1
|
Das S, Shaw AK, Das Sarma S, Koval M, Das Sarma J, Maulik M. Neurotropic Murine β-Coronavirus Infection Causes Differential Expression of Connexin 47 in Oligodendrocyte Subpopulations Associated with Demyelination. Mol Neurobiol 2024:10.1007/s12035-024-04482-0. [PMID: 39292341 DOI: 10.1007/s12035-024-04482-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
Gap junctions (GJs) play a crucial role in the survival of oligodendrocytes and myelination of the central nervous system (CNS). In this study, we investigated the spatiotemporal changes in the expression of oligodendroglial GJ protein connexin 47 (Cx47), its primary astroglial coupling partner, Cx43, and their association with demyelination following intracerebral infection with mouse hepatitis virus (MHV). Neurotropic strains of MHV, a β-coronavirus, induce an acute encephalomyelitis followed by a chronic demyelinating disease that shares similarities with the human disease multiple sclerosis (MS). Our results reveal that Cx47 GJs are persistently lost in mature oligodendrocytes, not only in demyelinating lesions but also in surrounding normal appearing white and gray matter areas, following an initial loss of astroglial Cx43 GJs during acute infection. At later stages after viral clearance, astroglial Cx43 GJs re-emerge but mature oligodendrocytes fail to fully re-establish GJs with astrocytes due to lack of Cx47 GJ expression. In contrast, at this later demyelinating stage, the increased oligodendrocyte precursor cells appear to exhibit Cx47 GJs. Our findings further highlight varying degrees of demyelination in distinct spinal cord regions, with the thoracic cord showing the most pronounced demyelination. The regional difference in demyelination correlates well with dynamic changes in the proportion of different oligodendrocyte lineage cells exhibiting differential Cx47 GJ expression, suggesting an important mechanism of progressive demyelination even after viral clearance.
Collapse
Affiliation(s)
- Soubhik Das
- Biotechnology Research and Innovation Council - National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, 741251, West Bengal, India
| | - Archana Kumari Shaw
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, West Bengal, India
| | - Subhajit Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, West Bengal, India
| | - Michael Koval
- Departments of Medicine and Cell Biology, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, 741246, West Bengal, India
| | - Mahua Maulik
- Biotechnology Research and Innovation Council - National Institute of Biomedical Genomics (BRIC-NIBMG), Kalyani, 741251, West Bengal, India.
| |
Collapse
|
2
|
Mireles-Ramírez MA, Pacheco-Moises FP, González-Usigli HA, Sánchez-Rosales NA, Hernández-Preciado MR, Delgado-Lara DLC, Hernández-Cruz JJ, Ortiz GG. Neuromyelitis optica spectrum disorder: pathophysiological approach. Int J Neurosci 2024; 134:826-838. [PMID: 36453541 DOI: 10.1080/00207454.2022.2153046] [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: 03/12/2022] [Revised: 11/19/2022] [Accepted: 11/24/2022] [Indexed: 12/02/2022]
Abstract
Aim: To review the main pathological findings of Neuromyelitis Optica Spectrum Disorder (NMOSD) associated with the presence of autoantibodies to aquaporin-4 (AQP4) as well as the mechanisms of astrocyte dysfunction and demyelination. Methods: An comprehensive search of the literature in the field was carried out using the database of The National Center for Biotechnology Information from . Systematic searches were performed until July 2022. Results: NMOSD is an inflammatory and demyelinating disease of the central nervous system mainly in the areas of the optic nerves and spinal cord, thus explaining mostly the clinical findings. Other areas affected in NMOSD are the brainstem, hypothalamus, and periventricular regions. Relapses in NMOSD are generally severe and patients only partially recover. NMOSD includes clinical conditions where autoantibodies to aquaporin-4 (AQP4-IgG) of astrocytes are detected as well as similar clinical conditions where such antibodies are not detected. AQP4 are channel-forming integral membrane proteins of which AQ4 isoforms are able to aggregate in supramolecular assemblies termed orthogonal arrays of particles (OAP) and are essential in the regulation of water homeostasis and the adequate modulation of neuronal activity and circuitry. AQP4 assembly in orthogonal arrays of particles is essential for AQP4-IgG pathogenicity since AQP4 autoantibodies bind to OAPs with higher affinity than for AQP4 tetramers. NMOSD has a complex background with prominent roles for genes encoding cytokines and cytokine receptors. AQP4 autoantibodies activate the complement-mediated inflammatory demyelination and the ensuing damage to AQP4 water channels, leading to water influx, necrosis and axonal loss. Conclusions: NMOSD as an astrocytopathy is a nosological entity different from multiple sclerosis with its own serological marker: immunoglobulin G-type autoantibodies against the AQP4 protein which elicits a complement-dependent cytotoxicity and neuroinflammation. Some patients with typical manifestations of NMSOD are AQP4 seronegative and myelin oligodendrocyte glycoprotein positive. Thus, the detection of autoantibodies against AQP4 or other autoantibodies is crucial for the correct treatment of the disease and immunosuppressant therapy is the first choice.
Collapse
Affiliation(s)
- Mario A Mireles-Ramírez
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | - Fermín P Pacheco-Moises
- Department of Chemistry, University Center of Exact Sciences and Engineering; University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Héctor A González-Usigli
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | - Nayeli A Sánchez-Rosales
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | - Martha R Hernández-Preciado
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
| | | | - José J Hernández-Cruz
- Department of Philosophical and Methodological Disciplines and Service of Molecular Biology in medicine HC, University Health Sciences Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| | - Genaro Gabriel Ortiz
- Department of Neurology, High Specialty Medical Unit, Western National Medical Center of the Mexican Institute of Social Security, Guadalajara, Jalisco, Mexico
- Department of Philosophical and Methodological Disciplines and Service of Molecular Biology in medicine HC, University Health Sciences Center, University of Guadalajara, Guadalajara, Jalisco, Mexico
| |
Collapse
|
3
|
Takase EO, Yamasaki R, Nagata S, Watanabe M, Masaki K, Yamaguchi H, Kira JI, Takeuchi H, Isobe N. Astroglial connexin 43 is a novel therapeutic target for chronic multiple sclerosis model. Sci Rep 2024; 14:10877. [PMID: 38740862 PMCID: PMC11091090 DOI: 10.1038/s41598-024-61508-2] [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: 10/29/2023] [Accepted: 05/07/2024] [Indexed: 05/16/2024] Open
Abstract
In chronic stages of multiple sclerosis (MS) and its animal model, experimental autoimmune encephalitis (EAE), connexin (Cx)43 gap junction channel proteins are overexpressed because of astrogliosis. To elucidate the role of increased Cx43, the central nervous system (CNS)-permeable Cx blocker INI-0602 was therapeutically administered. C57BL6 mice with chronic EAE initiated by MOG35-55 received INI-0602 (40 mg/kg) or saline intraperitoneally every other day from days post-immunization (dpi) 17-50. Primary astroglia were employed to observe calcein efflux responses. In INI-0602-treated mice, EAE clinical signs improved significantly in the chronic phase, with reduced demyelination and decreased CD3+ T cells, Iba-1+ and F4/80+ microglia/macrophages, and C3+GFAP+ reactive astroglia infiltration in spinal cord lesions. Flow cytometry analysis of CD4+ T cells from CNS tissues revealed significantly reduced Th17 and Th17/Th1 cells (dpi 24) and Th1 cells (dpi 50). Multiplex array of cerebrospinal fluid showed significantly suppressed IL-6 and significantly increased IL-10 on dpi 24 in INI-0602-treated mice, and significantly suppressed IFN-γ and MCP-1 on dpi 50 in the same group. In vitro INI-0602 treatment inhibited ATP-induced calcium propagations of Cx43+/+ astroglial cells to similar levels of those of Cx43-/- cells. Astroglial Cx43 hemichannels represent a novel therapeutic target for chronic EAE and MS.
Collapse
Affiliation(s)
- Ezgi Ozdemir Takase
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Satoshi Nagata
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Mitsuru Watanabe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroo Yamaguchi
- School of Physical Therapy, Faculty of Rehabilitation, Reiwa Health Sciences University, Fukuoka, Japan
| | - Jun-Ichi Kira
- Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, Ookawa, Japan
- Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, Fukuoka, Japan
| | - Hideyuki Takeuchi
- Department of Neurology and Stroke Medicine, Graduate School of Medicine, Yokohama City University, 3-9 Fukuura, Kanazawa-ku, Yokohama, 236-0004, Japan.
- Department of Neurology, Graduate School of Medicine, International University of Health and Welfare, Narita, Japan.
- Center for Intractable Neurological Diseases and Dementia, International University of Health and Welfare Atami Hospital, Atami, Japan.
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
4
|
Okolo CA, Maran JJ, Watts A, Maripillan J, Harkiolaki M, Martínez AD, Green CR, Mugisho OO. Correlative light and X-ray tomography jointly unveil the critical role of connexin43 channels on inflammation-induced cellular ultrastructural alterations. Heliyon 2024; 10:e27888. [PMID: 38560181 PMCID: PMC10979075 DOI: 10.1016/j.heliyon.2024.e27888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 03/06/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
Non-junctional connexin43 (Cx43) plasma membrane hemichannels have been implicated in several inflammatory diseases, particularly playing a role in ATP release that triggers activation of the inflammasome. Therapies targeting the blocking of the hemichannels to prevent the pathological release or uptake of ions and signalling molecules through its pores are of therapeutic interest. To date, there is no close-to-native, high-definition documentation of the impact of Cx43 hemichannel-mediated inflammation on cellular ultrastructure, neither is there a robust account of the ultrastructural changes that occur following treatment with selective Cx43 hemichannel blockers such as Xentry-Gap19 (XG19). A combination of same-sample correlative high-resolution three-dimensional fluorescence microscopy and soft X-ray tomography at cryogenic temperatures, enabled in the identification of novel 3D molecular interactions within the cellular milieu when comparing behaviour in healthy states and during the early onset or late stages under inflammatory conditions. Notably, our findings suggest that XG19 blockage of connexin hemichannels under pro-inflammatory conditions may be crucial in preventing the direct degradation of connexosomes by lysosomes, without affecting connexin protein translation and trafficking. We also delineated fine and gross cellular phenotypes, characteristic of inflammatory insult or road-to-recovery from inflammation, where XG19 could indirectly prevent and reverse inflammatory cytokine-induced mitochondrial swelling and cellular hypertrophy through its action on Cx43 hemichannels. Our findings suggest that XG19 might have prophylactic and therapeutic effects on the inflammatory response, in line with functional studies.
Collapse
Affiliation(s)
- Chidinma Adanna Okolo
- Beamline B24, Life Sciences Division, Diamond Light Source, Didcot, Oxfordshire, United Kingdom
| | - Jack Jonathan Maran
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, New Zealand
| | - Amy Watts
- Beamline B24, Life Sciences Division, Diamond Light Source, Didcot, Oxfordshire, United Kingdom
| | - Jaime Maripillan
- Centro Interdisciplinario de Neurociencias de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
| | - Maria Harkiolaki
- Beamline B24, Life Sciences Division, Diamond Light Source, Didcot, Oxfordshire, United Kingdom
| | - Agustín D. Martínez
- Centro Interdisciplinario de Neurociencias de Valparaíso (CINV), Universidad de Valparaíso, Valparaíso, Chile
| | - Colin R. Green
- Department of Ophthalmology, University of Auckland, New Zealand
| | - Odunayo Omolola Mugisho
- Buchanan Ocular Therapeutics Unit, Department of Ophthalmology, New Zealand National Eye Centre, University of Auckland, New Zealand
| |
Collapse
|
5
|
Li J, Qi H, Chen Y, Zhu X. Epilepsy and demyelination: Towards a bidirectional relationship. Prog Neurobiol 2024; 234:102588. [PMID: 38378072 DOI: 10.1016/j.pneurobio.2024.102588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Accepted: 02/13/2024] [Indexed: 02/22/2024]
Abstract
Demyelination stands out as a prominent feature in individuals with specific types of epilepsy. Concurrently, individuals with demyelinating diseases, such as multiple sclerosis (MS) are at a greater risk of developing epilepsy compared to non-MS individuals. These bidirectional connections raise the question of whether both pathological conditions share common pathogenic mechanisms. This review focuses on the reciprocal relationship between epilepsy and demyelination diseases. We commence with an overview of the neurological basis of epilepsy and demyelination diseases, followed by an exploration of how our comprehension of these two disorders has evolved in tandem. Additionally, we discuss the potential pathogenic mechanisms contributing to the interactive relationship between these two diseases. A more nuanced understanding of the interplay between epilepsy and demyelination diseases has the potential to unveiling the molecular intricacies of their pathological relationships, paving the way for innovative directions in future clinical management and treatment strategies for these diseases.
Collapse
Affiliation(s)
- Jiayi Li
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China; Clinical Medicine, Medical School of Southeast University, Nanjing, China
| | - Honggang Qi
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China
| | - Yuzhou Chen
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China; Clinical Medicine, Medical School of Southeast University, Nanjing, China
| | - Xinjian Zhu
- Department of Pharmacology, Medical School of Southeast University, Nanjing, China.
| |
Collapse
|
6
|
Yang Z, Yang L, Zhang J, Qian C, Zhao Y. AS602801 treatment suppresses breast cancer metastasis to the brain by interfering with gap-junction communication by regulating Cx43 expression. Drug Dev Res 2024; 85:e22124. [PMID: 37859299 DOI: 10.1002/ddr.22124] [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: 07/16/2023] [Revised: 09/21/2023] [Accepted: 10/08/2023] [Indexed: 10/21/2023]
Abstract
AS602801 has been reported as a potential drug candidate against brain metastasis by suppressing the gap-junction communication between lung cancer stem cells and astrocytes. In this study, we aimed to study the molecular mechanism underlying the role of AS602801 in the treatment of brain metastasis in breast cancer. We utilized female athymic BALB/c nude mice and MDA-MB-231/BT-474BR cells to establish experimental models. Polymerase chain reaction assays were performed to observe changes in the connexin 43 (Cx43) messenger RNA (mRNA) and c-Jun N-terminal kinase (JNK) mRNA levels. Dye transfer assay was used to observe the effect of AS602801 on cell-cell communication. An organotypic blood-brain barrier (BBB) model was utilized to observe the effect of AS602801 on transmigration through the BBB barrier. MTT (3-[4,5-dimethylthiazol-2-yl]-2,5 diphenyl tetrazolium bromide) assay and flow cytometry were performed to evaluate the proliferation and apoptosis of breast cancer cells co-cultivated with astrocytes. AS602801 inhibited the upregulation of Cx43 and JNK in brain metastasized breast cancer cells in a dose-dependent manner. Also, AS602801 significantly decreased the dye transfer rate from astrocytes to breast cancer cells, indicating the inhibitory effect of AS602801 on cell-cell communication. The transmigration ability of breast cancer cells co-cultured with astrocytes was decreased by AS602801. Furthermore, AS602801 reduced the elevated Cx43/JNK mRNA expression in the co-astrocyte group while suppressing the increased proliferation and promoting the decreased apoptosis of breast cancer cells co-cultivated with astrocytes. AS602801 also suppressed the brain metastasis of breast cancer cells and increased mouse survival. AS602801 downregulates the expressions of JNK and Cx43 to suppress the gap-junction activity. AS602801 also inhibits the communication between breast cancer cells and astrocytes, thus contributing to the treatment of brain metastasis in breast cancer.
Collapse
Affiliation(s)
- Zhigang Yang
- Department of General Surgery, Shidong Hospital, Yangpu District, Shanghai, China
| | - Liguo Yang
- Department of General Surgery, Shidong Hospital, Yangpu District, Shanghai, China
| | - Jun Zhang
- Department of General Surgery, Shidong Hospital, Yangpu District, Shanghai, China
| | - Chenzeyue Qian
- Department of General Surgery, Shidong Hospital, Yangpu District, Shanghai, China
| | - Yi Zhao
- Department of General Surgery, Shidong Hospital, Yangpu District, Shanghai, China
| |
Collapse
|
7
|
Wang H, Yang X, Lai H, Sun Y, Yan X, Ai Q, Lin M, Yang S, Yang Y, Chu S, Wang Z, Chen N. Novel antidepressant mechanism of hypericin: Role of connexin 43-based gap junctions. Biomed Pharmacother 2023; 167:115545. [PMID: 37734259 DOI: 10.1016/j.biopha.2023.115545] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 09/13/2023] [Accepted: 09/18/2023] [Indexed: 09/23/2023] Open
Abstract
Hypericin is widely utilized for its precise antidepressant properties, but its exact antidepressant mechanism remains unclear. Gap junctions, which were predominantly expressed in astrocytes in the central nervous system, are concerned with the pathogenesis of depression. However, the role of hypericin in gap junctional dysfunction in depression has rarely been investigated. Here, we found that gap junctions were ultra-structurally broadened in the chronic unpredictable stress (CUS) rat model of depression, while hypericin repaired the dysfunction of gap junctions. Suppression of gap junctions by bilateral injection of carbenoxolone (CBX) in the prefrontal cortex of rats significantly inhibited the restoration of gap junctional dysfunction in depression by hypericin. Meanwhile, hypericin failed to show antidepressant benefits. Furthermore, in corticosterone (CORT)-stimulated primary astrocytes derived from neonatal rats, hypericin dramatically reversed the phosphorylation of connexin 43 (Cx43), normalizing the expression of Cx43 and thereby ameliorating gap junctional dysfunction. Comparatively, CBX inhibited the remission of hypericin on gap junctional intercellular communication function. Gap junctional function might be a novel therapeutic target for hypericin in the treatment of depression and provide potential novel insights into the antidepressant mechanism of other herbal ingredients.
Collapse
Affiliation(s)
- Huiqin Wang
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha 410208, Hunan, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Xueying Yang
- School of Chinese Materia Medica, Guangdong Pharmaceutical University, Guangzhou 510006, Guangdong, China
| | - Huaqing Lai
- Science and Technology Innovation Center, Guangzhou University of Chinese Medicine, Guangzhou 510405, Guangdong, China
| | - Yang Sun
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha 410208, Hunan, China
| | - Xu Yan
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Qidi Ai
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha 410208, Hunan, China
| | - Meiyu Lin
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha 410208, Hunan, China
| | - Songwei Yang
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha 410208, Hunan, China
| | - Yantao Yang
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha 410208, Hunan, China
| | - Shifeng Chu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China
| | - Zhenzhen Wang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| | - Naihong Chen
- Hunan University of Chinese Medicine & Hunan Engineering Technology Center of Standardization and Function of Chinese Herbal Decoction Pieces, Changsha 410208, Hunan, China; State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica & Neuroscience Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
8
|
Nagata S, Yamasaki R, Takase EO, Iida K, Watanabe M, Masaki K, Wijering MHC, Yamaguchi H, Kira JI, Isobe N. Iguratimod Ameliorates the Severity of Secondary Progressive Multiple Sclerosis in Model Mice by Directly Inhibiting IL-6 Production and Th17 Cell Migration via Mitigation of Glial Inflammation. BIOLOGY 2023; 12:1217. [PMID: 37759616 PMCID: PMC10525689 DOI: 10.3390/biology12091217] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Revised: 08/30/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
We previously reported a novel secondary progressive multiple sclerosis (SPMS) model, progressive experimental autoimmune encephalomyelitis (pEAE), in oligodendroglia-specific Cx47-inducible conditional knockout (Cx47 icKO) mice. Based on our prior study showing the efficacy of iguratimod (IGU), an antirheumatic drug, for acute EAE treatment, we aimed to elucidate the effect of IGU on the SPMS animal model. We induced pEAE by immunizing Cx47 icKO mice with myelin oligodendrocyte glycoprotein peptide 35-55. IGU was orally administered from 17 to 50 days post-immunization. We also prepared a primary mixed glial cell culture and measured cytokine levels in the culture supernatant after stimulation with designated cytokines (IL-1α, C1q, TNF-α) and lipopolysaccharide. A migration assay was performed to evaluate the effect of IGU on the migration ability of T cells toward mixed glial cell cultures. IGU treatment ameliorated the clinical signs of pEAE, decreased the demyelinated area, and attenuated glial inflammation on immunohistochemical analysis. Additionally, IGU decreased the intrathecal IL-6 level and infiltrating Th17 cells. The migration assay revealed reduced Th17 cell migration and IL-6 levels in the culture supernatant after IGU treatment. Collectively, IGU successfully mitigated the clinical signs of pEAE by suppressing Th17 migration through inhibition of IL-6 production by proinflammatory-activated glial cells.
Collapse
Affiliation(s)
- Satoshi Nagata
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Ezgi Ozdemir Takase
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Kotaro Iida
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Mitsuru Watanabe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Marion Heleen Cathérine Wijering
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- Section Molecular Neurobiology, Department of Biomedical Sciences of Cells & Systems, University of Groningen, University Medical Center Groningen (UMCG), MS Center Noord Nederland, 9713 AV Groningen, The Netherlands
| | - Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
- School of Physical Therapy, Faculty of Rehabilitation, Reiwa Health Sciences University, Fukuoka 811-0213, Japan
| | - Jun-ichi Kira
- Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, Fukuoka 831-8501, Japan
- Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, Fukuoka 810-0022, Japan
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
9
|
Ambati R, Kho LK, Prentice D, Thompson A. Osmotic demyelination syndrome: novel risk factors and proposed pathophysiology. Intern Med J 2023; 53:1154-1162. [PMID: 35717664 DOI: 10.1111/imj.15855] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 04/30/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Osmotic demyelination syndrome (ODS) is non-inflammatory demyelination in response to an osmotic challenge. It can be pontine or extrapontine in presentation. AIMS To retrospectively review cases involving ODS and define the spectrum of causes, risk factors, clinical and radiological presentations, and functional outcomes. RESULTS The study utilised data from 15 patients with a mean age of 53.6 years. Malnutrition (9; 60%) and chronic alcoholism (10; 66.7%) were the most common associated disorders. Two (13.3%) patients had severe hyponatraemia (<120 mmol/L). The average highest single-day change was 5.1 mmol/L. Radiologically, 14 (93.3%) had pontine and 6 (40%) had extra-pontine lesions. Hypokalaemia (14; 93.3%) and hypophosphataemia (9; 60%) were commonly associated. Common clinical manifestations include altered consciousness/encephalopathy (9; 60%), dysphagia (4; 26.7%) and limb weakness (4; 26.7%). At 3 months, two (14.3%) had died and six (40%) were functionally independent (modified Rankin scale 0-2). CONCLUSION We found that ODS occurred despite appropriate correction rates of hyponatraemia. Factors such as malnutrition, chronic alcoholism, hypokalaemia and hypophosphataemia are thought to play a role in its pathogenesis. Approximately half of the patients survived and became functionally independent.
Collapse
Affiliation(s)
- Ravi Ambati
- Department of Neurology, Royal Perth Hospital, Perth, Western Australia, Australia
- Department of Neurology, St. John of God Midland Hospital, Perth, Western Australia, Australia
| | - Lay K Kho
- Department of Neurology, Royal Perth Hospital, Perth, Western Australia, Australia
- Department of Neurology, St. John of God Midland Hospital, Perth, Western Australia, Australia
| | - David Prentice
- Department of Neurology, Royal Perth Hospital, Perth, Western Australia, Australia
- Department of Neurology, St. John of God Midland Hospital, Perth, Western Australia, Australia
| | - Andrew Thompson
- Neurological Intervention and Imaging Service of WA (NIISwa), Royal Perth and Sir Charles Gairdner Hospitals, Perth, Western Australia, Australia
| |
Collapse
|
10
|
Luo W, Xu H, Xu L, Jiang W, Chen C, Chang Y, Liu C, Tian Z, Qiu X, Xie C, Li X, Chen H, Lai S, Wu L, Cui Y, Tang C, Qiu W. Remyelination in neuromyelitis optica spectrum disorder is promoted by edaravone through mTORC1 signaling activation. Glia 2023; 71:284-304. [PMID: 36089914 DOI: 10.1002/glia.24271] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 08/15/2022] [Accepted: 08/27/2022] [Indexed: 01/28/2023]
Abstract
Neuromyelitis optica spectrum disorder (NMOSD) is a severe inflammatory autoimmune disease of the central nervous system that is manifested as secondary myelin loss. Oligodendrocyte progenitor cells (OPCs) are the principal source of myelinating oligodendrocytes (OLs) and are abundant in demyelinated regions of NMOSD patients, thus possibly representing a cellular target for pharmacological intervention. To explore the therapeutic compounds that enhance myelination due to endogenous OPCs, we screened the candidate drugs in mouse neural progenitor cell (NPC)-derived OPCs. We identified drug edaravone, which is approved by the Food and Drug Administration (FDA), as a promoter of OPC differentiation into mature OLs. Edaravone enhanced remyelination in organotypic slice cultures and in mice, even when edaravone was administered following NMO-IgG-induced demyelination, and ameliorated motor impairment in a systemic mouse model of NMOSD. The results of mechanistic studies in NMO-IgG-treated mice and the biopsy samples of the brain tissues of NMOSD patients indicated that the mTORC1 signaling pathway was significantly inhibited, and edaravone promoted OPC maturation and remyelination by activating mTORC1 signaling. Furthermore, pharmacological activation of mTORC1 signaling significantly enhanced myelin regeneration in NMOSD. Thus, edaravone is a potential therapeutic agent that promotes lesion repair in NMOSD patients by enhancing OPC maturation.
Collapse
Affiliation(s)
- Wenjing Luo
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Huiming Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Li Xu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Wei Jiang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Chen Chen
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Yanyu Chang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Chunxin Liu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Zhenming Tian
- Department of Spine Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Xiusheng Qiu
- Vaccine Research Institute, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Chichu Xie
- Department of Clinical Immunology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Xuejia Li
- Guangzhou SALIAI Stem Cell Science and Technology Co., Ltd., Guangdong Saliai Stem Cell Research Institute, Guangzhou, Guangdong Province, China
| | - Haijia Chen
- Guangzhou SALIAI Stem Cell Science and Technology Co., Ltd., Guangdong Saliai Stem Cell Research Institute, Guangzhou, Guangdong Province, China
| | - Shuiqing Lai
- Department of Endocrinology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, Guangdong Province, China
| | - Longjun Wu
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | - Yaxiong Cui
- State Key Laboratory of Membrane Biology, Tsinghua-Peking Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Advanced Innovation Center for Structural Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Changyong Tang
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| | - Wei Qiu
- Department of Neurology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong Province, China
| |
Collapse
|
11
|
Xue H, Wu M, Wang Y, Zhao Y, Zhang M, Zhang H. The circadian rhythms regulated by Cx43-signaling in the pathogenesis of Neuromyelitis Optica. Front Immunol 2023; 13:1021703. [PMID: 36726988 PMCID: PMC9885795 DOI: 10.3389/fimmu.2022.1021703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 12/29/2022] [Indexed: 01/18/2023] Open
Abstract
Introduction Neuromyelitis Optica (NMO) is an inflammatory demyelinating disease of the central nervous system (CNS). NMO manifests as selective and severe attacks on axons and myelin of the optic nerve and spinal cord, resulting in necrotic cavities. The circadian rhythms are well demonstrated to profoundly impact cellular function, behavior, and disease. This study is aimed to explore the role and molecular basis of circadian rhythms in NMO. Methods We used an Aquaporin 4(AQP4) IgG-induced NMO cell model in isolated astrocytes. The expression of Cx43 and Bmal1 were detected by real-time PCR and Western Blot. TAT-Gap19 and DQP-1105 were used to inhibit Cx43 and glutamate receptor respectively. The knockdown of Bmal1 were performed with the shRNA containing adenovirus. The levels of glutamate, anterior visual pathway (AVP), and vasoactive intestinal peptide (VIP) were quantified by ELISA kits. Results We found that Bmal1 and Clock, two essential components of the circadian clock, were significantly decreased in NMO astrocytes, which were reversed by Cx43 activation (linoleic acid) or glutamate. Moreover, the expression levels of Bmal1 and Clock were also decreased by Cx43 blockade (TAT-Gap19) or glutamate receptor inhibition (DQP-1105). Furthermore, adenovirus-mediated Bmal1 knockdown by shRNA (Ad-sh-Bmal1) dramatically decreased the levels of glutamate, AVP, and VIP from neurons, and significantly down-regulated the protein level of Cx43 in NMO astrocytes with Cx43 activation (linoleic acid) or glutamate treatment. However, Bmal1 knockdown did not alter these levels in normal astrocytes with Cx43 blockade (TAT-Gap19) or glutamate receptor inhibition (DQP-1105). Discussion Collectively, these results suggest that Cx43-glutamate signaling would be a critical upstream regulator that contributes to the NMO-induced rhythmic damage in SCN astrocytes.
Collapse
Affiliation(s)
- Huiru Xue
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China,First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Minghui Wu
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yongle Wang
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yunfei Zhao
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Meini Zhang
- Department of Neurology, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China,*Correspondence: Meini Zhang, ; Hui Zhang,
| | - Hui Zhang
- First Clinical Medical College of Shanxi Medical University, Taiyuan, Shanxi, China,Department of Medical Imaging, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China,*Correspondence: Meini Zhang, ; Hui Zhang,
| |
Collapse
|
12
|
Nishimura Y, Masaki K, Matsuse D, Yamaguchi H, Tanaka T, Matsuo E, Hayashida S, Watanabe M, Matsushita T, Sadashima S, Sasagasako N, Yamasaki R, Isobe N, Iwaki T, Kira J. Early and extensive alterations of glial connexins, distal oligodendrogliopathy type demyelination, and nodal/paranodal pathology are characteristic of multiple system atrophy. Brain Pathol 2022; 33:e13131. [PMID: 36368713 PMCID: PMC10154368 DOI: 10.1111/bpa.13131] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 10/24/2022] [Indexed: 11/13/2022] Open
Abstract
The pathological hallmark of multiple system atrophy (MSA) is aberrant accumulation of phosphorylated α-synuclein in oligodendrocytes, forming glial cytoplasmic inclusions (GCIs). Extensive demyelination occurs particularly in the olivopontocerebellar and striatonigral pathways, but its precise mechanism remains elusive. Glial connexins (Cxs), which form gap junction channels between astrocytes and oligodendrocytes, play critical roles in myelin maintenance, and have not been studied in MSA. Therefore, we immunohistochemically investigated glial Cx changes in the cerebellar afferent fibers in 15 autopsied patients with MSA. We classified demyelinating lesions into three stages based on Klüver-Barrera staining: early (Stage I), intermediate (Stage II), and late (Stage III) stages showing subtle, moderate, and severe myelin reduction, respectively. Myelin-associated glycoprotein, but not myelin oligodendrocyte glycoprotein, was preferentially decreased in Stage I, suggesting distal oligodendrogliopathy type demyelination. Accumulation of phosphorylated α-synuclein in oligodendrocytes was frequently seen in Stage I but less frequently observed in Stages II and III. Tubulin polymerization-promoting protein (TPPP/p25α)-positive oligodendrocytes were preserved in Stage I but successively decreased in Stages II and III. Even at Stage I, Cx32 was nearly absent from myelin, despite the relative preservation of other nodal proteins, such as neurofascin, claudin-11/oligodendrocyte-specific protein, and contactin-associated protein 1, which successively decreased in the later stages. Cx32 was re-distributed in the oligodendrocyte cytoplasm and co-localized with GCIs. Cx47 gradually decreased at the oligodendrocyte surface in a stage-dependent manner but was not co-localized with GCIs. Astrocytic Cx43 was down-regulated in Stage I but up-regulated in Stages II and III, reflecting astrogliosis. Cx43/Cx47 gap junctions significantly decreased from Stage I to III. Activated microglia/macrophages and T cells infiltrated in Stage I rather than Stages II and III. Therefore, early and extensive alterations of glial Cxs, particularly Cx32 loss, occur in MSA and may accelerate distal oligodendrogliopathy type demyelination and nodal/paranodal dysfunction through disruption of inter-glial communication.
Collapse
Affiliation(s)
- Yuji Nishimura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Dai Matsuse
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Tatsunori Tanaka
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
- Sumitomo Pharma Osaka Japan
| | - Eriko Matsuo
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Shotaro Hayashida
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Mitsuru Watanabe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Takuya Matsushita
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Shoko Sadashima
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
- Department of Neuropathology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Naokazu Sasagasako
- Department of Neurology, Neuro‐Muscular Center National Omuta Hospital Fukuoka Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Toru Iwaki
- Department of Neuropathology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
| | - Jun‐ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences Kyushu University Fukuoka Japan
- Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka International University of Health and Welfare Ookawa Japan
- Department of Neurology, Brain and Nerve Center Fukuoka Central Hospital Fukuoka Japan
| |
Collapse
|
13
|
Ma W, Huang C, Yang L, Luo J. MRI findings of autoimmune glial fibrillary acidic protein astrocytopathy involving infratentorial: Case report. Radiol Case Rep 2022; 17:2515-2518. [PMID: 35601378 PMCID: PMC9114157 DOI: 10.1016/j.radcr.2022.04.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 10/25/2022] Open
|
14
|
Wang B, Li X, Li H, Xiao L, Zhou Z, Chen K, Gui L, Hou X, Fan R, Chen K, Wu W, Li H, Hu X. Clinical, Radiological and Pathological Characteristics Between Cerebral Small Vessel Disease and Multiple Sclerosis: A Review. Front Neurol 2022; 13:841521. [PMID: 35812110 PMCID: PMC9263123 DOI: 10.3389/fneur.2022.841521] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/19/2022] [Indexed: 11/13/2022] Open
Abstract
Cerebral small vessel disease (CSVD) and multiple sclerosis (MS) are a group of diseases associated with small vessel lesions, the former often resulting from the vascular lesion itself, while the latter originating from demyelinating which can damage the cerebral small veins. Clinically, CSVD and MS do not have specific signs and symptoms, and it is often difficult to distinguish between the two from the aspects of the pathology and imaging. Therefore, failure to correctly identify and diagnose the two diseases will delay early intervention, which in turn will affect the long-term functional activity for patients and even increase their burden of life. This review has summarized recent studies regarding their similarities and difference of the clinical manifestations, pathological features and imaging changes in CSVD and MS, which could provide a reliable basis for the diagnosis and differentiation of the two diseases in the future.
Collapse
Affiliation(s)
- Bijia Wang
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xuegang Li
- Department of Neurosurgery, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Haoyi Li
- Department of Neurosurgery, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Li Xiao
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Zhenhua Zhou
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Kangning Chen
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Li Gui
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xianhua Hou
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Rong Fan
- Department of Neurology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Kang Chen
- Department of Radiology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wenjing Wu
- Department of Radiology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Haitao Li
- Department of Radiology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- *Correspondence: Haitao Li
| | - Xiaofei Hu
- Department of Radiology, First Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- Xiaofei Hu
| |
Collapse
|
15
|
Cacciaguerra L, Carotenuto A, Pagani E, Mistri D, Radaelli M, Martinelli V, Filippi M, Rocca MA. MRI EVALUATION OF PERIVASCULAR SPACE ABNORMALITIES IN NEUROMYELITIS OPTICA. Ann Neurol 2022; 92:173-183. [PMID: 35596582 PMCID: PMC9544484 DOI: 10.1002/ana.26419] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/19/2022] [Accepted: 05/19/2022] [Indexed: 11/08/2022]
Abstract
Objective Astrocytes outline the perivascular space (PVS) and regulate fluid exchange through the aquaporin‐4 water channel. As neuromyelitis optica is an autoimmune astrocytopathy targeting aquaporin‐4, we hypothesized that it could be associatied with PVS abnormalities. Methods A total of 34 patients, and 46 age‐ and sex‐matched healthy controls from two independent cohorts (exploratory and validation dataset) underwent a standardized 3.0‐T magnetic resonance imaging protocol including conventional and diffusion tensor imaging. Susceptibility‐weighted imaging was also acquired in the exploratory dataset. We evaluated macroscopic and microstructural abnormalities of PVS in terms of enlargement and water diffusivity (DTI‐ALPS index). In the exploration dataset, a susceptibility‐weighted sequence was used to draw the regions of interest for the DTI‐ALPS index calculation in areas having veins perpendicular to lateral ventricles. Between‐group comparisons, correlations, and regression models were run to assess associations between PVS abnormalities, and clinical and magnetic resonance imaging variables. Results Patients had a higher frequency of severe PVS enlargement in the centrum semiovale (29.4% vs 8.7%), which correlated with brain atrophy, deep grey matter atrophy, and poorer cognitive performance (r‐values range: −0.44, −0.36; p values: 0.01–0.046). In both datasets, patients had reduced DTI‐ALPS index compared with controls (p values 0.004–0.038). Lower DTI‐ALPS index, deep gray matter volume, and cortical volume could discriminate between patients and controls (R2 = 0.62), whereas lower DTI‐ALPS index, higher number of myelitis, and higher T2‐lesion volume were associated with worse disability (R2 = 0.55). Interpretation Patients with neuromyelitis optica spectrum disorder are characterized by abnormal enlargement and impaired water diffusion along the PVS, whose clinical implications suggest a direct correlation with disease pathogenesis and severity. ANN NEUROL 2022;92:173–183
Collapse
Affiliation(s)
- Laura Cacciaguerra
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Antonio Carotenuto
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisabetta Pagani
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Damiano Mistri
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marta Radaelli
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Massimo Filippi
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurorehabilitation Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurophysiology Service, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| | - Maria A Rocca
- Neuroimaging Research Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy.,Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
16
|
Kunkl M, Amormino C, Tedeschi V, Fiorillo MT, Tuosto L. Astrocytes and Inflammatory T Helper Cells: A Dangerous Liaison in Multiple Sclerosis. Front Immunol 2022; 13:824411. [PMID: 35211120 PMCID: PMC8860818 DOI: 10.3389/fimmu.2022.824411] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 01/13/2022] [Indexed: 11/15/2022] Open
Abstract
Multiple Sclerosis (MS) is a neurodegenerative autoimmune disorder of the central nervous system (CNS) characterized by the recruitment of self-reactive T lymphocytes, mainly inflammatory T helper (Th) cell subsets. Once recruited within the CNS, inflammatory Th cells produce several inflammatory cytokines and chemokines that activate resident glial cells, thus contributing to the breakdown of blood-brain barrier (BBB), demyelination and axonal loss. Astrocytes are recognized as key players of MS immunopathology, which respond to Th cell-defining cytokines by acquiring a reactive phenotype that amplify neuroinflammation into the CNS and contribute to MS progression. In this review, we summarize current knowledge of the astrocytic changes and behaviour in both MS and experimental autoimmune encephalomyelitis (EAE), and the contribution of pathogenic Th1, Th17 and Th1-like Th17 cell subsets, and CD8+ T cells to the morphological and functional modifications occurring in astrocytes and their pathological outcomes.
Collapse
Affiliation(s)
- Martina Kunkl
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Carola Amormino
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| | - Valentina Tedeschi
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Maria Teresa Fiorillo
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy
| | - Loretta Tuosto
- Department of Biology and Biotechnology Charles Darwin, Sapienza University, Rome, Italy.,Laboratory Affiliated to Istituto Pasteur Italia-Fondazione Cenci Bolognetti, Sapienza University, Rome, Italy
| |
Collapse
|
17
|
Kim H, Lee EJ, Lim YM, Kim KK. Glial Fibrillary Acidic Protein in Blood as a Disease Biomarker of Neuromyelitis Optica Spectrum Disorders. Front Neurol 2022; 13:865730. [PMID: 35370870 PMCID: PMC8968934 DOI: 10.3389/fneur.2022.865730] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
Glial fibrillary acidic protein (GFAP) is a type III intermediate filament protein found in astrocytes in the brain. Damaged astrocytes release GFAP into cerebrospinal fluid and blood. Thus, GFAP levels in these body fluids may reflect the disease state of neuromyelitis optica spectrum disorder (NMOSD), which includes astrocytopathy, characterized by pathogenic antibodies against aquaporin 4 located on astrocytes. Recently, single-molecule array technology that can detect these synaptic proteins in blood, even in the subfemtomolar range, has been developed. Emerging evidence suggests that GFAP protein is a strong biomarker candidate for NMOSD. This mini-review provides basic information about GFAP protein and innovative clinical data that show the potential clinical value of blood GFAP levels as a biomarker for NMOSD.
Collapse
Affiliation(s)
- Hyunjin Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Eun-Jae Lee
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Department of Medicine, Asan Medical Institute of Convergence Science and Technology, Seoul, South Korea
| | - Young-Min Lim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Kwang-Kuk Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
18
|
Vakrakou AG, Brinia ME, Svolaki I, Argyrakos T, Stefanis L, Kilidireas C. Immunopathology of Tumefactive Demyelinating Lesions-From Idiopathic to Drug-Related Cases. Front Neurol 2022; 13:868525. [PMID: 35418930 PMCID: PMC8997292 DOI: 10.3389/fneur.2022.868525] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Accepted: 02/18/2022] [Indexed: 11/13/2022] Open
Abstract
Tumefactive demyelinating lesions (TDL) represent a diagnostic dilemma for clinicians, and in rare atypical cases a collaboration of a neuroradiologist, a neurologist, and a neuropathologist is warranted for accurate diagnosis. Recent advances in neuropathology have shown that TDL represent an umbrella under which many different diagnostic entities can be responsible. TDL can emerge not only as part of the spectrum of classic multiple sclerosis (MS) but also can represent an idiopathic monophasic disease, a relapsing disease with recurrent TDL, or could be part of the myelin oligodendrocyte glycoprotein (MOG)- and aquaporin-4 (AQP4)-associated disease. TDL can appear during the MS disease course, and increasingly cases arise showing an association with specific drug interventions. Although TDL share common features with classic MS lesions, they display some unique features, such as extensive and widespread demyelination, massive and intense parenchymal infiltration by macrophages along with lymphocytes (mainly T but also B cells), dystrophic changes in astrocytes, and the presence of Creutzfeldt cells. This article reviews the existent literature regarding the neuropathological findings of tumefactive demyelination in various disease processes to better facilitate the identification of disease signatures. Recent developments in immunopathology of central nervous system disease suggest that specific pathological immune features (type of demyelination, infiltrating cell type distribution, specific astrocyte pathology and complement deposition) can differentiate tumefactive lesions arising as part of MS, MOG-associated disease, and AQP4 antibody-positive neuromyelitis optica spectrum disorder. Lessons from immunopathology will help us not only stratify these lesions in disease entities but also to better organize treatment strategies. Improved advances in tissue biomarkers should pave the way for prompt and accurate diagnosis of TDL leading to better outcomes for patients.
Collapse
Affiliation(s)
- Aigli G. Vakrakou
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Maria-Evgenia Brinia
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Svolaki
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Leonidas Stefanis
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Constantinos Kilidireas
- Demyelinating Diseases Unit, 1st Department of Neurology, School of Medicine, Aeginition Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
19
|
Tittarelli A. Connexin channels modulation in pathophysiology and treatment of immune and inflammatory disorders. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166258. [PMID: 34450245 DOI: 10.1016/j.bbadis.2021.166258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/28/2021] [Accepted: 08/19/2021] [Indexed: 12/16/2022]
Abstract
Connexin-mediated intercellular communication mechanisms include bidirectional cell-to-cell coupling by gap junctions and release/influx of molecules by hemichannels. These intercellular communications have relevant roles in numerous immune system activities. Here, we review the current knowledge about the function of connexin channels, mainly those formed by connexin-43, on immunity and inflammation. Focusing on those evidence that support the design and development of therapeutic tools to modulate connexin expression and/or channel activities with treatment potential for infections, wounds, cancer, and other inflammatory conditions.
Collapse
Affiliation(s)
- Andrés Tittarelli
- Programa Institucional de Fomento a la Investigación, Desarrollo e Innovación, Universidad Tecnológica Metropolitana, Santiago 8940577, Chile.
| |
Collapse
|
20
|
Abstract
Fifty years have passed since the discovery of glial fibrillary acidic protein (GFAP) by Lawrence Eng and colleagues. Now recognized as a member of the intermediate filament family of proteins, it has become a subject for study in fields as diverse as structural biology, cell biology, gene expression, basic neuroscience, clinical genetics and gene therapy. This review covers each of these areas, presenting an overview of current understanding and controversies regarding GFAP with the goal of stimulating continued study of this fascinating protein.
Collapse
Affiliation(s)
- Albee Messing
- Waisman Center, University of Wisconsin-Madison.,Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison
| | - Michael Brenner
- Department of Neurobiology, University of Alabama-Birmingham
| |
Collapse
|
21
|
Understanding Abnormal c-JNK/p38MAPK Signaling Overactivation Involved in the Progression of Multiple Sclerosis: Possible Therapeutic Targets and Impact on Neurodegenerative Diseases. Neurotox Res 2021; 39:1630-1650. [PMID: 34432262 DOI: 10.1007/s12640-021-00401-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 08/01/2021] [Accepted: 08/09/2021] [Indexed: 12/16/2022]
Abstract
Demyelination, immune dysregulation, and neuroinflammation are the most common triggers of motor neuron disorders such as multiple sclerosis (MS). MS is a chronic demyelinating neurodegenerative disease of the central nervous system caused by abnormal immune activation, which causes myelin sheath damage. Cell signal transduction pathways are required for a variety of physiological and pathological processes in the brain. When these signaling systems become overactive, they can lead to disease progression. In various physiological conditions, abnormal mitogen-activated protein kinase (MAPK) activation is associated with several physiological dysfunctions that cause neurodegeneration. Previous research indicates that c-JNK and p38MAPK signaling play critical roles in neuronal growth and differentiation. c-JNK/p38MAPK is a member of the MAPK family, which regulates metabolic pathways, cell proliferation, differentiation, and apoptosis that control certain neurological activities. During brain injuries, c-JNK/p38MAPK also affects neuronal elastic properties, nerve growth, and cognitive processing. This review systematically linked abnormal c-JNK/p38MAPK signaling activation to multiple neuropathological pathways in MS and related neurological dysfunctions. MS progression is linked to genetic defects, oligodendrocyte destruction, glial overactivation, and immune dysregulation. We concluded that inhibiting both the c-JNK/p38MAPK signaling pathways can promote neuroprotection and neurotrophic effects against the clinical-pathological presentation of MS and influence other neurological disorders. As a result, the potential benefits of c-JNK/p38MAPK downregulation for the development of disease-modifying treatment interventions in the future could include MS prevention and related neurocomplications.
Collapse
|
22
|
Moulson AJ, Squair JW, Franklin RJM, Tetzlaff W, Assinck P. Diversity of Reactive Astrogliosis in CNS Pathology: Heterogeneity or Plasticity? Front Cell Neurosci 2021; 15:703810. [PMID: 34381334 PMCID: PMC8349991 DOI: 10.3389/fncel.2021.703810] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 07/02/2021] [Indexed: 01/02/2023] Open
Abstract
Astrocytes are essential for the development and homeostatic maintenance of the central nervous system (CNS). They are also critical players in the CNS injury response during which they undergo a process referred to as "reactive astrogliosis." Diversity in astrocyte morphology and gene expression, as revealed by transcriptional analysis, is well-recognized and has been reported in several CNS pathologies, including ischemic stroke, CNS demyelination, and traumatic injury. This diversity appears unique to the specific pathology, with significant variance across temporal, topographical, age, and sex-specific variables. Despite this, there is limited functional data corroborating this diversity. Furthermore, as reactive astrocytes display significant environmental-dependent plasticity and fate-mapping data on astrocyte subsets in the adult CNS is limited, it remains unclear whether this diversity represents heterogeneity or plasticity. As astrocytes are important for neuronal survival and CNS function post-injury, establishing to what extent this diversity reflects distinct established heterogeneous astrocyte subpopulations vs. environmentally dependent plasticity within established astrocyte subsets will be critical for guiding therapeutic development. To that end, we review the current state of knowledge on astrocyte diversity in the context of three representative CNS pathologies: ischemic stroke, demyelination, and traumatic injury, with the goal of identifying key limitations in our current knowledge and suggesting future areas of research needed to address them. We suggest that the majority of identified astrocyte diversity in CNS pathologies to date represents plasticity in response to dynamically changing post-injury environments as opposed to heterogeneity, an important consideration for the understanding of disease pathogenesis and the development of therapeutic interventions.
Collapse
Affiliation(s)
- Aaron J. Moulson
- Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
| | - Jordan W. Squair
- Department of Clinical Neuroscience, Faculty of Life Sciences, Center for Neuroprosthetics and Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), NeuroRestore, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Robin J. M. Franklin
- Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Wolfram Tetzlaff
- International Collaboration on Repair Discoveries (ICORD), Vancouver, BC, Canada
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
- Department of Surgery, University of British Columbia, Vancouver, BC, Canada
| | - Peggy Assinck
- Wellcome Trust - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Centre for Regenerative Medicine, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
23
|
Une H, Yamasaki R, Nagata S, Yamaguchi H, Nakamuta Y, Indiasari UC, Cui Y, Shinoda K, Masaki K, Götz M, Kira JI. Brain gray matter astroglia-specific connexin 43 ablation attenuates spinal cord inflammatory demyelination. J Neuroinflammation 2021; 18:126. [PMID: 34090477 PMCID: PMC8180177 DOI: 10.1186/s12974-021-02176-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 05/19/2021] [Indexed: 11/10/2022] Open
Abstract
Background Brain astroglia are activated preceding the onset of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We characterized the effects of brain astroglia on spinal cord inflammation, focusing on astroglial connexin (Cx)43, because we recently reported that Cx43 has a critical role in regulating neuroinflammation. Methods Because glutamate aspartate transporter (GLAST)+ astroglia are enriched in the brain gray matter, we generated Cx43fl/fl;GLAST-CreERT2/+ mice that were brain gray matter astroglia-specific Cx43 conditional knockouts (Cx43 icKO). EAE was induced by immunization with myelin oligodendroglia glycoprotein (MOG) 35–55 peptide 10 days after tamoxifen injection. Cx43fl/fl mice were used as controls. Results Acute and chronic EAE signs were significantly milder in Cx43 icKO mice than in controls whereas splenocyte MOG-specific responses were unaltered. Histologically, Cx43 icKO mice showed significantly less demyelination and fewer CD45+ infiltrating immunocytes, including F4/80+ macrophages, and Iba1+ microglia in the spinal cord than controls. Microarray analysis of the whole cerebellum revealed marked upregulation of anti-inflammatory A2-specific astroglia gene sets in the pre-immunized phase and decreased proinflammatory A1-specific and pan-reactive astroglial gene expression in the onset phase in Cx43 icKO mice compared with controls. Astroglia expressing C3, a representative A1 marker, were significantly decreased in the cerebrum, cerebellum, and spinal cord of Cx43 icKO mice compared with controls in the peak phase. Isolated Cx43 icKO spinal microglia showed more anti-inflammatory and less proinflammatory gene expression than control microglia in the pre-immunized phase. In particular, microglial expression of Ccl2, Ccl5, Ccl7, and Ccl8 in the pre-immunized phase and of Cxcl9 at the peak phase was lower in Cx43 icKO than in controls. Spinal microglia circularity was significantly lower in Cx43 icKO than in controls in the peak phase. Significantly lower interleukin (IL)-6, interferon-γ, and IL-10 levels were present in cerebrospinal fluid from Cx43 icKO mice in the onset phase compared with controls. Conclusions The ablation of Cx43 in brain gray matter astroglia attenuates EAE by promoting astroglia toward an anti-inflammatory phenotype and suppressing proinflammatory activation of spinal microglia partly through depressed cerebrospinal fluid proinflammatory cytokine/chemokine levels. Brain astroglial Cx43 might be a novel therapeutic target for MS. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02176-1.
Collapse
Affiliation(s)
- Hayato Une
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Satoshi Nagata
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yuko Nakamuta
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ulfa Camelia Indiasari
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yiwen Cui
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Koji Shinoda
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Magdalena Götz
- Physiological Genomics, Biomedical Center, Ludwig-Maximilians University Munich, Munich, Germany
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, 3-1-1, Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan. .,Translational Neuroscience Center, Graduate School of Medicine, and School of Pharmacy at Fukuoka, International University of Health and Welfare, 137-1 Enokizu, Ookawa, Fukuoka, 831-8501, Japan. .,Department of Neurology, Brain and Nerve Center, Fukuoka Central Hospital, International University of Health and Welfare, 2-6-11 Yakuin, Chuo-ku, Fukuoka, 810-0022, Japan.
| |
Collapse
|
24
|
Astrocytes in Multiple Sclerosis-Essential Constituents with Diverse Multifaceted Functions. Int J Mol Sci 2021; 22:ijms22115904. [PMID: 34072790 PMCID: PMC8198285 DOI: 10.3390/ijms22115904] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/24/2021] [Accepted: 05/25/2021] [Indexed: 11/19/2022] Open
Abstract
In multiple sclerosis (MS), astrocytes respond to the inflammatory stimulation with an early robust process of morphological, transcriptional, biochemical, and functional remodeling. Recent studies utilizing novel technologies in samples from MS patients, and in an animal model of MS, experimental autoimmune encephalomyelitis (EAE), exposed the detrimental and the beneficial, in part contradictory, functions of this heterogeneous cell population. In this review, we summarize the various roles of astrocytes in recruiting immune cells to lesion sites, engendering the inflammatory loop, and inflicting tissue damage. The roles of astrocytes in suppressing excessive inflammation and promoting neuroprotection and repair processes is also discussed. The pivotal roles played by astrocytes make them an attractive therapeutic target. Improved understanding of astrocyte function and diversity, and the mechanisms by which they are regulated may lead to the development of novel approaches to selectively block astrocytic detrimental responses and/or enhance their protective properties.
Collapse
|
25
|
Gómez-Pinedo U, García-Ávila Y, Gallego-Villarejo L, Matías-Guiu JA, Benito-Martín MS, Esteban-García N, Sanclemente-Alamán I, Pytel V, Moreno-Jiménez L, Sancho-Bielsa F, Vidorreta-Ballesteros L, Montero-Escribano P, Matías-Guiu J. Sera from Patients with NMOSD Reduce the Differentiation Capacity of Precursor Cells in the Central Nervous System. Int J Mol Sci 2021; 22:5192. [PMID: 34068922 PMCID: PMC8155872 DOI: 10.3390/ijms22105192] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/11/2021] [Accepted: 05/11/2021] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION AQP4 (aquaporin-4)-immunoglobulin G (IgG)-mediated neuromyelitis optica spectrum disorder (NMOSD) is an inflammatory demyelinating disease that affects the central nervous system, particularly the spinal cord and optic nerve; remyelination capacity in neuromyelitis optica is yet to be determined, as is the role of AQP4-IgG in cell differentiation. MATERIAL AND METHODS We included three groups-a group of patients with AQP4-IgG-positive neuromyelitis optica, a healthy group, and a sham group. We analyzed differentiation capacity in cultures of neurospheres from the subventricular zone of mice by adding serum at two different times: early and advanced stages of differentiation. We also analyzed differentiation into different cell lines. RESULTS AND CONCLUSIONS The effect of sera from patients with NMOSD on precursor cells differs according to the degree of differentiation, and probably affects oligodendrocyte progenitor cells from NG2 cells to a lesser extent than cells from the subventricular zone; however, the resulting oligodendrocytes may be compromised in terms of maturation and possibly limited in their ability to generate myelin. Furthermore, these cells decrease in number with age. It is very unlikely that the use of drugs favoring the migration and differentiation of oligodendrocyte progenitor cells in multiple sclerosis would be effective in the context of neuromyelitis optica, but cell therapy with oligodendrocyte progenitor cells seems to be a potential alternative.
Collapse
Affiliation(s)
- Ulises Gómez-Pinedo
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| | - Yolanda García-Ávila
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| | - Lucía Gallego-Villarejo
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| | - Jordi A. Matías-Guiu
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| | - María Soledad Benito-Martín
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| | - Noelia Esteban-García
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| | - Inmaculada Sanclemente-Alamán
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| | - Vanesa Pytel
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| | - Lidia Moreno-Jiménez
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| | - Francisco Sancho-Bielsa
- Department of Physiology, Ciudad Real School of Medicine, Universidad de Castilla-La Mancha, 13001 Ciudad Real, Spain;
| | - Lucía Vidorreta-Ballesteros
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| | - Paloma Montero-Escribano
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| | - Jorge Matías-Guiu
- Laboratory of Neurobiology, Department of Neurology, Institute of Neurosciences, San Carlos Health Research Institute, Universidad Complutense, 28040 Madrid, Spain; (Y.G.-Á.); (L.G.-V.); (J.A.M.-G.); (M.S.B.-M.); (N.E.-G.); (I.S.-A.); (V.P.); (L.M.-J.); (L.V.-B.); (P.M.-E.); (J.M.-G.)
| |
Collapse
|
26
|
Richard C, Ruiz A, Cavagna S, Bigotte M, Vukusic S, Masaki K, Suenaga T, Kira JI, Giraudon P, Marignier R. Connexins in neuromyelitis optica: a link between astrocytopathy and demyelination. Brain 2021; 143:2721-2732. [PMID: 32889550 DOI: 10.1093/brain/awaa227] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/01/2020] [Accepted: 05/06/2020] [Indexed: 01/26/2023] Open
Abstract
Neuromyelitis optica, a rare neuroinflammatory demyelinating disease of the CNS, is characterized by the presence of specific pathogenic autoantibodies directed against the astrocytic water channel aquaporin 4 (AQP4) and is now considered as an astrocytopathy associated either with complement-dependent astrocyte death or with astrocyte dysfunction. However, the link between astrocyte dysfunction and demyelination remains unclear. We propose glial intercellular communication, supported by connexin hemichannels and gap junctions, to be involved in demyelination process in neuromyelitis optica. Using mature myelinated cultures, we demonstrate that a treatment of 1 h to 48 h with immunoglobulins purified from patients with neuromyelitis optica (NMO-IgG) is responsible for a complement independent demyelination, compared to healthy donors' immunoglobulins (P < 0.001). In parallel, patients' immunoglobulins induce an alteration of connexin expression characterized by a rapid loss of astrocytic connexins at the membrane followed by an increased size of gap junction plaques (+60%; P < 0.01). This was co-observed with connexin dysfunction with gap junction disruption (-57%; P < 0.001) and increased hemichannel opening (+17%; P < 0.001), associated with glutamate release. Blocking connexin 43 hemichannels with a specific peptide was able to prevent demyelination in co-treatment with patients compared to healthy donors' immunoglobulins. By contrast, the blockade of connexin 43 gap junctions with another peptide was detrimental for myelin (myelin density -48%; P < 0.001). Overall, our results suggest that dysregulation of connexins would play a pathogenetic role in neuromyelitis optica. The further identification of mechanisms leading to connexin dysfunction and soluble factors implicated, would provide interesting therapeutic strategies for demyelinating disorders.
Collapse
Affiliation(s)
- Chloé Richard
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Anne Ruiz
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Sylvie Cavagna
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Maxime Bigotte
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Sandra Vukusic
- Service de neurologie, sclérose en plaques, pathologies de la myéline et neuro-inflammation, Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, Lyon, France.,Centre de référence des maladies inflammatoires rares du cerveau et de la moelle, Lyon, France
| | - Katsuhisa Masaki
- Department of Neurology, Neurological institute, Graduate School of Medical Sciences, Kyushu University
| | | | - Jun-Ichi Kira
- Department of Neurology, Neurological institute, Graduate School of Medical Sciences, Kyushu University
| | - Pascale Giraudon
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France
| | - Romain Marignier
- INSERM U1028, CNRS UMR 5292, Lyon1 University, Center for Research in Neuroscience of Lyon, Lyon, France.,Service de neurologie, sclérose en plaques, pathologies de la myéline et neuro-inflammation, Hôpital Neurologique Pierre Wertheimer Hospices Civils de Lyon, Lyon, France.,Centre de référence des maladies inflammatoires rares du cerveau et de la moelle, Lyon, France
| |
Collapse
|
27
|
Mozafari S, Deboux C, Laterza C, Ehrlich M, Kuhlmann T, Martino G, Baron-Van Evercooren A. Beneficial contribution of induced pluripotent stem cell-progeny to Connexin 47 dynamics during demyelination-remyelination. Glia 2020; 69:1094-1109. [PMID: 33301181 PMCID: PMC7984339 DOI: 10.1002/glia.23950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 11/26/2020] [Accepted: 11/30/2020] [Indexed: 12/17/2022]
Abstract
Oligodendrocytes are extensively coupled to astrocytes, a phenomenon ensuring glial homeostasis and maintenance of central nervous system myelin. Molecular disruption of this communication occurs in demyelinating diseases such as multiple sclerosis. Less is known about the vulnerability and reconstruction of the panglial network during adult demyelination‐remyelination. Here, we took advantage of lysolcithin‐induced demyelination to investigate the expression dynamics of the oligodendrocyte specific connexin 47 (Cx47) and to some extent that of astrocyte Cx43, and whether this dynamic could be modulated by grafted induced pluripotent stem cell (iPSC)‐neural progeny. Our data show that disruption of Cx43‐Cx47 mediated hetero‐cellular gap‐junction intercellular communication following demyelination is larger in size than demyelination. Loss of Cx47 expression is timely rescued during remyelination and accelerated by the grafted neural precursors. Moreover, mouse and human iPSC‐derived oligodendrocytes express Cx47, which co‐labels with astrocyte Cx43, indicating their integration into the panglial network. These data suggest that in rodents, full lesion repair following transplantation occurs by panglial reconstruction in addition to remyelination. Targeting panglial elements by cell therapy or pharmacological compounds may help accelerating or stabilizing re/myelination in myelin disorders.
Collapse
Affiliation(s)
- Sabah Mozafari
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| | - Cyrille Deboux
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| | - Cecilia Laterza
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy.,Industrial Engineering Department, University of Padova, Padova, Italy
| | - Marc Ehrlich
- Department of Cell and Developmental Biology, Max Planck Institute for Molecular Biomedicine, Münster, Germany.,Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster, Germany
| | - Gianvito Martino
- Institute of Experimental Neurology-DIBIT 2, Division of Neuroscience, IRCCS San Raffaele Hospital and Vita San Raffaele University, Milan, Italy
| | - Anne Baron-Van Evercooren
- INSERM, U1127, Paris, France.,CNRS, UMR 7225, Paris, France.,Sorbonne Université UPMC Paris 06, UM-75, Paris, France.,ICM-GH Pitié-Salpêtrière, Paris, France
| |
Collapse
|
28
|
Bozic I, Savic D, Lavrnja I. Astrocyte phenotypes: Emphasis on potential markers in neuroinflammation. Histol Histopathol 2020; 36:267-290. [PMID: 33226087 DOI: 10.14670/hh-18-284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Astrocytes, the most abundant glial cells in the central nervous system (CNS), have numerous integral roles in all CNS functions. They are essential for synaptic transmission and support neurons by providing metabolic substrates, secreting growth factors and regulating extracellular concentrations of ions and neurotransmitters. Astrocytes respond to CNS insults through reactive astrogliosis, in which they go through many functional and molecular changes. In neuroinflammatory conditions reactive astrocytes exert both beneficial and detrimental functions, depending on the context and heterogeneity of astrocytic populations. In this review we profile astrocytic diversity in the context of neuroinflammation; with a specific focus on multiple sclerosis (MS) and its best-described animal model experimental autoimmune encephalomyelitis (EAE). We characterize two main subtypes, protoplasmic and fibrous astrocytes and describe the role of intermediate filaments in the physiology and pathology of these cells. Additionally, we outline a variety of markers that are emerging as important in investigating astrocytic biology in both physiological conditions and neuroinflammation.
Collapse
Affiliation(s)
- Iva Bozic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Danijela Savic
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Irena Lavrnja
- Institute for Biological Research "Sinisa Stankovic", National Institute of Republic of Serbia, University of Belgrade, Belgrade, Serbia.
| |
Collapse
|
29
|
Mozafari S, Baron-Van Evercooren A. Human stem cell-derived oligodendrocytes: From humanized animal models to cell therapy in myelin diseases. Semin Cell Dev Biol 2020; 116:53-61. [PMID: 33082116 DOI: 10.1016/j.semcdb.2020.09.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 09/21/2020] [Accepted: 09/22/2020] [Indexed: 12/15/2022]
Abstract
Oligodendrocytes are main targets in demyelinating and dysmyelinating diseases of the central nervous system (CNS), but are also involved in accidental, neurodegenerative and psychiatric disorders. The underlying pathology of these diseases is not fully understood and treatments are still lacking. The recent discovery of the induced pluripotent stem cell (iPSC) technology has open the possibility to address the biology of human oligodendroglial cells both in the dish and in vivo via engraftment in animal models, and paves the way for the development of treatment for myelin disorders. In this review, we make a short overview of the different sources human oligodendroglial cells, and animal models available for pre-clinical cell therapy. We discuss the anatomical and functional benefit of grafted iPSC-progenitors over their brain counterparts, their use in disease modeling and the missing gaps that still prevent to study their biology in the most integrated way, and to translate iPSC-stem cell based therapy to the clinic.
Collapse
Affiliation(s)
- Sabah Mozafari
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, UMR 7225, Sorbonne Université UM75, F-75013 Paris, France; CNRS, UMR 7225, Paris, France; Sorbonne Universités, Université Pierre et MarieCurie Paris 06, UM-75, Paris, France
| | - Anne Baron-Van Evercooren
- Institut du Cerveau et de la Moelle Epinière-Groupe Hospitalier Pitié-Salpêtrière, INSERM, U1127, CNRS, UMR 7225, Sorbonne Université UM75, F-75013 Paris, France; CNRS, UMR 7225, Paris, France; Sorbonne Universités, Université Pierre et MarieCurie Paris 06, UM-75, Paris, France.
| |
Collapse
|
30
|
Cohen-Salmon M, Slaoui L, Mazaré N, Gilbert A, Oudart M, Alvear-Perez R, Elorza-Vidal X, Chever O, Boulay AC. Astrocytes in the regulation of cerebrovascular functions. Glia 2020; 69:817-841. [PMID: 33058289 DOI: 10.1002/glia.23924] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are the most numerous type of neuroglia in the brain and have a predominant influence on the cerebrovascular system; they control perivascular homeostasis, the integrity of the blood-brain barrier, the dialogue with the peripheral immune system, the transfer of metabolites from the blood, and blood vessel contractility in response to neuronal activity. These regulatory processes occur in a specialized interface composed of perivascular astrocyte extensions that almost completely cover the cerebral blood vessels. Scientists have only recently started to study how this interface is formed and how it influences cerebrovascular functions. Here, we review the literature on the astrocytes' role in the regulation of the cerebrovascular system. We cover the anatomy and development of the gliovascular interface, the known gliovascular functions, and molecular factors, the latter's implication in certain pathophysiological situations, and recent cutting-edge experimental tools developed to examine the astrocytes' role at the vascular interface. Finally, we highlight some open questions in this field of research.
Collapse
Affiliation(s)
- Martine Cohen-Salmon
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Leila Slaoui
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Noémie Mazaré
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Alice Gilbert
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Marc Oudart
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Rodrigo Alvear-Perez
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Xabier Elorza-Vidal
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Oana Chever
- Normandie University, UNIROUEN, INSERM, DC2N, IRIB, Rouen, France
| | - Anne-Cécile Boulay
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| |
Collapse
|
31
|
Angeli S, Kousiappa I, Stavrou M, Sargiannidou I, Georgiou E, Papacostas SS, Kleopa KA. Altered Expression of Glial Gap Junction Proteins Cx43, Cx30, and Cx47 in the 5XFAD Model of Alzheimer's Disease. Front Neurosci 2020; 14:582934. [PMID: 33117125 PMCID: PMC7575794 DOI: 10.3389/fnins.2020.582934] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/14/2020] [Indexed: 11/13/2022] Open
Abstract
Glial gap junction proteins, called connexins (Cxs), form gap junctions in the central nervous system (CNS) to allow the bidirectional cytosolic exchange of molecules between adjacent cells. Their involvement in inheritable diseases and the use of experimental animal models that closely mimic such diseases revealed the critical role of glial GJs in myelination and homeostasis. Cxs are also implicated in acquired demyelinating disorders, such as Multiple Sclerosis (MS) and Alzheimer's disease (AD). Animal and human studies have revealed a role of the astrocytic Cx43 in the progression of AD but the role of Cx47, which is the main partner of Cx43 in the astrocyte-oligodendrocyte GJs is still unknown. The aim of this study was to investigate the astrocytic connexins, Cx43 and Cx30 in relation to oligodendrocytic Cx47 in the cortex and thalamus of the 5XFAD mouse model of AD. The model was characterized by increased Aβ deposition, gliosis, neuronal loss, and memory impairment. Compared to wild-type mice, Cx43 and Cx30 showed increased immunoreactivity in older 5XFAD mice, reflecting astrogliosis, while Cx47 immunoreactivity was reduced. Moreover, Cx47 GJ plaques showed reduced colocalization with Cx43 plaques. Oligodendrocyte precursor cells (OPCs) and mature oligodendrocyte populations were also depleted, and myelin deficits were observed. Our findings indicate reduced astrocyte-oligodendrocyte gap junction connectivity and possibly a shift in Cx43 expression toward astrocyte-astrocyte gap junctions and/or hemichannels, that could impair oligodendrocyte homeostasis and myelination. However, other factors, such as Aβ toxicity, could directly affect oligodendrocyte survival in AD. Our study provides evidence that Cxs might have implications in the progression of AD, although the role of oligodendrocyte Cxs in AD requires further investigation.
Collapse
Affiliation(s)
- Stella Angeli
- Neurobiology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Ioanna Kousiappa
- Neurobiology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Marios Stavrou
- Department of Electrical and Computer Engineering, Faculty of Engineering, University of Cyprus, Nicosia, Cyprus
| | - Irene Sargiannidou
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Elena Georgiou
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Savvas S. Papacostas
- Neurobiology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
- Dementia and Cognitive Disorders Center, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Medical School, University of Nicosia, Nicosia, Cyprus
| | - Kleopas A. Kleopa
- Cyprus School of Molecular Medicine, Nicosia, Cyprus
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Center for Neuromuscular disorders, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- Center for Multiple Sclerosis and Related Disorders, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| |
Collapse
|
32
|
Matsushita T, Masaki K, Isobe N, Sato S, Yamamoto K, Nakamura Y, Watanabe M, Suenaga T, Kira JI. Genetic factors for susceptibility to and manifestations of neuromyelitis optica. Ann Clin Transl Neurol 2020; 7:2082-2093. [PMID: 32979043 PMCID: PMC7664265 DOI: 10.1002/acn3.51147] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/15/2020] [Accepted: 07/11/2020] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE To identify genetic factors associated with susceptibility to and clinical features of neuromyelitis optica spectrum disorders (NMOSD). METHODS Genome-wide single nucleotide polymorphism (SNP) genotyping was conducted in 211 Japanese patients with NMOSD fulfilling the 2006 criteria with or without anti-aquaporin-4 (AQP4) antibody and 1,919 Japanese healthy controls (HCs). HLA-DRB1 and HLA-DPB1 alleles were genotyped in 184 NMOSD cases and 317 HCs. Multiple sclerosis (MS) risk alleles outside the major histocompatibility complex (MHC) region were tested in NMOSD and MS genetic burden (MSGB) scores were compared between HCs and NMOSD. RESULTS A SNP (rs1964995) in the MHC region was associated with NMOSD susceptibility (odds ratio (OR) = 2.33, P = 4.07 × 10-11 ). HLA-DRB1*08:02 (OR = 2.86, P = 3.03 × 10-4 ) and HLA-DRB1*16:02 (OR = 8.39, P = 1.92 × 10-3 ) were risk alleles for NMOSD susceptibility whereas HLA-DRB1*09:01 was protective (OR = 0.27, P = 1.06 × 10-5 ). Three MS risk variants were associated with susceptibility and MSGB scores were significantly higher in NMOSD than in HCs (P = 0.0095). A SNP in the KCNMA1 (potassium calcium-activated channel subfamily M alpha 1) gene was associated with disability score with genome-wide significance (rs1516512, P = 2.33 × 10-8 ) and transverse myelitis (OR = 1.77, P = 0.011). KCNMA1 was immunohistochemically detected in the perivascular endfeet of astrocytes and its immunoreactivity was markedly diminished in active spinal cord lesions in NMOSD. INTERPRETATION Specific HLA-DRB1 alleles confer NMOSD susceptibility and KCNMA1 is associated with disability and transverse myelitis in NMOSD.
Collapse
Affiliation(s)
- Takuya Matsushita
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriko Isobe
- Department of Neurological Therapeutics, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Shinya Sato
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ken Yamamoto
- Department of Medical Chemistry, Kurume University School of Medicine, Kurume, Japan
| | - Yuri Nakamura
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Mitsuru Watanabe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
33
|
Papaneophytou C, Georgiou E, Kleopa KA. The role of oligodendrocyte gap junctions in neuroinflammation. Channels (Austin) 2020; 13:247-263. [PMID: 31232168 PMCID: PMC6602578 DOI: 10.1080/19336950.2019.1631107] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Gap junctions (GJs) provide channels for direct cell-to-cell connectivity serving the homeostasis in several organs of vertebrates including the central (CNS) and peripheral (PNS) nervous systems. GJs are composed of connexins (Cx), which show a highly distinct cellular and subcellular expression pattern. Oligodendrocytes, the myelinating cells of the CNS, are characterized by extensive GJ connectivity with each other as well as with astrocytes. The main oligodendrocyte connexins forming these GJ channels are Cx47 and Cx32. The importance of these channels has been highlighted by the discovery of human diseases caused by mutations in oligodendrocyte connexins, manifesting with leukodystrophy or transient encephalopathy. Experimental models have provided further evidence that oligodendrocyte GJs are essential for CNS myelination and homeostasis, while a strong inflammatory component has been recognized in the absence of oligodendrocyte connexins. Further studies revealed that connexins are also disrupted in multiple sclerosis (MS) brain, and in experimental models of induced inflammatory demyelination. Moreover, induced demyelination was more severe and associated with higher degree of CNS inflammation in models with oligodendrocyte GJ deficiency, suggesting that disrupted connexin expression in oligodendrocytes is not only a consequence but can also drive a pro-inflammatory environment in acquired demyelinating disorders such as MS. In this review, we summarize the current insights from human disorders as well as from genetic and acquired models of demyelination related to oligodendrocyte connexins, with the remaining challenges and perspectives.
Collapse
Affiliation(s)
- Christos Papaneophytou
- a Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine , Nicosia , Cyprus.,b Department of Life and Health Sciences, School of Sciences and Engineering , University of Nicosia , Nicosia , Cyprus
| | - Elena Georgiou
- a Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine , Nicosia , Cyprus
| | - Kleopas A Kleopa
- a Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine , Nicosia , Cyprus.,c Neurology Clinics , the Cyprus Institute of Neurology and Genetics, and the Cyprus School of Molecular Medicine , Nicosia , Cyprus
| |
Collapse
|
34
|
Caufriez A, Böck D, Martin C, Ballet S, Vinken M. Peptide-based targeting of connexins and pannexins for therapeutic purposes. Expert Opin Drug Discov 2020; 15:1213-1222. [PMID: 32539572 DOI: 10.1080/17460441.2020.1773787] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Connexin and pannexin (hemi)channels play an important role in paracrine and autocrine signaling pathways. The opening of these cellular pores is linked to a wide range of diseases. Therefore, pharmacological closing of connexin and pannexin (hemi)channels seems a promising therapeutic strategy. However, the currently available inhibitors cope with recurring problems concerning selectivity, specificity, stability and/or solubility. AREAS COVERED A number of peptides that mimic specific regions in the native sequence of connexins and pannexins have the potential to overcome some of these hurdles. In this paper, an overview is provided on these peptide-based inhibitors of connexin and pannexin (hemi)channels for therapeutic purposes. The authors also provide the reader with their expert perspectives on the future of these peptide-based inhibitors. EXPERT OPINION Peptide mimetics can become valuable tools in the treatment of connexin-related and pannexin-related diseases. This can be made possible provided that available peptides are optimized, and new peptide mimetics are designed based on knowledge of the mechanisms underlying the gating control of connexin and pannexin (hemi)channels.
Collapse
Affiliation(s)
- Anne Caufriez
- Department of in Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , 1090, Brussels, Belgium
| | - Denise Böck
- Department of in Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , 1090, Brussels, Belgium
| | - Charlotte Martin
- Department of Organic Chemistry, Vrije Universiteit Brussel , 1050, Brussels, Belgium
| | - Steven Ballet
- Department of Organic Chemistry, Vrije Universiteit Brussel , 1050, Brussels, Belgium
| | - Mathieu Vinken
- Department of in Vitro Toxicology and Dermato-Cosmetology, Vrije Universiteit Brussel , 1090, Brussels, Belgium
| |
Collapse
|
35
|
Rocca MA, Cacciaguerra L, Filippi M. Moving beyond anti-aquaporin-4 antibodies: emerging biomarkers in the spectrum of neuromyelitis optica. Expert Rev Neurother 2020; 20:601-618. [DOI: 10.1080/14737175.2020.1764352] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Maria A. Rocca
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Cacciaguerra
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Massimo Filippi
- Neuroimaging Research Unit, Institute of Experimental Neurology, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Neurophysiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
36
|
Takahashi S. Metabolic compartmentalization between astroglia and neurons in physiological and pathophysiological conditions of the neurovascular unit. Neuropathology 2020; 40:121-137. [PMID: 32037635 PMCID: PMC7187297 DOI: 10.1111/neup.12639] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Revised: 12/09/2019] [Accepted: 12/09/2019] [Indexed: 12/17/2022]
Abstract
Astroglia or astrocytes, the most abundant cells in the brain, are interposed between neuronal synapses and microvasculature in the brain gray matter. They play a pivotal role in brain metabolism as well as in the regulation of cerebral blood flow, taking advantage of their unique anatomical location. In particular, the astroglial cellular metabolic compartment exerts supportive roles in dedicating neurons to the generation of action potentials and protects them against oxidative stress associated with their high energy consumption. An impairment of normal astroglial function, therefore, can lead to numerous neurological disorders including stroke, neurodegenerative diseases, and neuroimmunological diseases, in which metabolic derangements accelerate neuronal damage. The neurovascular unit (NVU), the major components of which include neurons, microvessels, and astroglia, is a conceptual framework that was originally used to better understand the pathophysiology of cerebral ischemia. At present, the NVU is a tool for understanding normal brain physiology as well as the pathophysiology of numerous neurological disorders. The metabolic responses of astroglia in the NVU can be either protective or deleterious. This review focuses on three major metabolic compartments: (i) glucose and lactate; (ii) fatty acid and ketone bodies; and (iii) D- and L-serine. Both the beneficial and the detrimental roles of compartmentalization between neurons and astroglia will be discussed. A better understanding of the astroglial metabolic response in the NVU is expected to lead to the development of novel therapeutic strategies for diverse neurological diseases.
Collapse
Affiliation(s)
- Shinichi Takahashi
- Department of Neurology and StrokeSaitama Medical University International Medical CenterSaitamaJapan
- Department of PhysiologyKeio University School of MedicineTokyoJapan
| |
Collapse
|
37
|
Kim HJ, Kim MJ, Mostafa MN, Park JH, Choi HS, Kim YS, Choi EK. RhoA/ROCK Regulates Prion Pathogenesis by Controlling Connexin 43 Activity. Int J Mol Sci 2020; 21:ijms21041255. [PMID: 32070020 PMCID: PMC7072953 DOI: 10.3390/ijms21041255] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Revised: 02/10/2020] [Accepted: 02/11/2020] [Indexed: 11/16/2022] Open
Abstract
Scrapie infection, which converts cellular prion protein (PrPC) into the pathological and infectious isoform (PrPSc), leads to neuronal cell death, glial cell activation and PrPSc accumulation. Previous studies reported that PrPC regulates RhoA/Rho-associated kinase (ROCK) signaling and that connexin 43 (Cx43) expression is upregulated in in vitro and in vivo prion-infected models. However, whether there is a link between RhoA/ROCK and Cx43 in prion disease pathogenesis is uncertain. Here, we investigated the role of RhoA/ROCK signaling and Cx43 in prion diseases using in vitro and in vivo models. Scrapie infection induced RhoA activation, accompanied by increased phosphorylation of LIM kinase 1/2 (LIMK1/2) at Thr508/Thr505 and cofilin at Ser3 and reduced phosphorylation of RhoA at Ser188 in hippocampal neuronal cells and brains of mice. Scrapie infection-induced RhoA activation also resulted in PrPSc accumulation followed by a reduction in the interaction between RhoA and p190RhoGAP (a GTPase-activating protein). Interestingly, scrapie infection significantly enhanced the interaction between RhoA and Cx43. Moreover, RhoA and Cx43 colocalization was more visible in both the membrane and cytoplasm of scrapie-infected hippocampal neuronal cells than in controls. Finally, RhoA and ROCK inhibition reduced PrPSc accumulation and the RhoA/Cx43 interaction, leading to decreased Cx43 hemichannel activity in scrapie-infected hippocampal neuronal cells. These findings suggest that RhoA/ROCK regulates Cx43 activity, which may have an important role in the pathogenesis of prion disease.
Collapse
Affiliation(s)
- Hee-Jun Kim
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do 14066, Korea; (H.-J.K.); (M.-J.K.); (M.N.M.); (J.-H.P.); (H.-S.C.); (Y.-S.K.)
| | - Mo-Jong Kim
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do 14066, Korea; (H.-J.K.); (M.-J.K.); (M.N.M.); (J.-H.P.); (H.-S.C.); (Y.-S.K.)
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, Gangwon-do 24252, Korea
| | - Mohd Najib Mostafa
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do 14066, Korea; (H.-J.K.); (M.-J.K.); (M.N.M.); (J.-H.P.); (H.-S.C.); (Y.-S.K.)
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, Gangwon-do 24252, Korea
| | - Jeong-Ho Park
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do 14066, Korea; (H.-J.K.); (M.-J.K.); (M.N.M.); (J.-H.P.); (H.-S.C.); (Y.-S.K.)
| | - Hong-Seok Choi
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do 14066, Korea; (H.-J.K.); (M.-J.K.); (M.N.M.); (J.-H.P.); (H.-S.C.); (Y.-S.K.)
| | - Yong-Sun Kim
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do 14066, Korea; (H.-J.K.); (M.-J.K.); (M.N.M.); (J.-H.P.); (H.-S.C.); (Y.-S.K.)
- Department of Microbiology, College of Medicine, Hallym University, Chuncheon, Gangwon-do 24252, Korea
| | - Eun-Kyoung Choi
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do 14066, Korea; (H.-J.K.); (M.-J.K.); (M.N.M.); (J.-H.P.); (H.-S.C.); (Y.-S.K.)
- Department of Biomedical Gerontology, Graduate School of Hallym University, Chuncheon, Gangwon-do 24252, Korea
- Correspondence: ; Tel.: +82-31-380-1893; Fax: +82-31-388-3427
| |
Collapse
|
38
|
Oligodendroglial connexin 47 regulates neuroinflammation upon autoimmune demyelination in a novel mouse model of multiple sclerosis. Proc Natl Acad Sci U S A 2020; 117:2160-2169. [PMID: 31932428 DOI: 10.1073/pnas.1901294117] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
In multiple sclerosis plaques, oligodendroglial connexin (Cx) 47 constituting main gap junction channels with astroglial Cx43 is persistently lost. As mice with Cx47 single knockout exhibit no demyelination, the roles of Cx47 remain undefined. We aimed to clarify the effects of oligodendroglia-specific Cx47 inducible conditional knockout (icKO) on experimental autoimmune encephalomyelitis (EAE) induced by myelin oligodendrocyte glycoprotein peptide (MOG35-55) in PLP/CreERT;Cx47fl/fl mice at 14 d after tamoxifen injection. Cx47 icKO mice demonstrated exacerbation of acute and chronic relapsing EAE with more pronounced demyelination than Cx47 flox (fl)/fl littermates. CD3+ T cells more abundantly infiltrated the spinal cord in Cx47 icKO than in Cx47 fl/fl mice throughout the acute to chronic phases. CXCR3-CCR6+CD4+ and IL17+IFNγ-CD4+ helper T (Th) 17 cells isolated from spinal cord and brain tissues were significantly increased in Cx47 icKO mice compared with Cx47 fl/fl mice, while MOG35-55-specific proliferation and proinflammatory cytokine production of splenocytes were unaltered. Microarray analysis of isolated microglia revealed stronger microglial activation toward proinflammatory and injury-response phenotypes with increased expressions of chemokines that can attract Th17 cells, including Ccl2, Ccl3, Ccl4, Ccl7, and Ccl8, in Cx47 icKO mice compared with Cx47 fl/fl mice. In Cx47 icKO mice, NOS2+ and MHC class II+ microglia were more enriched immunohistochemically, and A1-specific astroglial gene expressions and astroglia immunostained for C3, a representative A1 astrocyte marker, were significantly increased at the acute phase, compared with Cx47 fl/fl mice. These findings suggest that oligodendroglia-specific Cx47 ablation induces severe inflammation upon autoimmune demyelination, underscoring a critical role for Cx47 in regulating neuroinflammation.
Collapse
|
39
|
Droguerre M, Tsurugizawa T, Duchêne A, Portal B, Guiard BP, Déglon N, Rouach N, Hamon M, Mouthon F, Ciobanu L, Charvériat M. A New Tool for In Vivo Study of Astrocyte Connexin 43 in Brain. Sci Rep 2019; 9:18292. [PMID: 31797899 PMCID: PMC6892890 DOI: 10.1038/s41598-019-54858-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 11/20/2019] [Indexed: 01/22/2023] Open
Abstract
Astrocytes are glial cells organized in dynamic and structured networks in the brain. These plastic networks, involving key proteins such as connexin 43 (Cx43), are engaged in fine neuronal tuning and have recently been considered as emerging therapeutic targets in central nervous system disorders. We developed and validated a new application of the manganese-enhanced magnetic resonance imaging (MEMRI) technique allowing in vivo investigations of astrocyte-neuron interactions through quantification of brain Cx43 functional activity. The proof of concept has been achieved by quantification of MEMRI signals in brain after either local astrocyte-specific Cx43 knockdown with shRNA or systemic administration of Cx43 blockers. Unilateral hippocampal Cx43 genetical silencing was associated with an ipsilateral local increase of MEMRI signal. Furthermore, Cx43 blockers also enhanced MEMRI signal responses in hippocampus. Altogether, these data reveal the MEMRI technique as a tool for quantitative imaging of in vivo Cx43-dependent function in astrocytes under physiological and pathological conditions.
Collapse
Affiliation(s)
| | | | | | - Benjamin Portal
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31330, Toulouse, France
| | - Bruno P Guiard
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université de Toulouse, CNRS, UPS, 31330, Toulouse, France
| | - Nicole Déglon
- Laboratory of Neurotherapies and NeuroModulation, Neuroscience research Center (CRN), Lausanne University Hospital (CHUV) and University of Lausanne, 1011, Lausanne, Switzerland.,Laboratory of Neurotherapies and NeuroModulation, Department of Clinical Neuroscience (DNC), Lausanne University Hospital (CHUV) and University of Lausanne, 1011, Lausanne, Switzerland
| | - Nathalie Rouach
- Laboratory of Neuroglial Interactions in Cerebral Physiopathology, Center for Interdisciplinary Research in Biology, Collège de France, CNRS UMR 7241, INSERM U1050, Labex Memolife, PSL Research University, Paris, 75005, France
| | - Michel Hamon
- Theranexus, 60 Avenue Rockefeller, 69008, Lyon, France
| | | | | | | |
Collapse
|
40
|
Molina-Gonzalez I, Miron VE. Astrocytes in myelination and remyelination. Neurosci Lett 2019; 713:134532. [PMID: 31589903 DOI: 10.1016/j.neulet.2019.134532] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Revised: 09/13/2019] [Accepted: 10/01/2019] [Indexed: 02/07/2023]
Abstract
Astrocytes are known to play critical roles in central nervous system development, homeostasis, and response to injury. In addition to well-defined functions in synaptic signalling and blood-brain barrier control, astrocytes are now emerging as important contributors to white matter health. Here, we review the roles of astrocytes in myelin formation and regeneration (remyelination), focusing on both direct interactions with oligodendrocyte lineage cells, and indirect influences via crosstalk with central nervous system resident macrophages, microglia.
Collapse
Affiliation(s)
- Irene Molina-Gonzalez
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom
| | - Veronique E Miron
- Medical Research Council Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.
| |
Collapse
|
41
|
Astrocytes in multiple sclerosis and experimental autoimmune encephalomyelitis: Star-shaped cells illuminating the darkness of CNS autoimmunity. Brain Behav Immun 2019; 80:10-24. [PMID: 31125711 DOI: 10.1016/j.bbi.2019.05.029] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/16/2019] [Accepted: 05/20/2019] [Indexed: 12/18/2022] Open
Abstract
Neuropathology in the human autoimmune disease multiple sclerosis (MS) is considered to be mediated by autoreactive leukocytes, such as T cells, B cells, and macrophages. However, the inflammation and tissue damage in MS and its animal model experimental autoimmune encephalomyelitis (EAE) is also critically regulated by astrocytes, the most abundant cell population in the central nervous system (CNS). Under physiological conditions, astrocytes are integral to the development and function of the CNS, whereas in CNS autoimmunity, astrocytes influence the pathogenesis, progression, and recovery of the diseases. In this review, we summarize recent advances in astrocytic functions in the context of MS and EAE, which are categorized into two opposite aspects, one being detrimental and the other beneficial. Inhibition of the detrimental functions and/or enhancement of the beneficial functions of astrocytes might be favorable for the treatment of MS.
Collapse
|
42
|
The contribution of astrocytes to the neuroinflammatory response in multiple sclerosis and experimental autoimmune encephalomyelitis. Acta Neuropathol 2019; 137:757-783. [PMID: 30847559 DOI: 10.1007/s00401-019-01980-7] [Citation(s) in RCA: 159] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/21/2019] [Accepted: 02/23/2019] [Indexed: 02/06/2023]
Abstract
Neuroinflammation is the coordinated response of the central nervous system (CNS) to threats to its integrity posed by a variety of conditions, including autoimmunity, pathogens and trauma. Activated astrocytes, in concert with other cellular elements of the CNS and immune system, are important players in the modulation of the neuroinflammatory response. During neurological disease, they produce and respond to cellular signals that often lead to dichotomous processes, which can promote further damage or contribute to repair. This occurs also in multiple sclerosis (MS), where astrocytes are now recognized as key components of its immunopathology. Evidence supporting this role has emerged not only from studies in MS patients, but also from animal models, among which the experimental autoimmune encephalomyelitis (EAE) model has proved especially instrumental. Based on this premise, the purpose of the present review is to summarize the current knowledge of astrocyte behavior in MS and EAE. Following a brief description of the pathological characteristics of the two diseases and the main functional roles of astrocytes in CNS physiology, we will delve into the specific responses of this cell population, analyzing MS and EAE in parallel. We will define the temporal and anatomical profile of astroglial activation, then focus on key processes they participate in. These include: (1) production and response to soluble mediators (e.g., cytokines and chemokines), (2) regulation of oxidative stress, and (3) maintenance of BBB integrity and function. Finally, we will review the state of the art on the available methods to measure astroglial activation in vivo in MS patients, and how this could be exploited to optimize diagnosis, prognosis and treatment decisions. Ultimately, we believe that integrating the knowledge obtained from studies in MS and EAE may help not only better understand the pathophysiology of MS, but also uncover new signals to be targeted for therapeutic intervention.
Collapse
|
43
|
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating disorder. Although all MS patients initially show a relapsing-remitting course, 20-50% subsequently enter a chronic progressive course at 10-20 years after onset that greatly influences their activities of daily living. There are 2.5 million MS patients worldwide with large regional and racial differences. In particular, there are many MS patients among Caucasians living in Europe, while the disease is relatively rare in Asians and Africans.Although MS is regarded as an autoimmune disease, many factors such as genetic background, environmental factors, and sex are involved in its pathogenesis. While the immunological mechanisms remain to be fully elucidated, invasion of autoreactive T cells into the central nervous system (CNS) tissue is considered the first step of the disease. These T cells react with myelin antigens and initiate demyelination of the CNS by activating cytotoxic T cells, macrophages, and B cells through the release of inflammatory cytokines. As a treatment option, disease-modifying therapies have recently been developed to prevent the recurrence of MS in addition to conventional treatment with corticosteroids for acute relapse. However, there are still few effective treatments for the chronic progressive phase, and it is thus imperative to decipher the mechanism for chronic progression.
Collapse
|
44
|
Fang M, Yamasaki R, Li G, Masaki K, Yamaguchi H, Fujita A, Isobe N, Kira JI. Connexin 30 Deficiency Attenuates Chronic but Not Acute Phases of Experimental Autoimmune Encephalomyelitis Through Induction of Neuroprotective Microglia. Front Immunol 2018; 9:2588. [PMID: 30464764 PMCID: PMC6234958 DOI: 10.3389/fimmu.2018.02588] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2018] [Accepted: 10/19/2018] [Indexed: 01/25/2023] Open
Abstract
Glial connexins (Cxs) form gap junction channels through which a pan-glial network plays key roles in maintaining homeostasis of the central nervous system (CNS). In multiple sclerosis (MS) and its animal model, experimental autoimmune encephalomyelitis (EAE), expression of astrocytic Cx43 is lost in acute lesions but upregulated in chronic plaques, while astrocytic Cx30 is very low in normal white matter and changes in its expression have not been convincingly shown. In Cx30 or Cx43 single knockout (KO) mice and even in Cx30/Cx43 double KO mice, acute EAE is unaltered. However, the effects of Cx30/Cx43 deficiency on chronic EAE remains to be elucidated. We aimed to clarify the roles of Cx30 in chronic neuroinflammation by studying EAE induced by myelin oligodendrocyte glycoprotein peptide 35–55 in Cx30 KO mice. We found that Cx30 deficiency improved the clinical symptoms and demyelination of chronic but not acute EAE without influencing CD3+ T cell infiltration. Furthermore, increased ramified microglia in the naïve state and induced earlier and stronger microglial activation in the acute and chronic phases of EAE was observed. These activated microglia had an anti-inflammatory phenotype, as shown by the upregulation of arginase-1 and brain-derived neurotrophic factor and the downregulation of nitric oxide synthase 2. In the naïve state, Cx30 deficiency induced modest enlargement of astrocytic processes in the spinal cord gray matter and a partial reduction of Cx43 expression in the spinal cord white matter. These astrocytes in Cx30 KO mice showed earlier and stronger activation during the acute phase of EAE, with upregulated A2 astrocyte markers and a significant decrease in Cx43 in the chronic phases. Spinal cord neurons and axons were more preserved in Cx30 KO mice than in littermates in the chronic phase of EAE. These findings suggest that Cx30 deficiency increased ramified microglia in the CNS in the naïve state and improved chronic EAE through redirecting microglia toward an anti-inflammatory phenotype, suggesting a hitherto unknown critical role of astrocytic Cx30 in regulating microglial number and functional state.
Collapse
Affiliation(s)
- Mei Fang
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Ryo Yamasaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Guangrui Li
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Katsuhisa Masaki
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Hiroo Yamaguchi
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Atsushi Fujita
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Noriko Isobe
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.,Department of Neurological Therapeutics, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Jun-Ichi Kira
- Department of Neurology, Neurological Institute, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| |
Collapse
|
45
|
Function of Connexins in the Interaction between Glial and Vascular Cells in the Central Nervous System and Related Neurological Diseases. Neural Plast 2018; 2018:6323901. [PMID: 29983707 PMCID: PMC6015683 DOI: 10.1155/2018/6323901] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 05/06/2018] [Accepted: 05/14/2018] [Indexed: 02/05/2023] Open
Abstract
Neuronal signaling together with synapse activity in the central nervous system requires a precisely regulated microenvironment. Recently, the blood-brain barrier is considered as a “neuro-glia-vascular unit,” a structural and functional compound composed of capillary endothelial cells, glial cells, pericytes, and neurons, which plays a pivotal role in maintaining the balance of the microenvironment in and out of the brain. Tight junctions and adherens junctions, which function as barriers of the blood-brain barrier, are two well-known kinds in the endothelial cell junctions. In this review, we focus on the less-concerned contribution of gap junction proteins, connexins in blood-brain barrier integrity under physio-/pathology conditions. In the neuro-glia-vascular unit, connexins are expressed in the capillary endothelial cells and prominent in astrocyte endfeet around and associated with maturation and function of the blood-brain barrier through a unique signaling pathway and an interaction with tight junction proteins. Connexin hemichannels and connexin gap junction channels contribute to the physiological or pathological progress of the blood-brain barrier; in addition, the interaction with other cell-cell-adhesive proteins is also associated with the maintenance of the blood-brain barrier. Lastly, we explore the connexins and connexin channels involved in the blood-brain barrier in neurological diseases and any programme for drug discovery or delivery to target or avoid the blood-brain barrier.
Collapse
|
46
|
Affiliation(s)
- Ryo Yamasaki
- Department of Neurology; Neurological Institute; Graduate School of Medical Sciences; Kyushu University; Fukuoka Japan
| |
Collapse
|
47
|
Long Y, Liang J, Xu H, Huang Q, Yang J, Gao C, Qiu W, Lin S, Chen X. Autoimmune glial fibrillary acidic protein astrocytopathy in Chinese patients: a retrospective study. Eur J Neurol 2018; 25:477-483. [PMID: 29193473 DOI: 10.1111/ene.13531] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 11/02/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Y. Long
- Department of Neurology Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
- Institute of Neuroscience Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province Ministry of Education of China Collaborative Innovation Center for Neurogenetics and Channelopathies Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - J. Liang
- Department of Neurology Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
- Institute of Neuroscience Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province Ministry of Education of China Collaborative Innovation Center for Neurogenetics and Channelopathies Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - H. Xu
- Department of Neurology Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
- Institute of Neuroscience Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province Ministry of Education of China Collaborative Innovation Center for Neurogenetics and Channelopathies Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - Q. Huang
- Department of Neurology Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
- Institute of Neuroscience Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province Ministry of Education of China Collaborative Innovation Center for Neurogenetics and Channelopathies Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - J. Yang
- Department of Neurology Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
- Institute of Neuroscience Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province Ministry of Education of China Collaborative Innovation Center for Neurogenetics and Channelopathies Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - C. Gao
- Department of Neurology Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
- Institute of Neuroscience Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province Ministry of Education of China Collaborative Innovation Center for Neurogenetics and Channelopathies Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - W. Qiu
- Department of Neurology Third Affiliated Hospital of Sun Yat‐Sen University Guangzhou China
| | - S. Lin
- Department of Emergency Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
| | - X. Chen
- Department of Emergency Second Affiliated Hospital of Guangzhou Medical University Guangzhou China
| |
Collapse
|
48
|
Bose A, Basu R, Maulik M, Das Sarma J. Loss of Cx43-Mediated Functional Gap Junction Communication in Meningeal Fibroblasts Following Mouse Hepatitis Virus Infection. Mol Neurobiol 2018; 55:6558-6571. [PMID: 29327203 PMCID: PMC7090783 DOI: 10.1007/s12035-017-0861-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/21/2017] [Indexed: 10/28/2022]
Abstract
Mouse hepatitis virus (MHV) infection causes meningoencephalitis by disrupting the neuro-glial and glial-pial homeostasis. Recent studies suggest that MHV infection alters gap junction protein connexin 43 (Cx43)-mediated intercellular communication in brain and primary cultured astrocytes. In addition to astrocytes, meningeal fibroblasts also express high levels of Cx43. Fibroblasts in the meninges together with the basal lamina and the astrocyte endfeet forms the glial limitans superficialis as part of the blood-brain barrier (BBB). Alteration of glial-pial gap junction intercellular communication (GJIC) in MHV infection has the potential to affect the integrity of BBB. Till date, it is not known if viral infection can modulate Cx43 expression and function in cells of the brain meninges and thus affect BBB permeability. In the present study, we have investigated the effect of MHV infection on Cx43 localization and function in mouse brain meningeal cells and primary meningeal fibroblasts. Our results show that MHV infection reduces total Cx43 levels and causes its intracellular retention in the perinuclear compartments reducing its surface expression. Reduced trafficking of Cx43 to the cell surface in MHV-infected cells is associated with loss functional GJIC. Together, these data suggest that MHV infection can directly affect expression and cellular distribution of Cx43 resulting in loss of Cx43-mediated GJIC in meningeal fibroblasts, which may be associated with altered BBB function observed in acute infection.
Collapse
Affiliation(s)
- Abhishek Bose
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, Nadia, West Bengal, 741246, India
| | - Rahul Basu
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, Nadia, West Bengal, 741246, India
| | - Mahua Maulik
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, Nadia, West Bengal, 741246, India
| | - Jayasri Das Sarma
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata (IISER-K), Mohanpur, Nadia, West Bengal, 741246, India.
| |
Collapse
|
49
|
Allergic Inflammation Leads to Neuropathic Pain via Glial Cell Activation. J Neurosci 2017; 36:11929-11945. [PMID: 27881779 DOI: 10.1523/jneurosci.1981-16.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/23/2016] [Accepted: 09/27/2016] [Indexed: 01/08/2023] Open
Abstract
Allergic and atopic disorders have increased over the past few decades and have been associated with neuropsychiatric conditions, such as autism spectrum disorder and asthmatic amyotrophy. Myelitis presenting with neuropathic pain can occur in patients with atopic disorder; however, the relationship between allergic inflammation and neuropathic pain, and the underlying mechanism, remains to be established. We studied whether allergic inflammation affects the spinal nociceptive system. We found that mice with asthma, atopic dermatitis, or atopic diathesis had widespread and significantly more activated microglia and astroglia in the spinal cord than those without atopy, and displayed tactile allodynia. Microarray analysis of isolated microglia revealed a dysregulated phenotype showing upregulation of M1 macrophage markers and downregulation of M2 markers in atopic mice. Among the cell surface protein genes, endothelin receptor type B (EDNRB) was most upregulated. Immunohistochemical analysis revealed that EDNRB expression was enhanced in microglia and astroglia, whereas endothelin-1, an EDNRB ligand, was increased in serum, lungs, and epidermis of atopic mice. No EDNRA expression was found in the spinal cord. Expression of FBJ murine osteosarcoma viral oncogene homolog B was significantly higher in the dorsal horn neurons of asthma mice than nonatopic mice. The EDNRB antagonist BQ788 abolished glial and neural activation and allodynia. We found increased serum endothelin-1 in atopic patients with myelitis and neuropathic pain, and activation of spinal microglia and astroglia with EDNRB upregulation in an autopsied case. These results suggest that allergic inflammation induces diffuse glial activation, influencing the nociceptive system via the EDNRB pathway. SIGNIFICANCE STATEMENT The prevalence of allergic disorders has markedly increased over the past few decades. Allergic disorders are associated with neuropsychiatric conditions; however, the relationship between allergic inflammation and CNS complications is unknown. A peculiar myelitis presenting with persistent neuropathic pain has been reported in patients with allergic disorders. We studied how atopy exerts substantial influence on the nociceptive system. We found that mice with allergic disorders had severe allodynia with activated astroglia and microglia, and showed marked upregulation of endothelin-1 (ET-1) receptor type B (EDNRB) in the spinal cord. A selective EDNRB antagonist prevented allodynia and glial activation. Our findings suggest a novel mechanism whereby atopy induces glial activation and neuropathic pain via an ET-1/EDNRB pathway.
Collapse
|
50
|
Völgyi K, Gulyássy P, Todorov MI, Puska G, Badics K, Hlatky D, Kékesi KA, Nyitrai G, Czurkó A, Drahos L, Dobolyi A. Chronic Cerebral Hypoperfusion Induced Synaptic Proteome Changes in the rat Cerebral Cortex. Mol Neurobiol 2017; 55:4253-4266. [PMID: 28620701 DOI: 10.1007/s12035-017-0641-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/29/2017] [Indexed: 12/23/2022]
Abstract
Chronic cerebral hypoperfusion (CCH) evokes mild cognitive impairment (MCI) and contributes to the progression of vascular dementia and Alzheimer's disease (AD). How CCH induces these neurodegenerative processes that may spread along the synaptic network and whether they are detectable at the synaptic proteome level of the cerebral cortex remains to be established. In the present study, we report the synaptic protein changes in the cerebral cortex after stepwise bilateral common carotid artery occlusion (BCCAO) induced CCH in the rat. The occlusions were confirmed with magnetic resonance angiography 5 weeks after the surgery. Synaptosome fractions were prepared using sucrose gradient centrifugation from cerebral cortex dissected 7 weeks after the occlusion. The synaptic protein differences between the sham operated and CCH groups were analyzed with label-free nanoUHPLC-MS/MS. We identified 46 proteins showing altered abundance due to CCH. In particular, synaptic protein and lipid metabolism, as well as GABA shunt-related proteins showed increased while neurotransmission and synaptic assembly-related proteins showed decreased protein level changes in CCH rats. Protein network analysis of CCH-induced protein alterations suggested the importance of increased synaptic apolipoprotein E (APOE) level as a consequence of CCH. Therefore, the change in APOE level was confirmed with Western blotting. The identified synaptic protein changes would precede the onset of dementia-like symptoms in the CCH model, suggesting their importance in the development of vascular dementia.
Collapse
Affiliation(s)
- Katalin Völgyi
- MTA-ELTE NAP B Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Pázmány Péter sétány 1C, Budapest, H-1117, Hungary.
| | - Péter Gulyássy
- MTA-TTK NAP B MS Neuroproteomics Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Mihail Ivilinov Todorov
- MTA-ELTE NAP B Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Pázmány Péter sétány 1C, Budapest, H-1117, Hungary.,Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest, Hungary
| | - Gina Puska
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, Budapest, Hungary
| | - Kata Badics
- Laboratory of Proteomics, Institute of Biology, Eötvös Loránd University, Budapest, Hungary
| | - Dávid Hlatky
- Preclinical Imaging and Biomarker Laboratory, Pharmacology and Drug Safety Research, Richter Gedeon Plc, Budapest, Hungary
| | - Katalin Adrienna Kékesi
- MTA-TTK NAP B MS Neuroproteomics Research Group, Hungarian Academy of Sciences, Budapest, Hungary.,Department of Physiology and Neurobiology, Eötvös Loránd University, Budapest, Hungary
| | - Gabriella Nyitrai
- Preclinical Imaging and Biomarker Laboratory, Pharmacology and Drug Safety Research, Richter Gedeon Plc, Budapest, Hungary
| | - András Czurkó
- Preclinical Imaging and Biomarker Laboratory, Pharmacology and Drug Safety Research, Richter Gedeon Plc, Budapest, Hungary
| | - László Drahos
- MTA-TTK NAP B MS Neuroproteomics Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Arpád Dobolyi
- MTA-ELTE NAP B Laboratory of Molecular and Systems Neurobiology, Institute of Biology, Hungarian Academy of Sciences and Eötvös Loránd University, Pázmány Péter sétány 1C, Budapest, H-1117, Hungary
| |
Collapse
|