collapse
|
4
|
Medeiros-Furquim T, Miedema A, Schilder E, Brouwer N, Holtman IR, Kooistra SM, Eggen BJL. Microglia endotoxin tolerance is retained after enforced repopulation. Brain Behav Immun 2025:S0889-1591(25)00144-8. [PMID: 40274001 DOI: 10.1016/j.bbi.2025.04.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 03/15/2025] [Accepted: 04/08/2025] [Indexed: 04/26/2025] Open
Abstract
Microglia are crucial for CNS homeostasis and are involved in a wide range of neurodegenerative and neuroinflammatory diseases. Systemic inflammation and infections can contribute to neurodegeneration later in life by affecting microglia. Like other innate immune cells, microglia can develop innate immune memory (IMM) in response to an inflammatory challenge, altering their response to subsequent stimuli. IMM can ameliorate or worsen CNS pathology, but it is unclear if IMM can be reversed to restore microglia functions. Here, we investigated whether microglia depletion-repopulation by inhibition of the colony-stimulating factor 1 receptor with BLZ945 reversed LPS-induced endotoxin tolerance in mice. Repopulated microglia displayed a reduced expression of homeostatic genes and genes related to mitochondrial respiration and TCA cycle metabolism and an increased expression of immune effector and activation genes. Nonetheless the blunted inflammatory gene response after LPS-preconditioning was retained after a depletion-repopulation cycle. Our study highlights the persistence of endotoxin tolerance in microglia after a depletion-repopulation cycle which might impact the potential effectiveness of strategies targeted at microglia depletion for clinical applications.
Collapse
Affiliation(s)
- Tiago Medeiros-Furquim
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Anneke Miedema
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Edwin Schilder
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Nieske Brouwer
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Inge R Holtman
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Susanne M Kooistra
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Bart J L Eggen
- Department of Biomedical Sciences, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
5
|
Ribeiro AR, Pereira R, Barros C, Barateiro A, Alberro A, Basto AP, Graça L, Pinto MV, Santos FMF, Gois PMP, Howlett SE, Fernandes A. Experimental autoimmune encephalomyelitis pathogenesis alters along animal age: impact of S100B expression. J Neuroimmune Pharmacol 2025; 20:37. [PMID: 40227512 PMCID: PMC11997003 DOI: 10.1007/s11481-025-10195-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/20/2025] [Indexed: 04/15/2025]
Abstract
Multiple Sclerosis (MS) is the leading inflammatory and non-traumatic cause of disability in young adults, with late-onset MS emerging in middle-aged patients often resulting in poorer treatment responses and worse prognoses. The calcium-binding protein S100B is elevated in MS patients, and its targeting has shown promise in reducing disease severity in experimental autoimmune encephalomyelitis (EAE) models. However, most studies on MS pathology have focused on young animal models, leaving a gap in understanding the effects of age and S100B ablation on disease progression throughout the lifespan. This study aimed to characterize EAE in mice of different ages, examining demyelination, inflammation, and immune responses to determine whether S100B ablation could mitigate MS pathogenesis across the lifespan. EAE was induced in six cohorts of C57BL/6 mice: young adults (3 months), older adults (6 months), and middle-aged (12 months), including corresponding S100B knockout (KO) groups, followed for 23 days. Upon sacrifice, spinal cords were assessed via immunohistochemistry and Real-Time qPCR, while splenocytes were analyzed for immune cell characterization. Results indicated a more severe disease course in 12-month-old mice, marked by increased gliosis, inflammation, and impaired microglial phagocytic activity. Notably, S100B absence reduced gliosis and inflammatory markers across all ages, with 12-month-old S100B KO mice showing increased regulatory T cells. These findings highlight the exacerbating role of age and elevated S100B in MS progression, underscoring the importance of identifying age-specific MS markers and therapeutic targets.
Collapse
MESH Headings
- Animals
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- S100 Calcium Binding Protein beta Subunit/genetics
- S100 Calcium Binding Protein beta Subunit/biosynthesis
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Female
- Aging/metabolism
- Aging/pathology
- Age Factors
- Spinal Cord/pathology
- Spinal Cord/metabolism
- Male
Collapse
Affiliation(s)
- Ana Rita Ribeiro
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Raquel Pereira
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Catarina Barros
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Andreia Barateiro
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
- Departamento de Ciências Farmacêuticas E Do Medicamento, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Ainhoa Alberro
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
- IIS Biogipuzkoa Health Research Institute, San Sebastian, Spain
| | - Afonso P Basto
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- CIISA - Centro de Investigação Interdisciplinar em Sanidade Animal, Faculdade de Medicina Veterinária, Universidade de Lisboa, Lisbon, Portugal
- Laboratório Associado Para a Ciência Animal E Veterinária (AL4AnimalS), Lisbon, Portugal
| | - Luís Graça
- Gulbenkian Institute for Molecular Medicine, Lisbon, Portugal
- Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Maria Vaz Pinto
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Fábio M F Santos
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
| | - Pedro M P Gois
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal
- Departamento de Ciências Farmacêuticas E Do Medicamento, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal
| | - Susan E Howlett
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
- Department of Medicine (Geriatric Medicine), Dalhousie University, Halifax, NS, Canada
| | - Adelaide Fernandes
- Faculdade de Farmácia, Research Institute for Medicines (iMed.ULisboa), Universidade de Lisboa, Lisbon, Portugal.
- Departamento de Ciências Farmacêuticas E Do Medicamento, Faculdade de Farmácia, Universidade de Lisboa, Lisbon, Portugal.
| |
Collapse
|
6
|
Oruk S, Ergul Erkec O, Huyut Z, Acikgoz E. Neuroprotective effects of ghrelin in cuprizone-induced rat model of multiple sclerosis. Metab Brain Dis 2025; 40:176. [PMID: 40214860 PMCID: PMC11991981 DOI: 10.1007/s11011-025-01603-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 04/05/2025] [Indexed: 04/14/2025]
Abstract
Multiple sclerosis (MS) is an inflammatory central nervous system disease characterized by demyelination and axonal loss and is the main cause of non-traumatic neurological disability in young adults. Although there are several treatment approaches to manage the disease, there is no definitive cure for multiple sclerosis. Inflammation and oxidative stress are known to play important roles in the pathophysiology of MS. Ghrelin, a peptide secreted by the stomach, is reported to have neuroprotective properties through several pathways, including attenuating oxidative stress and inflammation. In the present study cuprizone (CPZ)-induced model of MS was used in Wistar albino rats to study the possible anti-inflammatory, antioxidant and neuroprotective effects of ghrelin. Rats were randomly divided into six groups: Control groups (Control35 and Control-S42), demyelination group, remyelination group, remyelination + ghrelin (20 µg/kg) group and remyelination + ghrelin (40 µg/kg) group. Y maze test was performed on the rats on their last day of the experiment. Oxidative stress and inflammatory parameters were investigated in brain using commercial kits by enzyme-linked immunosorbent assay (ELISA). Luxol fast blue (LFB) and hematoxylen&eosin (H&E) staining were performed in brain tissues. CPZ leads to a significant decrease in glutathione peroxidase (GSH-Px) levels and myelin content and a significant increase in malondialdehyde (MDA), tumor necrosis factor-alpha (TNF-ɑ), interleukin- 6 (IL- 6) levels, the number of lymphatic cells and inflammatory cells. A significant increase in the antioxidant parameter levels and a significant decrease in MDA levels were found in the ghrelin treated groups (p < 0.05). CPZ leads to irregular, fragmented, demyelinating nerve fibers. A more significant remyelination was observed in the ghrelin treated groups compared to the other groups (p < 0.05). In conclusion, ghrelin treatment showed neuroprotective and antioxidant properties and reduced demyelination in the CPZ-induced rat model of multiple sclerosis.
Collapse
Affiliation(s)
- Sezai Oruk
- Department of Medical Physiology, Institute of Health Sciences, Van Yuzuncu Yil University, Van, Turkey
| | - Ozlem Ergul Erkec
- Department of Physiology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey.
| | - Zubeyir Huyut
- Department of Biochemistry, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| | - Eda Acikgoz
- Department of Histology and Embryology, Faculty of Medicine, Van Yuzuncu Yil University, Van, Turkey
| |
Collapse
|
7
|
Sertbas M, Ulgen KO. Exploring Human Brain Metabolism via Genome-Scale Metabolic Modeling with Highlights on Multiple Sclerosis. ACS Chem Neurosci 2025; 16:1346-1360. [PMID: 40091499 PMCID: PMC11969529 DOI: 10.1021/acschemneuro.5c00006] [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: 01/06/2025] [Revised: 02/18/2025] [Accepted: 03/03/2025] [Indexed: 03/19/2025] Open
Abstract
Cerebral dysfunctions give rise to a wide range of neurological diseases due to the structural and functional complexity of the human brain stemming from the interactive cellular metabolism of its specific cells, including neurons and glial cells. In parallel with advances in isolation and measurement technologies, genome-scale metabolic models (GEMs) have become a powerful tool in the studies of systems biology to provide critical insights into the understanding of sophisticated eukaryotic systems. In this study, brain cell-specific GEMs were reconstructed for neurons, astrocytes, microglia, oligodendrocytes, and oligodendrocyte precursor cells by integrating single-cell RNA-seq data and global Human1 via a task-driven integrative network inference for tissues (tINIT) algorithm. Then, intercellular reactions among neurons, astrocytes, microglia, and oligodendrocytes were added to generate a combined brain model, iHumanBrain2690. This brain network was used in the prediction of metabolic alterations in glucose, ketone bodies, oxygen change, and reporter metabolites. Glucose supplementation increased the subsystems' activities in glycolysis, and ketone bodies elevated those in the TCA cycle and oxidative phosphorylation. Reporter metabolite analysis identified L-carnitine and arachidonate as the top reporter metabolites in gray and white matter microglia in multiple sclerosis (MS), respectively. Carbamoyl-phosphate was found to be the top reporter metabolite in primary progressive MS. Taken together, single and integrated iHumanBrain2690 metabolic networks help us elucidate complex metabolism in brain physiology and homeostasis in health and disease.
Collapse
Affiliation(s)
- Mustafa Sertbas
- Department
of Chemical Engineering, Bogazici University, 34342 Istanbul, Turkey
- Department
of Chemical Engineering, Istanbul Technical
University, 34469 Istanbul, Turkey
| | - Kutlu O. Ulgen
- Department
of Chemical Engineering, Bogazici University, 34342 Istanbul, Turkey
| |
Collapse
|
8
|
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] [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
|
9
|
Saika F, Sato T, Nakabayashi T, Fukazawa Y, Hino S, Suzuki K, Kiguchi N. Male-Dominant Spinal Microglia Contribute to Neuropathic Pain by Producing CC-Chemokine Ligand 4 Following Peripheral Nerve Injury. Cells 2025; 14:484. [PMID: 40214438 PMCID: PMC11987877 DOI: 10.3390/cells14070484] [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: 02/25/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Recent studies have revealed marked sex differences in pathophysiological roles of spinal microglia in neuropathic pain, with microglia contributing to pain exacerbation exclusively in males. However, the characteristics of pain-enhancing microglia, which are more prominent in males, remain poorly understood. Here, we reanalyzed a previously published single-cell RNA sequencing dataset and identified a microglial subpopulation that significantly increases in the spinal dorsal horn (SDH) of male mice following peripheral nerve injury. CC-chemokine ligand 4 (CCL4) was highly expressed in this subpopulation and its mRNA levels were increased in the SDH after partial sciatic nerve ligation (PSL) only in male mice. Notably, CCL4 expression was reduced in male mice following microglial depletion, indicating that microglia are the primary source of CCL4. Intrathecal administration of maraviroc, an inhibitor of the CCL4-CC-chemokine receptor 5 (CCR5) signaling pathway, after PSL, significantly suppressed mechanical allodynia only in male mice. Furthermore, intrathecal administration of CCL4 induced mechanical allodynia in both sexes, accompanied by increased expression of c-fos, a neuronal excitation marker, in the SDH. These findings highlight a sex-biased difference in the gene expression profile of spinal microglia following peripheral nerve injury, with elevated CCL4 expression in male mice potentially contributing to pain exacerbation.
Collapse
Affiliation(s)
- Fumihiro Saika
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan;
- Faculty of Wakayama Health Care Sciences, Takarazuka University of Medical and Health Care, Wakayama 640-8392, Japan
| | - Tetsuya Sato
- H.U. Group Research Institute G.K., Tokyo 197-0833, Japan;
| | | | - Yohji Fukazawa
- Department of Anatomy, Kansai University of Health Sciences, Osaka 590-0482, Japan;
| | - Shinjiro Hino
- Department of Medical Cell Biology, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan;
| | - Kentaro Suzuki
- Faculty of Life and Environmental Sciences, University of Yamanashi, Yamanashi 400-8510, Japan;
| | - Norikazu Kiguchi
- Department of Physiological Sciences, School of Pharmaceutical Sciences, Wakayama Medical University, Wakayama 640-8156, Japan;
| |
Collapse
|
10
|
Wei Y, Zhou X, Li Z, Liu Q, Ding H, Zhou Y, Yin RF, Zheng L. Genetically Programmed Single-Component Protein Hydrogel for Spinal Cord Injury Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405054. [PMID: 39792612 PMCID: PMC11904991 DOI: 10.1002/advs.202405054] [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: 05/09/2024] [Revised: 12/02/2024] [Indexed: 01/12/2025]
Abstract
Protein self-assembly allows for the formation of diverse supramolecular materials from relatively simple building blocks. In this study, a single-component self-assembling hydrogel is developed using the recombinant protein CsgA, and its successful application for spinal cord injury repair is demonstrated. Gelation is achieved by the physical entanglement of CsgA nanofibrils, resulting in a self-supporting hydrogel at low concentrations (≥5 mg mL-1). By leveraging the programmability of the CsgA gene sequence, the bioactive hydrogel is enhanced by fusing functional peptide GHK. GHK is recognized for its anti-inflammatory, antioxidant, and neurotrophic factor-stimulating properties, making it a valuable addition to the hydrogel for spinal cord injury repair applications. In vitro experiments demonstrate that the CsgA-GHK hydrogel can modulate microglial M2 polarization, promote neuronal differentiation of neural stem cells, and inhibit astrocyte differentiation. Additionally, the hydrogel shows efficacy in alleviating inflammation and promotes neuronal regeneration at the injury site, leading to significant functional recovery in a rat model with compression injury spinal cord cavity. These findings lay the groundwork for developing a modular design platform for recombinant CsgA protein hydrogels in tissue repair applications.
Collapse
Affiliation(s)
- Yi Wei
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Xiaolin Zhou
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Zhenhua Li
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Qing Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Han Ding
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Yunlong Zhou
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| | - Ruo-Feng Yin
- China-Japan Union Hospital, Jilin University, Changchun, Jilin, 130031, China
| | - Lifei Zheng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| |
Collapse
|
11
|
Clarence T, Bendl J, Cao X, Wang X, Zheng S, Hoffman GE, Kozlenkov A, Hong A, Iskhakova M, Jaiswal MK, Murphy S, Yu A, Haroutunian V, Dracheva S, Akbarian S, Fullard JF, Yuan GC, Lee D, Roussos P. Multiomic single-cell profiling identifies critical regulators of postnatal brain. Nat Genet 2025; 57:591-603. [PMID: 39962241 DOI: 10.1038/s41588-025-02083-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 01/08/2025] [Indexed: 03/15/2025]
Abstract
Human brain development spans from embryogenesis to adulthood, with dynamic gene expression controlled by cell-type-specific cis-regulatory element activity and three-dimensional genome organization. To advance our understanding of postnatal brain development, we simultaneously profiled gene expression and chromatin accessibility in 101,924 single nuclei from four brain regions across ten donors, covering five key postnatal stages from infancy to late adulthood. Using this dataset and chromosome conformation capture data, we constructed enhancer-based gene regulatory networks to identify cell-type-specific regulators of brain development and interpret genome-wide association study loci for ten main brain disorders. Our analysis connected 2,318 cell-specific loci to 1,149 unique genes, representing 41% of loci linked to the investigated traits, and highlighted 55 genes influencing several disease phenotypes. Pseudotime analysis revealed distinct stages of postnatal oligodendrogenesis and their regulatory programs. These findings provide a comprehensive dataset of cell-type-specific gene regulation at critical timepoints in postnatal brain development.
Collapse
Affiliation(s)
- Tereza Clarence
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Jaroslav Bendl
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xuan Cao
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Xinyi Wang
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Shiwei Zheng
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexey Kozlenkov
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Aram Hong
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Marina Iskhakova
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Manoj K Jaiswal
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Sarah Murphy
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alexander Yu
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vahram Haroutunian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Stella Dracheva
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA
| | - Schahram Akbarian
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - John F Fullard
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Guo-Cheng Yuan
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Donghoon Lee
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Mental Illness Research Education and Clinical Center, James J. Peters VA Medical Center, Bronx, NY, USA.
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, NY, USA.
| |
Collapse
|
12
|
Rahdar S, Basir Z, Tabandeh MR, Ghotbeddin Z, Khazaeel K. Betaine alleviates cerebellar endoplasmic reticulum stress and oxidative imbalance in a cuprizone model of multiple sclerosis in rat. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:2651-2664. [PMID: 39249501 DOI: 10.1007/s00210-024-03381-4] [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: 05/02/2024] [Accepted: 08/14/2024] [Indexed: 09/10/2024]
Abstract
Multiple sclerosis (MS) is the most common demyelinating disease of the central nervous system, especially the cerebellum, with numerous physical and mental symptoms. Oxidative stress caused by inflammation can play a role in the occurrence of this disease. Betaine, a natural methyl donor compound, has potent neuroprotective effects. Here, we investigated the effects of betaine on motor behavior, cerebellar histological changes, oxidative stress response, and endoplasmic reticulum stress in a cuprizone (CPZ)-induced multiple sclerosis model in male rats. Twenty Wistar adult male rats were randomly divided into four groups including control, MS, betaine-treated MS, and betaine groups. MS was induced by feeding animals with rodent chow containing 0.5% CPZ for 12 weeks. Betaine was daily administrated as 1% in drinking water for the last 6 weeks. The motor behavioral performance was evaluated by open field, rotarod, and reverse basket tests. Histological analysis of the cerebellum was performed by hematoxylin and eosin (H&E) and Cresyl violet (Nissl) staining. Oxidative stress factors (GSH, GSSG, GPX, GR, and GT) were assessed in the experimental groups and finally, the expression of ERS-associated proteins was measured using western blot analysis. Data showed that treatment with betaine could effectively prevent and reverse the adverse behavioral manifestation compared with the MS group. Betaine treatment protected cerebellar demyelination and neuron and Purkinje cell degeneration against CPZ-induced demyelination. Betaine attenuated the protein levels of ESR-related proteins in the cerebellum of MS rats and similarly increased the level of enzymes related to antioxidants in the cerebellum. Therefore, our results suggest that oral administration of betaine may be used as a novel adjunct therapy against cerebellar dysfunctions in an animal model of MS.
Collapse
Affiliation(s)
- Samaneh Rahdar
- Department of Basic Sciences, Division of Histology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Zahra Basir
- Department of Basic Sciences, Division of Histology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran.
| | - Mohammad Reza Tabandeh
- Department of Basic Sciences, Division of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Stem cells and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Zohreh Ghotbeddin
- Stem cells and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Department of Basic Sciences, Division of Physiology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Kaveh Khazaeel
- Stem cells and Transgenic Technology Research Center, Shahid Chamran University of Ahvaz, Ahvaz, Iran
- Department of Basic Sciences, Division of Anatomy and Embryology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
13
|
Boudriot E, Stephan M, Rabe F, Smigielski L, Schmitt A, Falkai P, Ziller MJ, Rossner MJ, Homan P, Papiol S, Raabe FJ. Genetic Analysis of Retinal Cell Types in Neuropsychiatric Disorders. JAMA Psychiatry 2025; 82:285-295. [PMID: 39775833 PMCID: PMC11883512 DOI: 10.1001/jamapsychiatry.2024.4230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 10/29/2024] [Indexed: 01/11/2025]
Abstract
Importance As an accessible part of the central nervous system, the retina provides a unique window to study pathophysiological mechanisms of brain disorders in humans. Imaging and electrophysiological studies have revealed retinal alterations across several neuropsychiatric and neurological disorders, but it remains largely unclear which specific cell types and biological mechanisms are involved. Objective To determine whether specific retinal cell types are affected by genomic risk for neuropsychiatric and neurological disorders and to explore the mechanisms through which genomic risk converges in these cell types. Design, Setting, and Participants This genetic association study combined findings from genome-wide association studies in schizophrenia, bipolar disorder, major depressive disorder, multiple sclerosis, Parkinson disease, Alzheimer disease, and stroke with retinal single-cell transcriptomic datasets from humans, macaques, and mice. To identify susceptible cell types, Multi-Marker Analysis of Genomic Annotation (MAGMA) cell-type enrichment analyses were applied and subsequent pathway analyses performed. The cellular top hits were translated to the structural level using retinal optical coherence tomography (acquired between 2009 and 2010) and genotyping data in the large population-based UK Biobank cohort study. Data analysis was conducted between 2022 and 2024. Main Outcomes and Measures Cell type-specific enrichment of genetic risk loading for neuropsychiatric and neurological disorder traits in the gene expression profiles of retinal cells. Results Expression profiles of amacrine cells (interneurons within the retina) were robustly enriched in schizophrenia genetic risk across mammalian species and in different developmental stages. This enrichment was primarily driven by genes involved in synapse biology. Moreover, expression profiles of retinal immune cell populations were enriched in multiple sclerosis genetic risk. No consistent cell-type associations were found for bipolar disorder, major depressive disorder, Parkinson disease, Alzheimer disease, or stroke. On the structural level, higher polygenic risk for schizophrenia was associated with thinning of the ganglion cell inner plexiform layer, which contains dendrites and synaptic connections of amacrine cells (B, -0.09; 95% CI, -0.16 to -0.03; P = .007; n = 36 349; mean [SD] age, 57.50 [8.00] years; 19 859 female [54.63%]). Higher polygenic risk for multiple sclerosis was associated with increased thickness of the retinal nerve fiber layer (B, 0.06; 95% CI, 0.02 to 0.10; P = .007; n = 36 371; mean [SD] age, 57.51 [8.00] years; 19 843 female [54.56%]). Conclusions and Relevance This study provides novel insights into the cellular underpinnings of retinal alterations in neuropsychiatric and neurological disorders and highlights the retina as a potential proxy to study synaptic pathology in schizophrenia.
Collapse
Affiliation(s)
- Emanuel Boudriot
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Marius Stephan
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- Systasy Bioscience, Munich, Germany
| | - Finn Rabe
- Department of Adult Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Lukasz Smigielski
- Department of Child and Adolescent Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Andrea Schmitt
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- German Center for Mental Health, Partner Site Munich-Augsburg, Germany
- Laboratory of Neurosciences, Institute of Psychiatry, University of São Paulo, São Paulo, Brazil
| | - Peter Falkai
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- German Center for Mental Health, Partner Site Munich-Augsburg, Germany
| | - Michael J. Ziller
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry, University of Münster, Münster, Germany
- Center for Soft Nanoscience, University of Münster, Münster, Germany
| | - Moritz J. Rossner
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
- Systasy Bioscience, Munich, Germany
| | - Philipp Homan
- Department of Adult Psychiatry and Psychotherapy, Psychiatric University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University of Zurich and Eidgenössische Technische Hochschule (ETH) Zurich, Zurich, Switzerland
| | - Sergi Papiol
- Max Planck Institute of Psychiatry, Munich, Germany
- Institute of Psychiatric Phenomics and Genomics, Ludwig Maximilian University Munich, Munich, Germany
| | - Florian J. Raabe
- Max Planck Institute of Psychiatry, Munich, Germany
- Department of Psychiatry and Psychotherapy, Ludwig Maximilian University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| |
Collapse
|
14
|
Magni G, Riboldi B, Marinelli A, Uboldi P, Bonacina F, Di Lorenzo C, Petroni K, Ceruti S. Prevention of motor relapses and associated trigeminal pain in experimental autoimmune encephalomyelitis by reducing neuroinflammation with a purple corn extract enriched in anthocyanins. Biomed Pharmacother 2025; 184:117906. [PMID: 39955855 DOI: 10.1016/j.biopha.2025.117906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025] Open
Abstract
Multiple sclerosis (MS) is the leading cause of disability in young adults, with about 2.5-3 million cases currently diagnosed. Pain is a common comorbid symptom of MS, and develops independently from motor impairments. Currently utilized drugs bear severe side effects; thus, new therapeutic strategies are needed, and nutraceutical supplements represent innovative and safe opportunities. We have selected a variety of anthocyanin-enriched purple corn, from which a water-soluble extract (Red extract) has been obtained and administered to male rats exposed to experimental autoimmune encephalomyelitis (EAE). Animals developed relapsing-remitting motor symptoms, accompanied by early onset trigeminal allodynia. The preventive administration of Red extract facilitated the remission of motor symptoms, prevented the development of relapses, and delayed and reduced the development of EAE-associated trigeminal pain. An overall inhibition of neuroinflammation, blunted microgliosis and astrogliosis, activation of autophagy and reduced immune cell infiltration in the brainstem, cervical and lumbar spinal cord were observed. Yellow corn extract, lacking anthocyanins, had no behavioral effects, despite a limited anti-inflammatory action. Therapeutic Red extract administration did not affect EAE motor symptoms, only partially reduced the development of trigeminal pain but maintained its ability to reduce neuroinflammation and glial cell activation and to promote autophagy. Overall, our data suggest that a nutraceutical supplement from anthocyanin-enriched purple corn represents an interesting option to limit the development of motor relapses and the chronicization of multiple sclerosis-associated pain, through the mitigation of neuroinflammation, of the infiltration of immune cells and the promotion of autophagy.
Collapse
Affiliation(s)
- Giulia Magni
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti" - Università degli Studi di Milano, Via Balzaretti, 9, Milan 20133, Italy.
| | - Benedetta Riboldi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti" - Università degli Studi di Milano, Via Balzaretti, 9, Milan 20133, Italy.
| | - Alessandra Marinelli
- Department of Biosciences - Università degli Studi di Milano, Via Celoria, 26, Milan 20133, Italy.
| | - Patrizia Uboldi
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti" - Università degli Studi di Milano, Via Balzaretti, 9, Milan 20133, Italy.
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti" - Università degli Studi di Milano, Via Balzaretti, 9, Milan 20133, Italy.
| | - Chiara Di Lorenzo
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti" - Università degli Studi di Milano, Via Balzaretti, 9, Milan 20133, Italy.
| | - Katia Petroni
- Department of Biosciences - Università degli Studi di Milano, Via Celoria, 26, Milan 20133, Italy.
| | - Stefania Ceruti
- Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti" - Università degli Studi di Milano, Via Balzaretti, 9, Milan 20133, Italy.
| |
Collapse
|
15
|
Fabian C, Mahajan S, Schmidt MHH. EGFL7: An emerging biomarker with great therapeutic potential. Pharmacol Ther 2025; 266:108764. [PMID: 39631508 DOI: 10.1016/j.pharmthera.2024.108764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 11/08/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024]
Abstract
EGFL7 is a factor involved in the regulation of various essential biological mechanisms. Endothelial cells and neurons secrete the EGFL7 protein into the extracellular matrix, where it interacts with other matrix proteins, thereby regulating several important signaling pathways. To date, extensive in vitro and in vivo studies have illuminated the central role of EGFL7 in governing major biological processes involving blood vessels and the central nervous system. Notably, EGFL7 has also emerged as a key factor in a spectrum of diseases including cancer, stroke, multiple sclerosis and preeclampsia. Its influence on various diseases and multiple regulatory pathways highlights EGFL7 as an emerging biomarker and therapeutic target. Thus, the multifaceted regulatory functions of EGFL7 will be discussed in the physiological context before delving into its involvement in the progression of different diseases. Finally, the review will provide an insight into the broad therapeutic potential of EGFL7 by describing its role as a powerful biomarker and discussing potential strategies to therapeutically target EGFL7 function in a plethora of human diseases.
Collapse
Affiliation(s)
- Carina Fabian
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technical University Dresden School of Medicine, Fetscherstraße 74, 01307 Dresden, Germany
| | - Sukrit Mahajan
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technical University Dresden School of Medicine, Fetscherstraße 74, 01307 Dresden, Germany
| | - Mirko H H Schmidt
- Institute of Anatomy, Medical Faculty Carl Gustav Carus, Technical University Dresden School of Medicine, Fetscherstraße 74, 01307 Dresden, Germany.
| |
Collapse
|
16
|
van Hijfte L, Geurts M, de Heer I, Ghisai SA, Balcioglu HE, Hoogstrate Y, Vallentgoed WR, Head R, Luning R, van den Bosch T, Westerman B, Wesseling P, Joyce JA, French P, Debets R. Gemistocytic tumor cells programmed for glial scarring characterize T cell confinement in IDH-mutant astrocytoma. Nat Commun 2025; 16:1156. [PMID: 39880824 PMCID: PMC11779865 DOI: 10.1038/s41467-025-56441-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025] Open
Abstract
Isocitrate dehydrogenase 1/2 mutant (IDHmt) astrocytoma is considered a T cell-deprived tumor, yet little is known regarding the phenotypes underlying T cell exclusion. Using bulk, single nucleus and spatial RNA and protein profiling, we demonstrate that a distinct spatial organization underlies T cell confinement to the perivascular space (T cell cuff) in IDHmt astrocytoma. T cell cuffs are uniquely characterized by a high abundance of gemistocytic tumor cells (GTC) in the surrounding stroma. Integrative analysis shows that GTC-high tumors are enriched for lymphocytes and tumor associated macrophages (TAM) and express immune cell migration and activation programs. Specifically, GTCs constitute a distinct sub-cluster of the astrocyte-like tumor cell state that co-localizes with immune reactive TAMs. Neighboring GTCs and TAMs express receptor-ligand pairs characteristic of reactive astrogliosis and glial scarring, such as SPP1/CD44 and IL-1β/IL1R1. Collectively, we reveal that T cell confinement in IDHmt astrocytomas associates with GTC-TAM networks that mimic glial scarring mechanisms.
Collapse
Affiliation(s)
- Levi van Hijfte
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
- Department of Medical Oncology, Laboratory of Tumor Immunology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| | - Marjolein Geurts
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
- Department of Medical Oncology, Laboratory of Tumor Immunology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Iris de Heer
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Santoesha A Ghisai
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Hayri E Balcioglu
- Department of Medical Oncology, Laboratory of Tumor Immunology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Youri Hoogstrate
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Wies R Vallentgoed
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Rania Head
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - Rosa Luning
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - Bart Westerman
- Department of Neurosurgery, Amsterdam UMC/VUMC, Amsterdam, The Netherlands
| | - Pieter Wesseling
- Department of Pathology, Amsterdam UMC/VUMC and Brain Tumour Center, Amsterdam, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Johanna A Joyce
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, Lausanne, Switzerland
- Agora Cancer Center Lausanne and Swiss Cancer Center Léman, Lausanne, Switzerland
| | - Pim French
- Department of Neurology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| | - Reno Debets
- Department of Medical Oncology, Laboratory of Tumor Immunology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands.
| |
Collapse
|
17
|
Liu X, Yang L, Su Z, Ma X, Liu Y, Ma L, Ma X, Ma M, Liu X, Zhang K, Chen X. Acupoint catgut embedding alleviates experimental autoimmune encephalomyelitis by modulating neuroinflammation and potentially inhibiting glia activation through JNK and ERK pathways. Front Neurosci 2025; 18:1520092. [PMID: 39850625 PMCID: PMC11755674 DOI: 10.3389/fnins.2024.1520092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 12/06/2024] [Indexed: 01/25/2025] Open
Abstract
Background Acupoint catgut embedding (ACE) is a traditional Chinese medicine technique commonly used for managing various disorders, including chronic inflammatory pain and allergic asthma. Despite its growing use, the neuroimmunological mechanisms underlying ACE treatment effects remain unclear. Methods This study investigated the roles and potential mechanisms of the effects of ACE in treating experimental autoimmune encephalomyelitis (EAE), a frequently used animal model of autoimmune neuroinflammation. The effects of ACE treatment were evaluated by monitoring body weight and EAE severity scores. Behavioral tests, histopathological analysis, ELISA, and flow cytometry were conducted to assess the therapeutic efficacy of ACE. RNA sequencing was performed to uncover ACE-associated transcriptional signatures in the spinal cords of EAE mice. Results The results were validated through western blotting, qRT-PCR, and immunofluorescence (IF) staining. In ACE-treated mice, EAE disease severity was significantly ameliorated, along with improvements in anxiety-like behaviors and reduced inflammation and demyelination. The ACE treatment restored immune imbalance in the EAE mice by decreasing Th17 and Th1 cells, while increasing Treg cells in peripheral immune organs and reducing serum inflammatory cytokine levels. RNA sequencing revealed significant suppression of the genes and pathways associated with reactive microglial and astrocytic activation, corroborated by IF studies. Additionally, ACE treatment could suppress the ERK and JNK signaling pathways at both RNA and protein levels. Conclusion These findings confirm the protective role of ACE in mitigating EAE symptoms by modulating microglial and astrocytic activity and regulating inflammatory cytokines.
Collapse
Affiliation(s)
- Xiaofang Liu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Liansheng Yang
- Department of Acupuncture and Moxibustion, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhumin Su
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xueying Ma
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Yingying Liu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Lili Ma
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaomeng Ma
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Mingxia Ma
- Department of Cardiology, Shanxi Province Cardiovascular Hospital, Taiyuan, China
| | - Xiaoyun Liu
- Department of General Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Kun Zhang
- Department of Acupuncture and Moxibustion, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Department of Allergy, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xiaohong Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
18
|
Vella VR, Holman PJ, Bodnar TS, Raineki C. Ontogenetic Neuroimmune Changes Following Prenatal Alcohol Exposure: Implications for Neurobehavioral Function. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1473:15-39. [PMID: 40128473 DOI: 10.1007/978-3-031-81908-7_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 03/26/2025]
Abstract
This chapter reviews the enduring effects of prenatal alcohol exposure (PAE) on neuroimmune function across the lifespan, including discussion of associated neurobehavioral alterations. Alcohol has potent teratogenic effects, with a large body of work linking PAE to perturbations in neuroimmune function. These PAE-related neuroimmune disturbances may have downstream effects on neurobehavioral function given the critical role of the neuroimmune system in central nervous system development. The neuroimmune system matures over time, playing distinct roles depending on the developmental processes occurring within that maturational stage. This chapter thus takes an ontogenetic approach to understanding how PAE induces unique neuroimmune changes across the lifespan, beginning with a review of changes in early life before moving into adolescence and ending in adulthood. The focus will be on work utilizing rodent models, which allow for more tightly controlled conditions than are possible in human research. The chapter concludes with a discussion of possible mechanisms underlying the developmental changes in neuroimmune function following PAE, with a specific focus on the role of the gut microbiota.
Collapse
Affiliation(s)
- Victoria R Vella
- Department of Psychology, Brock University, St. Catharines, ON, Canada
| | - Parker J Holman
- Department of Psychology, Brock University, St. Catharines, ON, Canada
| | - Tamara S Bodnar
- Department of Biological Sciences, University of Calgary, Calgary, AB, Canada
- Alberta Children's Hospital Research Institute, Calgary, AB, Canada
- Hotchkiss Brain Institute, Calgary, AB, Canada
| | - Charlis Raineki
- Department of Psychology, Brock University, St. Catharines, ON, Canada.
- Centre for Neuroscience, Brock University, St. Catharines, ON, Canada.
| |
Collapse
|
19
|
Desu HL, Thougaard E, Carney BN, Illiano P, Plastini MJ, Florimon Y, Mini A, Guastucci C, Kang B, Lee JK, Lambertsen KL, Brambilla R. TNFR2 signaling in oligodendrocyte precursor cells suppresses their immune-inflammatory function and detrimental microglia activation in CNS demyelinating disease. Brain Behav Immun 2025; 123:81-98. [PMID: 39243989 PMCID: PMC11624083 DOI: 10.1016/j.bbi.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 08/31/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
Multiple Sclerosis (MS) is a chronic degenerative disease of the central nervous system (CNS) characterized by inflammation, demyelination, and progressive neurodegeneration. These processes, combined with the failure of reparative remyelination initiated by oligodendrocyte precursor cells (OPCs), lead to irreversible neurological impairment. The cytokine tumor necrosis factor (TNF) has been implicated in CNS repair via activation of its cognate receptor TNFR2 in glia. Here, we demonstrate the important role of TNFR2 in regulating OPC function in vivo during demyelinating disease, and that TNFR2 expressed in OPCs modulates OPC-microglia interactions. In PdgfrαCreERT:Tnfrsf1bfl/fl:Eyfp mice with selective TNFR2 ablation in OPCs, we observed an earlier onset and disease peak in experimental autoimmune encephalomyelitis (EAE). This was associated with accelerated immune cell infiltration and increased microglia activation in the spinal cord. Similarly, PdgfrαCreERT:Tnfrsf1bfl/fl:Eyfp mice showed rapid and increased microglia reactivity compared to control mice in the corpus callosum after cuprizone-induced demyelination, followed by chronic reduction in the number of mature myelinating oligodendrocytes (OLs). With EAE and cuprizone models combined, we uncovered that TNFR2 does not have a cell autonomous role in OPC differentiation, but may be important for survival of newly formed mature OLs. Finally, using an in vitro approach, we demonstrated that factors released by Tnfrsf1b ablated OPCs drove microglia to develop an exacerbated "foamy" phenotype when incubated with myelin-rich spinal cord homogenate, aberrantly increasing lysosomal lipid accumulation. Together, our data indicate that TNFR2 signaling in OPCs is protective by dampening their immune-inflammatory activation and by suppressing neurotoxic microglia reactivity. This suggests that boosting TNFR2 activation or its downstream cascades could be an effective strategy to restore OPC reparative capacity in neuroimmune and demyelinating disease.
Collapse
MESH Headings
- Oligodendrocyte Precursor Cells/immunology
- Oligodendrocyte Precursor Cells/metabolism
- Animals
- Mice
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Mice, Transgenic
- Multiple Sclerosis/chemically induced
- Multiple Sclerosis/genetics
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Microglia/immunology
- Microglia/metabolism
- Microglia/pathology
- Signal Transduction/genetics
- Signal Transduction/immunology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Cell Survival/genetics
- Cell Survival/immunology
- Cuprizone/toxicity
- Cell Differentiation/genetics
- Cell Differentiation/immunology
- Mice, Inbred C57BL
- Male
- Female
- Corpus Callosum/cytology
- Corpus Callosum/immunology
- Corpus Callosum/pathology
Collapse
Affiliation(s)
- Haritha L Desu
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Estrid Thougaard
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark
| | - Brianna N Carney
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Placido Illiano
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Melanie J Plastini
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yoleinny Florimon
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Antonella Mini
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Chelsea Guastucci
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Brian Kang
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Jae K Lee
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Kate L Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark; BRIDGE-Brain Research Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark; Department of Neurology, Odense University Hospital, 5000 Odense C, Denmark
| | - Roberta Brambilla
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA; Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark; BRIDGE-Brain Research Inter Disciplinary Guided Excellence, Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark.
| |
Collapse
|
20
|
Wu ZY, Luo YF, Lu YP, Zhang YX, Han F. Targeting Brain Endothelial Gasdermin D: A Shortcut to Remodel the Blood-Brain Barrier. Neurosci Bull 2025; 41:189-191. [PMID: 39276179 PMCID: PMC11748715 DOI: 10.1007/s12264-024-01300-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 07/20/2024] [Indexed: 09/16/2024] Open
Affiliation(s)
- Zhou-Yue Wu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China
- International Joint Laboratory for Drug Targets of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Yi-Fan Luo
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China
- International Joint Laboratory for Drug Targets of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Ya-Ping Lu
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China
- International Joint Laboratory for Drug Targets of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
- Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China
| | - Yi-Xuan Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China
- International Joint Laboratory for Drug Targets of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Nanjing, 211166, China.
- International Joint Laboratory for Drug Targets of Critical Illnesses, School of Pharmacy, Nanjing Medical University, Nanjing, 211166, China.
- Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, 215002, China.
| |
Collapse
|
21
|
Bravo-San Pedro JM, Aranda F, Buqué A, Galluzzi L. Animal models of disease: Achievements and challenges. Methods Cell Biol 2025; 192:xv-xxi. [PMID: 39863396 DOI: 10.1016/s0091-679x(25)00026-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
Affiliation(s)
- José Manuel Bravo-San Pedro
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Aitziber Buqué
- Fox Chase Cancer Center, Philadelphia, PA, United States.
| | | |
Collapse
|
22
|
Wang X, Hu J, Xie S, Li W, Zhang H, Huang L, Qian Z, Zhao C, Zhang L. Hidden role of microglia during neurodegenerative disorders and neurocritical care: A mitochondrial perspective. Int Immunopharmacol 2024; 142:113024. [PMID: 39217875 DOI: 10.1016/j.intimp.2024.113024] [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: 05/07/2024] [Revised: 08/04/2024] [Accepted: 08/23/2024] [Indexed: 09/04/2024]
Abstract
The incidence of aging-related neurodegenerative disorders and neurocritical care diseases is increasing worldwide. Microglia, the main inflammatory cells in the brain, could be potential viable therapeutic targets for treating neurological diseases. Interestingly, mitochondrial functions, including energy metabolism, mitophagy and transfer, fission and fusion, and mitochondrial DNA expression, also change in activated microglia. Notably, mitochondria play an active and important role in the pathophysiology of neurodegenerative disorders and neurocritical care diseases. This review briefly summarizes the current knowledge on mitochondrial dysfunction in microglia in neurodegenerative disorders and neurocritical care diseases and comprehensively discusses the prospects of the application of neurological injury prevention and treatment targets by mitochondria.
Collapse
Affiliation(s)
- Xinrun Wang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Jiyun Hu
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Shucai Xie
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Wenchao Li
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Haisong Zhang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Li Huang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Zhaoxin Qian
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China
| | - Chunguang Zhao
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.
| | - Lina Zhang
- Department of Critical Care Medicine, Hunan Provincial Clinical Research Center for Critical Care Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, PR China.
| |
Collapse
|
23
|
Zhu H, Hu E, Guo X, Yuan Z, Jiang H, Zhang W, Tang T, Wang Y, Li T. Promoting remyelination in central nervous system diseases: Potentials and prospects of natural products and herbal medicine. Pharmacol Res 2024; 210:107533. [PMID: 39617281 DOI: 10.1016/j.phrs.2024.107533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024]
Abstract
Myelin damage is frequently associated with central nervous system (CNS) diseases and is a critical factor influencing neurological function and disease prognosis. Nevertheless, the majority of current treatments for the CNS concentrate on gray matter injury and repair strategies, while clinical interventions specifically targeting myelin repair remain unavailable. In recent years, natural products and herbal medicine have achieved considerable progress in the domain of myelin repair, given their remarkable curative effect and low toxic side effects, demonstrating significant therapeutic potential. In this review, we present a rather comprehensive account of the mechanisms underlying myelin formation, injury, and repair, with a particular emphasis on the interactions between oligodendrocytes and other glial cells. Furthermore, we summarize the natural products and herbal medicine currently employed in remyelination along with their mechanisms of action, highlighting the potential and challenges of certain natural compounds to enhance myelin repair. This review aims to facilitate the expedited development of innovative therapeutics derived from natural products and herbal medicine and furnish novel insights into myelin repair in the CNS.
Collapse
Affiliation(s)
- Haonan Zhu
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - En Hu
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Xin Guo
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhiqiang Yuan
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Haoying Jiang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Tao Tang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Yang Wang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Teng Li
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China.
| |
Collapse
|
24
|
Kadam R, Gupta M, Lazarov O, Prabhakar BS. Brain-immune interactions: implication for cognitive impairments in Alzheimer's disease and autoimmune disorders. J Leukoc Biol 2024; 116:1269-1290. [PMID: 38869088 DOI: 10.1093/jleuko/qiae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 05/09/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024] Open
Abstract
Progressive memory loss and cognitive dysfunction, encompassing deficits in learning, memory, problem solving, spatial reasoning, and verbal expression, are characteristics of Alzheimer's disease and related dementia. A wealth of studies has described multiple roles of the immune system in the development or exacerbation of dementia. Individuals with autoimmune disorders can also develop cognitive dysfunction, a phenomenon termed "autoimmune dementia." Together, these findings underscore the pivotal role of the neuroimmune axis in both Alzheimer's disease and related dementia and autoimmune dementia. The dynamic interplay between adaptive and innate immunity, both in and outside the brain, significantly affects the etiology and progression of these conditions. Multidisciplinary research shows that cognitive dysfunction arises from a bidirectional relationship between the nervous and immune systems, though the specific mechanisms that drive cognitive impairments are not fully understood. Intriguingly, this reciprocal regulation occurs at multiple levels, where neuronal signals can modulate immune responses, and immune system-related processes can influence neuronal viability and function. In this review, we consider the implications of autoimmune responses in various autoimmune disorders and Alzheimer's disease and explore their effects on brain function. We also discuss the diverse cellular and molecular crosstalk between the brain and the immune system, as they may shed light on potential triggers of peripheral inflammation, their effect on the integrity of the blood-brain barrier, and brain function. Additionally, we assess challenges and possibilities associated with developing immune-based therapies for the treatment of cognitive decline.
Collapse
Affiliation(s)
- Rashmi Kadam
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| | - Muskan Gupta
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Orly Lazarov
- Department of Anatomy and Cell Biology, University of Illinois College of Medicine, 808 S Wood street, MC 512, Chicago, Chicago, IL 60612, United States
| | - Bellur S Prabhakar
- Department of Microbiology and Immunology, University of Illinois College of Medicine, 835 S Wolcott street, MC 790, Chicago, Chicago, IL 60612, United States
| |
Collapse
|
25
|
Langlois J, Lange S, Ebeling M, Macnair W, Schmucki R, Li C, DeGeer J, Sudharshan TJJ, Yong VW, Shen YA, Harp C, Collin L, Keaney J. Fenebrutinib, a Bruton's tyrosine kinase inhibitor, blocks distinct human microglial signaling pathways. J Neuroinflammation 2024; 21:276. [PMID: 39465429 PMCID: PMC11514909 DOI: 10.1186/s12974-024-03267-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Bruton's tyrosine kinase (BTK) is an intracellular signaling enzyme that regulates B-lymphocyte and myeloid cell functions. Due to its involvement in both innate and adaptive immune compartments, BTK inhibitors have emerged as a therapeutic option in autoimmune disorders such as multiple sclerosis (MS). Brain-penetrant, small-molecule BTK inhibitors may also address compartmentalized neuroinflammation, which is proposed to underlie MS disease progression. BTK is expressed by microglia, which are the resident innate immune cells of the brain; however, the precise roles of microglial BTK and impact of BTK inhibitors on microglial functions are still being elucidated. Research on the effects of BTK inhibitors has been limited to rodent disease models. This is the first study reporting effects in human microglia. METHODS Here we characterize the pharmacological and functional properties of fenebrutinib, a potent, highly selective, noncovalent, reversible, brain-penetrant BTK inhibitor, in human microglia and complex human brain cell systems, including brain organoids. RESULTS We find that fenebrutinib blocks the deleterious effects of microglial Fc gamma receptor (FcγR) activation, including cytokine and chemokine release, microglial clustering and neurite damage in diverse human brain cell systems. Gene expression analyses identified pathways linked to inflammation, matrix metalloproteinase production and cholesterol metabolism that were modulated by fenebrutinib treatment. In contrast, fenebrutinib had no significant impact on human microglial pathways linked to Toll-like receptor 4 (TLR4) and NACHT, LRR and PYD domains-containing protein 3 (NLRP3) signaling or myelin phagocytosis. CONCLUSIONS Our study enhances the understanding of BTK functions in human microglial signaling that are relevant to MS pathogenesis and suggests that fenebrutinib could attenuate detrimental microglial activity associated with FcγR activation in people with MS.
Collapse
Affiliation(s)
- Julie Langlois
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Simona Lange
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Martin Ebeling
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Will Macnair
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Roland Schmucki
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Cenxiao Li
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, Canada
| | - Jonathan DeGeer
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - Tania J J Sudharshan
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - V Wee Yong
- Hotchkiss Brain Institute and the Department of Clinical Neurosciences, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, Canada
| | - Yun-An Shen
- Genentech, Inc., 1 DNA Way, South San Francisco, CA, USA
| | | | - Ludovic Collin
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland
| | - James Keaney
- Roche Pharma Research and Early Development, Neuroscience and Rare Diseases Discovery and Translational Area, Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| |
Collapse
|
26
|
Duan J, Lv A, Guo Z, Liu Q, Tian C, Yang Y, Bi J, Yu X, Peng G, Luo B, Cai Z, Xu B, Fu Y, Zhang J. CX3CR1 +/UCHL1 + microglial extracellular vesicles in blood: a potential biomarker for multiple sclerosis. J Neuroinflammation 2024; 21:254. [PMID: 39385200 PMCID: PMC11465848 DOI: 10.1186/s12974-024-03243-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/23/2024] [Indexed: 10/12/2024] Open
Abstract
In neuroinflammation, distinguishing microglia from macrophages and identifying microglial-specific biomarkers in peripheral blood pose significant challenges. This study comprehensively profiled the extracellular vesicles (EVs) of microglia and macrophages, respectively, revealing co-expressed EVs with UCHL1 and CX3CR1 as EVs derived specifically from microglia in human blood. After extensive validation, using optimized nano flow cytometry, we evaluated plasma CX3CR1+/UCHL1+ EVs across clinical cohorts [multiple sclerosis (MS), HTLV-1 associated myelopathy (HAM), Alzheimer's disease (AD), and Parkinson's disease (PD)], along with established neurodegenerative markers (NMDAR2A and NFL). The findings discovered a notable rise in CX3CR1+/UCHL1+ EVs in MS, particularly heightened in HAM, in contrast to controls. Conversely, AD and PD exhibited unaltered or diminished levels of microglial EVs. An integrated model of CX3CR1+/UCHL1+, NMDAR2A+, and NFL+ EVs demonstrated promising diagnostic potential for distinguishing MS from controls and HAM. As to the disease duration, CX3CR1+/UCHL1+ EVs increased in the initial five years of MS, stabilizing thereafter, whereas NMDAR2A+ and NFL+ EVs remained stable initially but increased significantly in the subsequent five years, suggesting their correlation with disease duration. This study uncovers unique blood microglial EVs with potential as biomarkers for MS diagnosis, differentiation from HAM, and correlation with disease duration.
Collapse
Affiliation(s)
- Jing Duan
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Aowei Lv
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhen Guo
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qi Liu
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Chen Tian
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Ying Yang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Jin Bi
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Xintong Yu
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China
| | - Guoping Peng
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Benyan Luo
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhijian Cai
- Institute of Immunology, Department of Orthopedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Bin Xu
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Ying Fu
- Department of Neurology, Institute of Neurology of First Affiliated Hospital, Fujian Medical University, Fuzhou, Fujian, China.
| | - Jing Zhang
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
- National Human Brain Bank for Health and Disease, Zhejiang University, Hangzhou, 310012, China.
| |
Collapse
|
27
|
Belousova O, Lopatina A, Melnikov M. The role of dopamine in the modulation of monocyte-induced Th17- and Th1-immune response in multiple sclerosis. Int Immunopharmacol 2024; 137:112540. [PMID: 38908080 DOI: 10.1016/j.intimp.2024.112540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 06/08/2024] [Accepted: 06/19/2024] [Indexed: 06/24/2024]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system (CNS) with autoimmune mechanism of development. The investigation of neuroimmune interaction is one of the most developing directions in MS pathogenesis study. Catecholamines are direct mediators of this interaction and can be involved in the pathogenesis of MS by modulating cells of both innate and adaptive immune systems. The aim of this study was to investigate the influence of dopamine and norepinephrine on the ability of monocytes of patients with relapsing-remitting MS, to induce Th17- and Th1-immune response, which play a crucial role in the autoimmunity of the CNS. We found, that both dopamine and norepinephrine modulate the production of Th17- (IL-23, IL-1β, and IL-6) and Th1-promoting (IL-12p70) cytokines by activated peripheral blood mononuclear cells or CD14+ monocytes in patients with MS and in healthy subjects. We also found the inhibitory effect of dopamine and norepinephrine on monocyte-induced production of IL-17 and IFN-γ by autologous CD4+ T-cells in both groups. Finally, the multidirectional role of D1- and D2-like dopaminergic receptors in the modulatory effect of dopamine on the ability of CD14+ monocytes to activate CD4+ T-cells was established, expanding the potential role of dopamine in the neuroimmune interaction.
Collapse
Affiliation(s)
- Olga Belousova
- Laboratory of Neuroimmunology, Federal Center of Brain Research and Neurotechnologies of the Federal Medical-Biological Agency of Russia, 117513, Moscow, Russia
| | - Anna Lopatina
- Laboratory of Neuroimmunology, Federal Center of Brain Research and Neurotechnologies of the Federal Medical-Biological Agency of Russia, 117513, Moscow, Russia
| | - Mikhail Melnikov
- Laboratory of Neuroimmunology, Federal Center of Brain Research and Neurotechnologies of the Federal Medical-Biological Agency of Russia, 117513, Moscow, Russia; Department of Neurology, Neurosurgery and Medical Genetics of Pirogov Russian National Research Medical University, 117997, Moscow, Russia; Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency, 115478, Moscow, Russia.
| |
Collapse
|
28
|
Ma H, Zhu M, Chen M, Li X, Feng X. The role of macrophage plasticity in neurodegenerative diseases. Biomark Res 2024; 12:81. [PMID: 39135084 PMCID: PMC11321226 DOI: 10.1186/s40364-024-00624-7] [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: 04/28/2024] [Accepted: 07/22/2024] [Indexed: 08/15/2024] Open
Abstract
Tissue-resident macrophages and recruited macrophages play pivotal roles in innate immunity and the maintenance of brain homeostasis. Investigating the involvement of these macrophage populations in eliciting pathological changes associated with neurodegenerative diseases has been a focal point of research. Dysregulated states of macrophages can compromise clearance mechanisms for pathological proteins such as amyloid-β (Aβ) in Alzheimer's disease (AD) and TDP-43 in Amyotrophic lateral sclerosis (ALS). Additionally, recent evidence suggests that abnormalities in the peripheral clearance of pathological proteins are implicated in the pathogenesis and progression of neurodegenerative diseases. Furthermore, numerous genome-wide association studies have linked genetic risk factors, which alter the functionality of various immune cells, to the accumulation of pathological proteins. This review aims to unravel the intricacies of macrophage biology in both homeostatic conditions and neurodegenerative disorders. To this end, we initially provide an overview of the modifications in receptor and gene expression observed in diverse macrophage subsets throughout development. Subsequently, we outlined the roles of resident macrophages and recruited macrophages in neurodegenerative diseases and the progress of targeted therapy. Finally, we describe the latest advances in macrophage imaging methods and measurement of inflammation, which may provide information and related treatment strategies that hold promise for informing the design of future investigations and therapeutic interventions.
Collapse
Affiliation(s)
- Hongyue Ma
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Mingxia Zhu
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Mengjie Chen
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Xiuli Li
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China
| | - Xinhong Feng
- Department of Neurology, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China.
| |
Collapse
|
29
|
Qi C, Ren H, Fan Y. Microglia specific alternative splicing alterations in multiple sclerosis. Aging (Albany NY) 2024; 16:11656-11667. [PMID: 39115871 PMCID: PMC11346782 DOI: 10.18632/aging.206045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024]
Abstract
Several aberrant alternative splicing (AS) events and their regulatory mechanisms are widely recognized in multiple sclerosis (MS). Yet the cell-type specific AS events have not been extensively examined. Here we assessed the diversity of AS events using web-based RNA-seq data of sorted CD15-CD11b+ microglia in white matter (WM) region from 10 patients with MS and 11 control subjects. The GSE111972 dataset was downloaded from GEO and ENA databases, aligned to the GRCh38 reference genome from ENSEMBL via STAR. rMATS was used to assess five types of AS events, alternative 3'SS (A3SS), alternative 5'SS (A5SS), skipped exon (SE), retained intron (RI) and mutually exclusive exons (MXE), followed by visualizing with rmats2sashimiplot and maser. Differential genes or transcripts were analyzed using the limma R package. Gene ontology (GO) analysis was performed with the clusterProfiler R package. 42,663 raw counts of AS events were identified and 132 significant AS events were retained based on the filtered criteria: 1) average coverage >10 and 2) delta percent spliced in (ΔPSI) >0.1. SE was the most common AS event (36.36%), followed by MXE events (32.58%), and RI (18.94%). Genes related to telomere maintenance and organization primarily underwent SE splicing, while genes associated with protein folding and mitochondrion organization were predominantly spliced in the MXE pattern. Conversely, genes experiencing RI were enriched in immune response and immunoglobulin production. In conclusion, we identified microglia-specific AS changes in the white matter of MS patients, which may shed light on novel pathological mechanisms underlying MS.
Collapse
Affiliation(s)
- Caiyun Qi
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Honglei Ren
- Department of Neurology, Tianjin Neurological Institute, Tianjin Institute of Immunology, State Key Laboratory of Experimental Hematology, Haihe Laboratory of Cell Ecosystem, Tianjin Medical University General Hospital, Tianjin, China
| | - Yong Fan
- Department of Obstetrics and Gynecology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, Guangdong-Hong Kong-Macao Greater Bay Area Higher Education Joint Laboratory of Maternal-Fetal Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
30
|
Keller CW, Mundt S, Segal BM. Editorial: CNS myeloid cell function in health and disease. Front Immunol 2024; 15:1459138. [PMID: 39086488 PMCID: PMC11288962 DOI: 10.3389/fimmu.2024.1459138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 07/09/2024] [Indexed: 08/02/2024] Open
Affiliation(s)
- Christian W. Keller
- Department of Neurology with Institute of Translational Neurology, University Hospital Münster, Münster, Germany
| | - Sarah Mundt
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Benjamin M. Segal
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, OH, United States
- The Neuroscience Research Institute, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
31
|
Kim M, Choi H, Jang DJ, Kim HJ, Sub Y, Gee HY, Choi C. Exploring the clinical transition of engineered exosomes designed for intracellular delivery of therapeutic proteins. Stem Cells Transl Med 2024; 13:637-647. [PMID: 38838263 PMCID: PMC11227971 DOI: 10.1093/stcltm/szae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 03/18/2024] [Indexed: 06/07/2024] Open
Abstract
Extracellular vesicles, particularly exosomes, have emerged as promising drug delivery systems owing to their unique advantages, such as biocompatibility, immune tolerability, and target specificity. Various engineering strategies have been implemented to harness these innate qualities, with a focus on enhancing the pharmacokinetic and pharmacodynamic properties of exosomes via payload loading and surface engineering for active targeting. This concise review outlines the challenges in the development of exosomes as drug carriers and offers insights into strategies for their effective clinical translation. We also highlight preclinical studies that have successfully employed anti-inflammatory exosomes and suggest future directions for exosome therapeutics. These advancements underscore the potential for integrating exosome-based therapies into clinical practice, heralding promise for future medical interventions.
Collapse
Affiliation(s)
| | - Hojun Choi
- ILIAS Biologics Inc., Daejeon 34014, Korea
| | - Deok-Jin Jang
- ILIAS Biologics Inc., Daejeon 34014, Korea
- Department of Ecological Science, College of Ecology and Environment, Kyungpook National University, Sangju 37224, Korea
| | | | - Yujin Sub
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Heon Yung Gee
- Department of Pharmacology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | | |
Collapse
|
32
|
Perussolo MC, Mogharbel BF, Saçaki CS, da Rosa NN, Irioda AC, de Oliveira NB, Appel JM, Lührs L, Meira LF, Guarita-Souza LC, Nagashima S, de Paula CBV, de Noronha L, Zotarelli-Filho IJ, Abdelwahid E, de Carvalho KAT. Cellular Therapy in Experimental Autoimmune Encephalomyelitis as an Adjuvant Treatment to Translate for Multiple Sclerosis. Int J Mol Sci 2024; 25:6996. [PMID: 39000105 PMCID: PMC11241124 DOI: 10.3390/ijms25136996] [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: 04/29/2024] [Revised: 06/11/2024] [Accepted: 06/20/2024] [Indexed: 07/16/2024] Open
Abstract
This study aims to evaluate and compare cellular therapy with human Wharton's jelly (WJ) mesenchymal stem cells (MSCs) and neural precursors (NPs) in experimental autoimmune encephalomyelitis (EAE), a preclinical model of Multiple Sclerosis. MSCs were isolated from WJ by an explant technique, differentiated to NPs, and characterized by cytometry and immunocytochemistry analysis after ethical approval. Forty-eight rats were EAE-induced by myelin basic protein and Freund's complete adjuvant. Forty-eight hours later, the animals received intraperitoneal injections of 250 ng/dose of Bordetella pertussis toxin. Fourteen days later, the animals were divided into the following groups: a. non-induced, induced: b. Sham, c. WJ-MSCs, d. NPs, and e. WJ-MSCs plus NPs. 1 × 105. Moreover, the cells were placed in a 10 µL solution and injected via a stereotaxic intracerebral ventricular injection. After ten days, the histopathological analysis for H&E, Luxol, interleukins, and CD4/CD8 was carried out. Statistical analyses demonstrated a higher frequency of clinical manifestation in the Sham group (15.66%) than in the other groups; less demyelination was seen in the treated groups than the Sham group (WJ-MSCs, p = 0.016; NPs, p = 0.010; WJ-MSCs + NPs, p = 0.000), and a lower cellular death rate was seen in the treated groups compared with the Sham group. A CD4/CD8 ratio of <1 showed no association with microglial activation (p = 0.366), astrocytes (p = 0.247), and cell death (p = 0.577) in WJ-MSCs. WJ-MSCs and NPs were immunomodulatory and neuroprotective in cellular therapy, which would be translated as an adjunct in demyelinating diseases.
Collapse
Affiliation(s)
- Maiara Carolina Perussolo
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba P.O. Box 80240-020, Paraná, Brazil; (M.C.P.); (B.F.M.); (C.S.S.); (N.N.d.R.); (A.C.I.); (N.B.d.O.); (J.M.A.); (L.L.)
| | - Bassam Felipe Mogharbel
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba P.O. Box 80240-020, Paraná, Brazil; (M.C.P.); (B.F.M.); (C.S.S.); (N.N.d.R.); (A.C.I.); (N.B.d.O.); (J.M.A.); (L.L.)
| | - Cláudia Sayuri Saçaki
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba P.O. Box 80240-020, Paraná, Brazil; (M.C.P.); (B.F.M.); (C.S.S.); (N.N.d.R.); (A.C.I.); (N.B.d.O.); (J.M.A.); (L.L.)
| | - Nádia Nascimento da Rosa
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba P.O. Box 80240-020, Paraná, Brazil; (M.C.P.); (B.F.M.); (C.S.S.); (N.N.d.R.); (A.C.I.); (N.B.d.O.); (J.M.A.); (L.L.)
| | - Ana Carolina Irioda
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba P.O. Box 80240-020, Paraná, Brazil; (M.C.P.); (B.F.M.); (C.S.S.); (N.N.d.R.); (A.C.I.); (N.B.d.O.); (J.M.A.); (L.L.)
| | - Nathalia Barth de Oliveira
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba P.O. Box 80240-020, Paraná, Brazil; (M.C.P.); (B.F.M.); (C.S.S.); (N.N.d.R.); (A.C.I.); (N.B.d.O.); (J.M.A.); (L.L.)
| | - Julia Maurer Appel
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba P.O. Box 80240-020, Paraná, Brazil; (M.C.P.); (B.F.M.); (C.S.S.); (N.N.d.R.); (A.C.I.); (N.B.d.O.); (J.M.A.); (L.L.)
| | - Larissa Lührs
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba P.O. Box 80240-020, Paraná, Brazil; (M.C.P.); (B.F.M.); (C.S.S.); (N.N.d.R.); (A.C.I.); (N.B.d.O.); (J.M.A.); (L.L.)
| | - Leanderson Franco Meira
- Experimental Laboratory of the Institute of Biology and Health Sciences, Pontifical Catholic University of Paraná, Curitiba P.O. Box 80215-901, Paraná, Brazil; (L.F.M.); (L.C.G.-S.)
| | - Luiz Cesar Guarita-Souza
- Experimental Laboratory of the Institute of Biology and Health Sciences, Pontifical Catholic University of Paraná, Curitiba P.O. Box 80215-901, Paraná, Brazil; (L.F.M.); (L.C.G.-S.)
| | - Seigo Nagashima
- Laboratory of Experimental Pathology, Graduate Program of Health Sciences, School of Medicine, Pontifical Catholic University of Paraná (PUCPR), Curitiba P.O. Box 80215-901, Paraná, Brazil; (S.N.); (C.B.V.d.P.); (L.d.N.)
| | - Caroline Busatta Vaz de Paula
- Laboratory of Experimental Pathology, Graduate Program of Health Sciences, School of Medicine, Pontifical Catholic University of Paraná (PUCPR), Curitiba P.O. Box 80215-901, Paraná, Brazil; (S.N.); (C.B.V.d.P.); (L.d.N.)
| | - Lucia de Noronha
- Laboratory of Experimental Pathology, Graduate Program of Health Sciences, School of Medicine, Pontifical Catholic University of Paraná (PUCPR), Curitiba P.O. Box 80215-901, Paraná, Brazil; (S.N.); (C.B.V.d.P.); (L.d.N.)
| | - Idiberto José Zotarelli-Filho
- Postgraduate Program in Food, Nutrition and Food Engineering, Institute of Biosciences, Humanities and Exact Sciences (IBILCE), São Paulo State University (UNESP), São José do Rio Preto P.O. Box 15054-000, São Paulo, Brazil;
| | - Eltyeb Abdelwahid
- Feinberg Cardiovascular Research Institute, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA;
| | - Katherine Athayde Teixeira de Carvalho
- Advanced Therapy and Cellular Biotechnology in Regenerative Medicine Department, The Pelé Pequeno Príncipe Research Institute, Child and Adolescent Health Research & Pequeno Príncipe Faculties, Curitiba P.O. Box 80240-020, Paraná, Brazil; (M.C.P.); (B.F.M.); (C.S.S.); (N.N.d.R.); (A.C.I.); (N.B.d.O.); (J.M.A.); (L.L.)
| |
Collapse
|
33
|
Catlin JP, Tooley CES. Exploring potential developmental origins of common neurodegenerative disorders. Biochem Soc Trans 2024; 52:1035-1044. [PMID: 38661189 PMCID: PMC11440815 DOI: 10.1042/bst20230422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 03/12/2024] [Accepted: 04/11/2024] [Indexed: 04/26/2024]
Abstract
In the United States, it is now estimated that 6.7 million people over the age of 65 are afflicted by Alzheimer's disease (AD), over 1 million people are living with Parkinson's disease (PD), and over 200 000 have or are at risk for developing Huntington's disease (HD). All three of these neurodegenerative diseases result in the ultimate death of distinct neuronal subtypes, and it is widely thought that age-related damage is the single biggest contributing factor to this neuronal death. However, recent studies are now suggesting that developmental defects during early neurogenesis could also play a role in the pathology of neurodegenerative diseases. Loss or overexpression of proteins associated with HD, PD, and AD also result in embryonic phenotypes but whether these developmental defects slowly unmask over time and contribute to age-related neurodegeneration remains highly debated. Here, we discuss known links between embryonic neurogenesis and neurodegenerative disorders (including common signaling pathways), potential compensatory mechanisms that could delay presentation of neurodegenerative disorders, and the types of model systems that could be used to study these links in vivo.
Collapse
Affiliation(s)
- James P. Catlin
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Christine E. Schaner Tooley
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| |
Collapse
|
34
|
Yan Y, Cho AN. Human Brain In Vitro Model for Pathogen Infection-Related Neurodegeneration Study. Int J Mol Sci 2024; 25:6522. [PMID: 38928228 PMCID: PMC11204318 DOI: 10.3390/ijms25126522] [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: 04/15/2024] [Revised: 05/21/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Recent advancements in stem cell biology and tissue engineering have revolutionized the field of neurodegeneration research by enabling the development of sophisticated in vitro human brain models. These models, including 2D monolayer cultures, 3D organoids, organ-on-chips, and bioengineered 3D tissue models, aim to recapitulate the cellular diversity, structural organization, and functional properties of the native human brain. This review highlights how these in vitro brain models have been used to investigate the effects of various pathogens, including viruses, bacteria, fungi, and parasites infection, particularly in the human brain cand their subsequent impacts on neurodegenerative diseases. Traditional studies have demonstrated the susceptibility of different 2D brain cell types to infection, elucidated the mechanisms underlying pathogen-induced neuroinflammation, and identified potential therapeutic targets. Therefore, current methodological improvement brought the technology of 3D models to overcome the challenges of 2D cells, such as the limited cellular diversity, incomplete microenvironment, and lack of morphological structures by highlighting the need for further technological advancements. This review underscored the significance of in vitro human brain cell from 2D monolayer to bioengineered 3D tissue model for elucidating the intricate dynamics for pathogen infection modeling. These in vitro human brain cell enabled researchers to unravel human specific mechanisms underlying various pathogen infections such as SARS-CoV-2 to alter blood-brain-barrier function and Toxoplasma gondii impacting neural cell morphology and its function. Ultimately, these in vitro human brain models hold promise as personalized platforms for development of drug compound, gene therapy, and vaccine. Overall, we discussed the recent progress in in vitro human brain models, their applications in studying pathogen infection-related neurodegeneration, and future directions.
Collapse
Affiliation(s)
- Yuwei Yan
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW 2008, Australia;
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2050, Australia
| | - Ann-Na Cho
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Darlington, NSW 2008, Australia;
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW 2050, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| |
Collapse
|
35
|
Beliën J, Swinnen S, D'hondt R, Verdú de Juan L, Dedoncker N, Matthys P, Bauer J, Vens C, Moylett S, Dubois B. CHIT1 at diagnosis predicts faster disability progression and reflects early microglial activation in multiple sclerosis. Nat Commun 2024; 15:5013. [PMID: 38866782 PMCID: PMC11169395 DOI: 10.1038/s41467-024-49312-y] [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: 11/09/2023] [Accepted: 05/30/2024] [Indexed: 06/14/2024] Open
Abstract
Multiple sclerosis (MS) is characterized by heterogeneity in disease course and prediction of long-term outcome remains a major challenge. Here, we investigate five myeloid markers - CHIT1, CHI3L1, sTREM2, GPNMB and CCL18 - in the cerebrospinal fluid (CSF) at diagnostic lumbar puncture in a longitudinal cohort of 192 MS patients. Through mixed-effects and machine learning models, we show that CHIT1 is a robust predictor for faster disability progression. Integrative analysis of 11 CSF and 26 central nervous system (CNS) parenchyma single-cell/nucleus RNA sequencing samples reveals CHIT1 to be predominantly expressed by microglia located in active MS lesions and enriched for lipid metabolism pathways. Furthermore, we find CHIT1 expression to accompany the transition from a homeostatic towards a more activated, MS-associated cell state in microglia. Neuropathological evaluation in post-mortem tissue from 12 MS patients confirms CHIT1 production by lipid-laden phagocytes in actively demyelinating lesions, already in early disease stages. Altogether, we provide a rationale for CHIT1 as an early biomarker for faster disability progression in MS.
Collapse
Affiliation(s)
- Jarne Beliën
- Laboratory for Neuroimmunology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Stijn Swinnen
- Laboratory for Neuroimmunology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - Robbe D'hondt
- Department of Public Health and Primary Care, KU Leuven, Kortrijk, Belgium
- Imec research group itec, KU Leuven, Kortrijk, Belgium
| | - Laia Verdú de Juan
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Nina Dedoncker
- Laboratory for Neuroimmunology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Laboratory of Immunobiology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Jan Bauer
- Department of Neuroimmunology, Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Celine Vens
- Department of Public Health and Primary Care, KU Leuven, Kortrijk, Belgium
- Imec research group itec, KU Leuven, Kortrijk, Belgium
| | - Sinéad Moylett
- Laboratory for Neuroimmunology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium
| | - Bénédicte Dubois
- Laboratory for Neuroimmunology, Department of Neurosciences, Leuven Brain Institute, KU Leuven, Leuven, Belgium.
- Department of Neurology, University Hospitals Leuven, Leuven, Belgium.
| |
Collapse
|
36
|
Kukanja P, Langseth CM, Rubio Rodríguez-Kirby LA, Agirre E, Zheng C, Raman A, Yokota C, Avenel C, Tiklová K, Guerreiro-Cacais AO, Olsson T, Hilscher MM, Nilsson M, Castelo-Branco G. Cellular architecture of evolving neuroinflammatory lesions and multiple sclerosis pathology. Cell 2024; 187:1990-2009.e19. [PMID: 38513664 DOI: 10.1016/j.cell.2024.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Revised: 12/13/2023] [Accepted: 02/23/2024] [Indexed: 03/23/2024]
Abstract
Multiple sclerosis (MS) is a neurological disease characterized by multifocal lesions and smoldering pathology. Although single-cell analyses provided insights into cytopathology, evolving cellular processes underlying MS remain poorly understood. We investigated the cellular dynamics of MS by modeling temporal and regional rates of disease progression in mouse experimental autoimmune encephalomyelitis (EAE). By performing single-cell spatial expression profiling using in situ sequencing (ISS), we annotated disease neighborhoods and found centrifugal evolution of active lesions. We demonstrated that disease-associated (DA)-glia arise independently of lesions and are dynamically induced and resolved over the disease course. Single-cell spatial mapping of human archival MS spinal cords confirmed the differential distribution of homeostatic and DA-glia, enabled deconvolution of active and inactive lesions into sub-compartments, and identified new lesion areas. By establishing a spatial resource of mouse and human MS neuropathology at a single-cell resolution, our study unveils the intricate cellular dynamics underlying MS.
Collapse
Affiliation(s)
- Petra Kukanja
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden.
| | - Christoffer M Langseth
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden.
| | - Leslie A Rubio Rodríguez-Kirby
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Eneritz Agirre
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Chao Zheng
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Amitha Raman
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden
| | - Chika Yokota
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden
| | - Christophe Avenel
- Department of Information Technology, Uppsala University, 752 37 Uppsala, Sweden; BioImage Informatics Facility, Science for Life Laboratory, SciLifeLab, 751 05 Uppsala, Sweden
| | - Katarina Tiklová
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden
| | - André O Guerreiro-Cacais
- Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, 171 76 Solna, Sweden
| | - Tomas Olsson
- Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institutet, Karolinska University Hospital, 171 76 Solna, Sweden
| | - Markus M Hilscher
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden
| | - Mats Nilsson
- Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Solna, 17154 Stockholm, Sweden.
| | - Gonçalo Castelo-Branco
- Laboratory of Molecular Neurobiology, Department of Medical Biochemistry and Biophysics, Biomedicum, Karolinska Institutet, 17177 Stockholm, Sweden.
| |
Collapse
|
37
|
He Z, Chen Q, Wang K, Lin J, Peng Y, Zhang J, Yan X, Jie Y. Single-cell transcriptomics analysis of cellular heterogeneity and immune mechanisms in neurodegenerative diseases. Eur J Neurosci 2024; 59:333-357. [PMID: 38221677 DOI: 10.1111/ejn.16242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 12/04/2023] [Accepted: 12/12/2023] [Indexed: 01/16/2024]
Abstract
Single-cell transcriptomics analysis is an advanced technology that can describe the intracellular transcriptome in complex tissues. It profiles and analyses datasets by single-cell RNA sequencing. Neurodegenerative diseases are identified by the abnormal apoptosis of neurons in the brain with few or no effective therapy strategies at present, which has been a growing healthcare concern and brought a great burden to society. The transcriptome of individual cells provides deep insights into previously unforeseen cellular heterogeneity and gene expression differences in neurodegenerative disorders. It detects multiple cell subsets and functional changes during pathological progression, which deepens the understanding of the molecular underpinnings and cellular basis of neurodegenerative diseases. Furthermore, the transcriptome analysis of immune cells shows the regulation of immune response. Different subtypes of immune cells and their interaction are found to contribute to disease progression. This finding enables the discovery of novel targets and biomarkers for early diagnosis. In this review, we emphasize the principles of the technology, and its recent progress in the study of cellular heterogeneity and immune mechanisms in neurodegenerative diseases. The application of single-cell transcriptomics analysis in neurodegenerative disorders would help explore the pathogenesis of these diseases and develop novel therapeutic methods.
Collapse
Affiliation(s)
- Ziping He
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, China
| | - Qianqian Chen
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Kaiyue Wang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
- Clinical Medicine Eight-Year Program, Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiang Lin
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Yilin Peng
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Jinlong Zhang
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
- Department of Forensic Science, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| | - Xisheng Yan
- Department of Cardiovascular Medicine, Wuhan Third Hospital & Tongren Hospital of Wuhan University, Wuhan, China
| | - Yan Jie
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
- Department of Forensic Science, School of Basic Medical Science, Xinjiang Medical University, Urumqi, China
| |
Collapse
|
38
|
Xu L, Min H, Saha A, Gunaratne A, Schwartzman J, Parrott R, Kurtzberg J, Filiano AJ. Mesenchymal stromal cells suppress microglial activation and tumor necrosis factor production. Cytotherapy 2024; 26:185-193. [PMID: 38054911 DOI: 10.1016/j.jcyt.2023.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 11/03/2023] [Accepted: 11/16/2023] [Indexed: 12/07/2023]
Abstract
BACKGROUND AIMS White matter diseases are commonly associated with microglial activation and neuroinflammation. Mesenchymal stromal cells (MSCs) have immunomodulatory properties and thus have the potential to be developed as cell therapy for white matter disease. MSCs interact with resident macrophages to alter the trajectory of inflammation; however, the impact MSCs have on central nervous system macrophages and the effect this has on the progression of white matter disease are unclear. METHODS In this study, we utilized numerous assays of varying complexity to model different aspects of white matter disease. These assays ranged from an in vivo spinal cord acute demyelination model to a simple microglial cell line activation assay. Our goal was to investigate the influence of human umbilical cord tissue MSCs on the activation of microglia. RESULTS MSCs reduced the production of tumor necrosis factor (TNF) by microglia and decreased demyelinated lesions in the spinal cord after acute focal injury. To determine if MSCs could directly suppress the activation of microglia and to develop an efficient potency assay, we utilized isolated primary microglia from mouse brains and the Immortalized MicroGlial Cell Line (IMG). MSCs suppressed the activation of microglia and the release of TNF after stimulation with lipopolysaccharide, a toll-like receptor agonist. CONCLUSIONS In this study, we demonstrated that MSCs altered the immune response after acute injury in the spinal cord. In numerous assays, MSCs suppressed activation of microglia and release of the pro-inflammatory cytokine TNF. Of these assays, IMG could be standardized and used as an effective potency assay to determine the efficacy of MSCs for treating white matter disease or other neuroinflammatory conditions associated with microglial activation.
Collapse
Affiliation(s)
- Li Xu
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Hyunjung Min
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Arjun Saha
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Aruni Gunaratne
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA; Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | | | - Roberta Parrott
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA
| | - Joanne Kurtzberg
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA; Department of Pediatrics, Duke University, Durham, North Carolina, USA
| | - Anthony J Filiano
- Marcus Center for Cellular Cures, Duke University, Durham, North Carolina, USA; Department of Neurosurgery, Duke University, Durham, North Carolina, USA; Department of Integrative Immunobiology, Duke University, Durham, North Carolina, USA; Department of Pathology, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
39
|
Greenberg BM. Bruton's Tyrosine Kinase Inhibitors for Multiple Sclerosis Treatment: A New Frontier. Neurol Clin 2024; 42:155-163. [PMID: 37980113 DOI: 10.1016/j.ncl.2023.07.006] [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: 11/20/2023]
Abstract
Multiple sclerosis (MS) can cause significant disability to patients via relapse-associated worsening and progression independent of relapses. The causes of neuronal and myelin damage can include lymphocyte-mediated inflammation and microglial activation. Bruton's tyrosine kinase (BTK) is an enzyme that mediates B cell activation and the proinflammatory phenotype of microglia. Inhibiting BTK provides a novel therapeutic target for MS but also has a complicated pharmacology based on binding specificity, CNS penetration, half-life, and enzyme inhibition characteristics. Multiple agents are being studied in phase 3 trials, and each agent will have unique efficacy and safety profiles that must be considered individually.
Collapse
Affiliation(s)
- Benjamin M Greenberg
- Department of Neurology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA.
| |
Collapse
|
40
|
Bravo-San Pedro JM, Aranda F, Buqué A, Galluzzi L. Preface. Methods Cell Biol 2024; 185:xvii-xxiv. [PMID: 38556455 DOI: 10.1016/s0091-679x(24)00112-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/02/2024]
Affiliation(s)
- José Manuel Bravo-San Pedro
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IdiSNA), Pamplona, Spain
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States
| |
Collapse
|
41
|
Bravo-San Pedro JM, Aranda F, Buqué A, Galluzzi L. Animal models of disease: Achievements and challenges. Methods Cell Biol 2024; 188:xv-xxi. [PMID: 38880531 DOI: 10.1016/s0091-679x(24)00164-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Affiliation(s)
- José Manuel Bravo-San Pedro
- Departamento de Fisiología, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Fernando Aranda
- Program of Immunology and Immunotherapy, Cima Universidad de Navarra, Pamplona, Spain; Navarra Institute for Health Research (IDISNA), Pamplona, Spain
| | - Aitziber Buqué
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, United States; Sandra and Edward Meyer Cancer Center, New York, NY, United States; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, United States.
| |
Collapse
|
42
|
Doghish AS, Elazazy O, Mohamed HH, Mansour RM, Ghanem A, Faraag AHI, Elballal MS, Elrebehy MA, Elesawy AE, Abdel Mageed SS, Mohammed OA, Nassar YA, Abulsoud AI, Raouf AA, Abdel-Reheim MA, Rashad AA, Elawady AS, Elsisi AM, Alsalme A, Ali MA. The role of miRNAs in multiple sclerosis pathogenesis, diagnosis, and therapeutic resistance. Pathol Res Pract 2023; 251:154880. [PMID: 37832353 DOI: 10.1016/j.prp.2023.154880] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Revised: 10/06/2023] [Accepted: 10/07/2023] [Indexed: 10/15/2023]
Abstract
In recent years, microRNAs (miRNAs) have gained increased attention from researchers around the globe. Although it is twenty nucleotides long, it can modulate several gene targets simultaneously. Their mal expression is a signature of various pathologies, and they provide the foundation to elucidate the molecular mechanisms of each pathology. Among the debilitating central nervous system (CNS) disorders with a growing prevalence globally is the multiple sclerosis (MS). Moreover, the diagnosis of MS is challenging due to the lack of disease-specific biomarkers, and the diagnosis mainly depends on ruling out other disabilities. MS could adversely affect patients' lives through its progression, and only symptomatic treatments are available as therapeutic options, but an exact cure is yet unavailable. Consequently, this review hopes to further the study of the biological features of miRNAs in MS and explore their potential as a therapeutic target.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt.
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Hend H Mohamed
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Biochemistry Department, Faculty of Science, Cairo University, Giza 12613, Egypt
| | - Reda M Mansour
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan 11795, Egypt; Biology Department, School of Biotechnology, Badr University in Cairo, Badr City, Cairo 11829, Egypt
| | - Aml Ghanem
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Ahmed H I Faraag
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt; Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan 11795, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt.
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Osama A Mohammed
- Department of Clinical Pharmacology, College of Medicine, University of Bisha, Bisha 61922, Saudi Arabia
| | - Yara A Nassar
- Biology Department, School of Biotechnology, Badr University in Cairo, Badr City, Cairo 11829, Egypt; Department of Botany, Faculty of Science, Mansoura University, Mansoura 35516, Egypt
| | - Ahmed I Abulsoud
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt; Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo 11785, Egypt
| | - Ahmed Amr Raouf
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef 62521, Egypt.
| | - Ahmed A Rashad
- Department of Clinical Pharmacy and Pharmacy Practice, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| | - Alaa S Elawady
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig 44519, Egypt
| | - Ahmed Mohammed Elsisi
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Cairo 11231, Egypt; Department of Biochemistry, Faculty of Pharmacy, Sinai University, Al-Arish, Egypt
| | - Ali Alsalme
- Chemistry Department, College of Science, King Saud University, Riyadh 1145, Saudi Arabia
| | - Mohamed A Ali
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Cairo 11829, Egypt
| |
Collapse
|
43
|
Belousova O, Lopatina A, Kuzmina U, Melnikov M. The role of biogenic amines in the modulation of monocytes in autoimmune neuroinflammation. Mult Scler Relat Disord 2023; 78:104920. [PMID: 37536214 DOI: 10.1016/j.msard.2023.104920] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 07/18/2023] [Accepted: 07/28/2023] [Indexed: 08/05/2023]
Abstract
Multiple sclerosis (MS) is inflammatory demyelinating and neurodegenerative disease of the central nervous system (CNS) with autoimmune mechanism of development. The study of the neuroimmune interactions is one of the most developing directions in the research of the pathogenesis of MS. The influence of biogenic amines on the pathogenesis of experimental autoimmune encephalomyelitis (EAE) and MS was shown by the modulation of subsets of T-helper cells and B-cells, which plays a crucial role in the autoimmunity of the CNS. However, along with T- and B-cells the critical involvement of mononuclear phagocytes such as dendritic cells, macrophages, and monocytes in the development of neuroinflammation also was shown. It was demonstrated that the activation of microglial cells (resident macrophages of the CNS) could initiate the neuroinflammation in the EAE, suggesting their role at an early stage of the disease. In contrast, monocytes, which migrate from the periphery into the CNS through the blood-brain barrier, mediate the effector phase of the disease and cause neurological disability in EAE. In addition, the clinical efficacy of the therapy with depletion of the monocytes in EAE was shown, suggesting their crucial role in the autoimmunity of the CNS. Biogenic amines, such as epinephrine, norepinephrine, dopamine, and serotonin are direct mediators of the neuroimmune interaction and may affect the pathogenesis of EAE and MS by modulating the immune cell activity and cytokine production. The anti-inflammatory effect of targeting the biogenic amines receptors on the pathogenesis of EAE and MS by suppression of Th17- and Th1-cells, which are critical for the CNS autoimmunity, was shown. However, the latest data showed the potential ability of biogenic amines to affect the functions of the mononuclear phagocytes and their involvement in the modulation of neuroinflammation. This article reviews the literature data on the role of monocytes in the pathogenesis of EAE and MS. The data on the effect of targeting of biogenic amine receptors on the function of monocytes are presented.
Collapse
Affiliation(s)
- Olga Belousova
- Laboratory of Neuroimmunology, Federal Center of Brain Research and Neurotechnology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Anna Lopatina
- Laboratory of Neuroimmunology, Federal Center of Brain Research and Neurotechnology of the Federal Medical-Biological Agency of Russia, Moscow, Russia
| | - Ulyana Kuzmina
- Laboratory of Neuroimmunology, Federal Center of Brain Research and Neurotechnology of the Federal Medical-Biological Agency of Russia, Moscow, Russia; Laboratory of Molecular Pharmacology and Immunology, Institute of Biochemistry and Genetics - Subdivision of the Ufa Federal Research Center of the Russian Academy of Science, Ufa, Russia
| | - Mikhail Melnikov
- Laboratory of Neuroimmunology, Federal Center of Brain Research and Neurotechnology of the Federal Medical-Biological Agency of Russia, Moscow, Russia; Department of Neurology, Neurosurgery and Medical Genetics, Pirogov Russian National Research Medical University, Moscow, Russia; Laboratory of Clinical Immunology, National Research Center Institute of Immunology of the Federal Medical-Biological Agency of Russia, Moscow, Russia.
| |
Collapse
|
44
|
Zong B, Yu F, Zhang X, Zhao W, Li S, Li L. Mechanisms underlying the beneficial effects of physical exercise on multiple sclerosis: focus on immune cells. Front Immunol 2023; 14:1260663. [PMID: 37841264 PMCID: PMC10570846 DOI: 10.3389/fimmu.2023.1260663] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 09/13/2023] [Indexed: 10/17/2023] Open
Abstract
Multiple sclerosis (MS) is a prevalent neuroimmunological illness that leads to neurological disability in young adults. Although the etiology of MS is heterogeneous, it is well established that aberrant activity of adaptive and innate immune cells plays a crucial role in its pathogenesis. Several immune cell abnormalities have been described in MS and its animal models, including T lymphocytes, B lymphocytes, dendritic cells, neutrophils, microglia/macrophages, and astrocytes, among others. Physical exercise offers a valuable alternative or adjunctive disease-modifying therapy for MS. A growing body of evidence indicates that exercise may reduce the autoimmune responses triggered by immune cells in MS. This is partially accomplished by restricting the infiltration of peripheral immune cells into the central nervous system (CNS) parenchyma, curbing hyperactivation of immune cells, and facilitating a transition in the balance of immune cells from a pro-inflammatory to an anti-inflammatory state. This review provides a succinct overview of the correlation between physical exercise, immune cells, and MS pathology, and highlights the potential benefits of exercise as a strategy for the prevention and treatment of MS.
Collapse
Affiliation(s)
- Boyi Zong
- College of Physical Education and Health, East China Normal University, Shanghai, China
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Fengzhi Yu
- College of Physical Education and Health, East China Normal University, Shanghai, China
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
- School of Exercise and Health, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai, China
| | - Xiaoyou Zhang
- School of Physical Education, Hubei University, Wuhan, China
| | - Wenrui Zhao
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Shichang Li
- College of Physical Education and Health, East China Normal University, Shanghai, China
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| | - Lin Li
- College of Physical Education and Health, East China Normal University, Shanghai, China
- Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai, China
| |
Collapse
|
45
|
Matsuzaka Y, Yashiro R. Unraveling the Immunopathogenesis of Multiple Sclerosis: The Dynamic Dance of Plasmablasts and Pathogenic T Cells. BIOLOGICS 2023; 3:232-252. [DOI: 10.3390/biologics3030013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory demyelinating disease of the central nervous system, characterized by multiple lesions occurring temporally and spatially. Additionally, MS is a disease that predominates in the white population. In recent years, there has been a rapid increase in the number of patients, and it often occurs in young people, with an average age of onset of around 30 years old, but it can also occur in children and the elderly. It is more common in women than men, with a male-to-female ratio of approximately 1:3. As the immunopathogenesis of MS, a group of B cells called plasmablasts controls encephalomyelitis via IL-10 production. These IL-10-producing B cells, called regulatory B cells, suppress inflammatory responses in experimental mouse models of autoimmune diseases including MS. Since it has been clarified that these regulatory B cells are plasmablasts, it is expected that the artificial control of plasmablast differentiation will lead to the development of new treatments for MS. Among CD8-positive T cells in the peripheral blood, the proportion of PD-1-positive cells is decreased in MS patients compared with healthy controls. The dysfunction of inhibitory receptors expressed on T cells is known to be the core of MS immunopathology and may be the cause of chronic persistent inflammation. The PD-1+ CD8+ T cells may also serve as indicators that reflect the condition of each patient in other immunological neurological diseases such as MS. Th17 cells also regulate the development of various autoimmune diseases, including MS. Thus, the restoration of weakened immune regulatory functions may be a true disease-modifying treatment. So far, steroids and immunosuppressants have been the mainstream for autoimmune diseases, but the problem is that this kills not only pathogenic T cells, but also lymphocytes, which are necessary for the body. From this understanding of the immune regulation of MS, we can expect the development of therapeutic strategies that target only pathogenic immune cells.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Ryu Yashiro
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|
46
|
Zong B, Yu F, Zhang X, Pang Y, Zhao W, Sun P, Li L. Mechanosensitive Piezo1 channel in physiology and pathophysiology of the central nervous system. Ageing Res Rev 2023; 90:102026. [PMID: 37532007 DOI: 10.1016/j.arr.2023.102026] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/29/2023] [Accepted: 07/29/2023] [Indexed: 08/04/2023]
Abstract
Since the discovery of the mechanosensitive Piezo1 channel in 2010, there has been a significant amount of research conducted to explore its regulatory role in the physiology and pathology of various organ systems. Recently, a growing body of compelling evidence has emerged linking the activity of the mechanosensitive Piezo1 channel to health and disease of the central nervous system. However, the exact mechanisms underlying these associations remain inadequately comprehended. This review systematically summarizes the current research on the mechanosensitive Piezo1 channel and its implications for central nervous system mechanobiology, retrospects the results demonstrating the regulatory role of the mechanosensitive Piezo1 channel on various cell types within the central nervous system, including neural stem cells, neurons, oligodendrocytes, microglia, astrocytes, and brain endothelial cells. Furthermore, the review discusses the current understanding of the involvement of the Piezo1 channel in central nervous system disorders, such as Alzheimer's disease, multiple sclerosis, glaucoma, stroke, and glioma.
Collapse
Affiliation(s)
- Boyi Zong
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Fengzhi Yu
- School of Exercise and Health, Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xiaoyou Zhang
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Yige Pang
- Department of Neurosurgery, Zibo Central Hospital, Zibo 255000, Shandong, China
| | - Wenrui Zhao
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua 321004, Zhejiang, China
| | - Peng Sun
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China
| | - Lin Li
- College of Physical Education and Health, East China Normal University, Shanghai 200241, China; Key Laboratory of Adolescent Health Assessment and Exercise Intervention of Ministry of Education, East China Normal University, Shanghai 200241, China.
| |
Collapse
|