Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.651
Filter
1.
BMJ Case Rep ; 17(5)2024 May 09.
Article in English | MEDLINE | ID: mdl-38724215

ABSTRACT

Autoimmune encephalitis due to glial fibrillar acidic protein (GFAP) astrocytopathy is a rare cause of subacute neuropsychiatric changes. In this case, a young patient presented with a viral prodrome and meningismus, followed by progressive encephalopathy and movement disorders over the span of 2 weeks. Due to his clinical trajectory, inflammatory cerebrospinal fluid (CSF) analysis, initial normal brain imaging and negative serum autoimmune encephalopathy panel, his initial diagnosis was presumed viral meningoencephalitis. The recurrence and progression of neuropsychiatric symptoms and myoclonus despite antiviral treatment prompted further investigation, inclusive of testing for CSF autoimmune encephalopathy autoantibodies, yielding a clinically meaningful, positive GFAP autoantibody. This case highlights the importance of appropriately testing both serum and CSF autoantibodies when an autoimmune encephalitic process is considered. Through this case, we review the clinical and radiographic manifestations of GFAP astrocytopathy, alongside notable pearls pertaining to this autoantibody syndrome and its management.


Subject(s)
Autoantibodies , Encephalitis , Glial Fibrillary Acidic Protein , Humans , Male , Glial Fibrillary Acidic Protein/blood , Glial Fibrillary Acidic Protein/immunology , Glial Fibrillary Acidic Protein/cerebrospinal fluid , Encephalitis/diagnosis , Encephalitis/immunology , Autoantibodies/blood , Autoantibodies/cerebrospinal fluid , Astrocytes/pathology , Astrocytes/immunology , Autoimmune Diseases of the Nervous System/diagnosis , Autoimmune Diseases of the Nervous System/immunology , Hashimoto Disease/diagnosis , Hashimoto Disease/blood , Diagnosis, Differential , Adult , Magnetic Resonance Imaging
2.
Methods Mol Biol ; 2807: 271-283, 2024.
Article in English | MEDLINE | ID: mdl-38743235

ABSTRACT

The blood-brain barrier (BBB) is one of several barriers between the brain and the peripheral blood system to maintain homeostasis. Understanding the interactions between infectious agents such as human immunodeficiency virus type 1 (HIV-1), which are capable of traversing the BBB and causing neuroinflammation requires modeling an authentic BBB in vitro. Such an in vitro BBB model also helps develop means of targeting viruses that reside in the brain via natural immune effectors such as antibodies. The BBB consists of human brain microvascular endothelial cells (HBMECs), astrocytes, and pericytes. Here we report in vitro methods to establish a dual-cell BBB model consisting of primary HBMECs and primary astrocytes to measure the integrity of the BBB and antibody penetration of the BBB, as well as a method to establish a single cell BBB model to study the impact of HIV-1 infected medium on the integrity of such a BBB.


Subject(s)
Astrocytes , Blood-Brain Barrier , Endothelial Cells , HIV Infections , HIV-1 , Blood-Brain Barrier/virology , Blood-Brain Barrier/metabolism , Humans , Astrocytes/virology , Astrocytes/metabolism , Astrocytes/immunology , Endothelial Cells/virology , Endothelial Cells/metabolism , Endothelial Cells/immunology , HIV-1/immunology , HIV-1/physiology , HIV Infections/virology , HIV Infections/immunology , Pericytes/virology , Pericytes/metabolism , Pericytes/immunology , Neuroinflammatory Diseases/virology , Neuroinflammatory Diseases/immunology , Coculture Techniques/methods , Cells, Cultured , Brain/virology , Brain/immunology , Brain/metabolism
3.
Trends Immunol ; 45(5): 320-321, 2024 May.
Article in English | MEDLINE | ID: mdl-38632002

ABSTRACT

Astrocytes are essential cells of the mammalian central nervous system (CNS), with key roles in development, homeostasis, and disease. Lee and colleagues recently showed that astrocytes can develop epigenetic memory, which enhances proinflammatory responses to subsequent stimulation, potentially driving sustained neurological disease pathology, such as in multiple sclerosis (MS).


Subject(s)
Astrocytes , Neuroinflammatory Diseases , Astrocytes/immunology , Humans , Animals , Neuroinflammatory Diseases/immunology , Multiple Sclerosis/immunology , Epigenesis, Genetic , Central Nervous System/immunology , Inflammation/immunology , Chronic Disease
4.
Front Immunol ; 15: 1361685, 2024.
Article in English | MEDLINE | ID: mdl-38665914

ABSTRACT

A 54-year-old Japanese man presented with headache and fever the day after SARS-CoV-2 vaccination. He became deeply unconscious within a week. Brain MRI showed periventricular linear enhancements and a few spotty lesions in the cerebral white matter. Cerebrospinal fluid (CSF) testing showed mild pleocytosis. He was treated with intravenous methylprednisolone and plasma exchange. However, the white matter lesions enlarged to involve the brainstem and cerebellum, and long cord spinal lesions appeared. Anti-glial fibrillary acidic protein (GFAP) antibody was positive in the CSF and serum, and he was therefore diagnosed as autoimmune GFAP-astrocytopathy (GFAP-A). In addition, high-dose immunoglobulin therapy was administered twice, but his symptoms did not improve; the white matter lesions enlarged further, and modified Rankin Scale score increased to 5. A brain biopsy specimen showed infiltration of macrophages and CD4 + lymphocytes together with neuron and oligodendrocytic injuries and glial scar. Although GFAP-A generally responds well to steroids, the present case developed GFAP-A following SARS-CoV-2 vaccination, with refractory to intensive immunosuppressive therapy and atypical pathologic findings of infiltration of CD4 + lymphocytes and demyelination.


Subject(s)
COVID-19 , Glial Fibrillary Acidic Protein , SARS-CoV-2 , Humans , Male , Middle Aged , Glial Fibrillary Acidic Protein/immunology , COVID-19/immunology , SARS-CoV-2/immunology , Immunosuppressive Agents/adverse effects , Immunosuppressive Agents/therapeutic use , Astrocytes/immunology , Astrocytes/pathology , COVID-19 Vaccines/adverse effects , COVID-19 Vaccines/immunology , Autoantibodies/blood , Autoantibodies/immunology , Vaccination/adverse effects , Brain/pathology , Brain/diagnostic imaging
5.
Mult Scler Relat Disord ; 85: 105527, 2024 May.
Article in English | MEDLINE | ID: mdl-38432014

ABSTRACT

OBJECTIVE: The differential diagnosis between autoimmune glial fibrillary acidic protein astrocytopathy (AGFAPA) mimicking tuberculous meningitis and tuberculous meningitis (TBM) remains challenging in clinical practice. This study aims to identify the clinical, laboratory parameters, and clinical score systems that may be helpful in differentiating AGFAPA from TBM. METHOD: Overall 22 AGFAPA patients who were initially misdiagnosed as TBM (AGFAPA-TBM) and 30 confirmed TBM patients were included. The clinical, laboratory, imaging parameters, Thwaites systems, and Lancet consensus scoring systems (LCSS) of all patients were reviewed. Logistic regression was employed to establish a diagnostic formula to differentiate AGFAPA-TBM from TBM. The receiver operating characteristic (ROC) curve was applied to determine the best diagnostic critical point of the formula. RESULTS: Urinary retention was more frequent in AGFAPA-TBM patients (72.7% vs 33.3%, p = 0.012). A significantly lower ratio of T-SPOT. TB was noted in AGFAPA-TBM patients (9.1% vs 82.1%, p < 0.001). We found the LCSS was able to differentiate AGFAPA-TBM from TBM (AUC value 0.918, 95% CI=0.897-0.924). Furthermore, we set up a new scoring system with three variables: urinary retention, T-SPOT. TB, and cerebral imaging criteria in LCSS. The proposed diagnostic score ranges from -8 to 2, and a score of ≥ 0 was suggestive of AGFAPA-TBM (AUC value 0.938, 95% CI=0.878-0.951). CONCLUSIONS: This study is the first to evaluate the Thwaites system and LCSS in AGFAPA-TBM and TBM. We provide an alternative diagnostic formula to differentiate AGFAPA-TBM from TBM and suggest testing for GFAP antibodies to avoid misdiagnosis when this scoring system meets AGFAPA-TBM.


Subject(s)
Glial Fibrillary Acidic Protein , Tuberculosis, Meningeal , Humans , Tuberculosis, Meningeal/diagnosis , Female , Male , Diagnosis, Differential , Glial Fibrillary Acidic Protein/immunology , Adult , Middle Aged , Young Adult , Retrospective Studies , Autoimmune Diseases of the Nervous System/diagnosis , Autoimmune Diseases of the Nervous System/immunology , Astrocytes/immunology , Autoantibodies/blood
6.
J Neuroimmunol ; 382: 578174, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37573633

ABSTRACT

We describe three cases of overlapping Epstein-Barr virus (EBV) Encephalitis and Autoimmune Glial Fibrillary Acidic Protein Astrocytopathy (GFAP-A). The three cases all presented with initial symptoms of fever, headache, coma, and posture tremor of the upper limbs, then followed by limb weakness and dysuria. All of the three cases were on ventilators. Case 1 and 2 improved dramatically after intravenous methylprednisoloneand immunoglobulin treatment. However, case 3 presented dyspneic, and died from gastrointestinal hemorrhage. The GFAP-A triggered by EBV intracranial infection could initially masquerade as EBV encephalitis only, and the detection of GFAP antibody is essential for differentiation.


Subject(s)
Astrocytes , Autoimmune Diseases of the Nervous System , Encephalitis , Epstein-Barr Virus Infections , Glial Fibrillary Acidic Protein , Humans , Antibodies , Astrocytes/immunology , Astrocytes/metabolism , Autoantibodies , Encephalitis/complications , Encephalitis/immunology , Encephalitis/therapy , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/therapy , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/immunology , Herpesvirus 4, Human , Immunoglobulins, Intravenous , Methylprednisolone/therapeutic use , Glucocorticoids/therapeutic use , Autoimmune Diseases of the Nervous System/complications , Autoimmune Diseases of the Nervous System/diagnosis , Autoimmune Diseases of the Nervous System/therapy , Diagnosis, Differential
7.
Proc Natl Acad Sci U S A ; 119(35): e2211310119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994674

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS). Astrocytes are the most abundant glial cells in the CNS, and their dysfunction contributes to the pathogenesis of MS and its animal model, experimental autoimmune encephalomyelitis (EAE). Recent advances highlight the pivotal role of cellular metabolism in programming immune responses. However, the underlying immunometabolic mechanisms that drive astrocyte pathogenicity remain elusive. Nicotinamide adenine dinucleotide (NAD+) is a vital coenzyme involved in cellular redox reactions and a substrate for NAD+-dependent enzymes. Cellular NAD+ levels are dynamically controlled by synthesis and degradation, and dysregulation of this balance has been associated with inflammation and disease. Here, we demonstrate that cell-autonomous generation of NAD+ via the salvage pathway regulates astrocyte immune function. Inhibition of nicotinamide phosphoribosyltransferase (NAMPT), a key enzyme in the salvage pathway, results in depletion of NAD+, inhibits oxidative phosphorylation, and limits astrocyte inflammatory potential. We identified CD38 as the main NADase up-regulated in reactive mouse and human astrocytes in models of neuroinflammation and MS. Genetic or pharmacological blockade of astrocyte CD38 activity augmented NAD+ levels, suppressed proinflammatory transcriptional reprogramming, impaired chemotactic potential to inflammatory monocytes, and ameliorated EAE. We found that CD38 activity is mediated via calcineurin/NFAT signaling in mouse and human reactive astrocytes. Thus, NAMPT-NAD+-CD38 circuitry in astrocytes controls their ability to meet their energy demands and drives the expression of proinflammatory transcriptional modules, contributing to CNS pathology in EAE and, potentially, MS. Our results identify candidate therapeutic targets in MS.


Subject(s)
ADP-ribosyl Cyclase 1 , Astrocytes , Encephalomyelitis, Autoimmune, Experimental , Multiple Sclerosis , NAD , ADP-ribosyl Cyclase 1/metabolism , Animals , Astrocytes/immunology , Astrocytes/metabolism , Autoimmunity , Central Nervous System/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Mice , Multiple Sclerosis/immunology , NAD/metabolism , Nicotinamide Phosphoribosyltransferase/metabolism
9.
Exp Neurol ; 348: 113943, 2022 02.
Article in English | MEDLINE | ID: mdl-34863998

ABSTRACT

Astrocytes control many processes of the nervous system in health and disease, and respond to injury quickly. Astrocytes produce neuroprotective factors in the injured brain to clear cellular debris and to orchestrate neurorestorative processes that are beneficial for neurological recovery after traumatic brain injury (TBI). However, astrocytes also become dysregulated and produce cytotoxic mediators that hinder CNS repair by induction of neuronal dysfunction and cell death. Hence, we discuss the potential role of astrocytes in neuropathological processes such as neuroinflammation, neurogenesis, synaptogenesis and blood-brain barrier repair after TBI. Thus, an improved understanding of the dual role of astrocytes may advance our knowledge of post-brain injury recovery, and provide opportunities for the development of novel therapeutic strategies for TBI.


Subject(s)
Astrocytes/metabolism , Blood-Brain Barrier/metabolism , Brain Injuries, Traumatic/metabolism , Brain/metabolism , Inflammation Mediators/metabolism , Animals , Astrocytes/immunology , Blood-Brain Barrier/immunology , Brain/immunology , Brain Injuries, Traumatic/immunology , Cell Death/physiology , Humans , Inflammation Mediators/immunology , Neurogenesis/physiology , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/metabolism
10.
Cell Rep ; 37(13): 110158, 2021 12 28.
Article in English | MEDLINE | ID: mdl-34965428

ABSTRACT

Non-neuronal responses in neurodegenerative disease have received increasing attention as important contributors to disease pathogenesis and progression. Here we utilize single-cell RNA sequencing to broadly profile 13 cell types in three different mouse models of Alzheimer disease (AD), capturing the effects of tau-only, amyloid-only, or combined tau-amyloid pathology. We highlight microglia, oligodendrocyte, astrocyte, and T cell responses and compare them across these models. Notably, we identify two distinct transcriptional states for oligodendrocytes emerging differentially across disease models, and we determine their spatial distribution. Furthermore, we explore the impact of Trem2 deletion in the context of combined pathology. Trem2 knockout mice exhibit severely blunted microglial responses to combined tau and amyloid pathology, but responses from non-microglial cell types (oligodendrocytes, astrocytes, and T cells) are relatively unchanged. These results delineate core transcriptional states that are engaged in response to AD pathology, and how they are influenced by a key AD risk gene, Trem2.


Subject(s)
Alzheimer Disease/pathology , Amyloid/chemistry , Astrocytes/pathology , Membrane Glycoproteins/physiology , Oligodendroglia/pathology , Receptors, Immunologic/physiology , T-Lymphocytes/immunology , tau Proteins/metabolism , Alzheimer Disease/immunology , Alzheimer Disease/metabolism , Animals , Astrocytes/immunology , Astrocytes/metabolism , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodendroglia/immunology , Oligodendroglia/metabolism
11.
Viruses ; 13(12)2021 11 25.
Article in English | MEDLINE | ID: mdl-34960633

ABSTRACT

The environment of the central nervous system (CNS) represents a double-edged sword in the context of viral infections. On the one hand, the infectious route for viral pathogens is restricted via neuroprotective barriers; on the other hand, viruses benefit from the immunologically quiescent neural environment after CNS entry. Both the herpes simplex virus (HSV) and the rabies virus (RABV) bypass the neuroprotective blood-brain barrier (BBB) and successfully enter the CNS parenchyma via nerve endings. Despite the differences in the molecular nature of both viruses, each virus uses retrograde transport along peripheral nerves to reach the human CNS. Once inside the CNS parenchyma, HSV infection results in severe acute inflammation, necrosis, and hemorrhaging, while RABV preserves the intact neuronal network by inhibiting apoptosis and limiting inflammation. During RABV neuroinvasion, surveilling glial cells fail to generate a sufficient type I interferon (IFN) response, enabling RABV to replicate undetected, ultimately leading to its fatal outcome. To date, we do not fully understand the molecular mechanisms underlying the activation or suppression of the host inflammatory responses of surveilling glial cells, which present important pathways shaping viral pathogenesis and clinical outcome in viral encephalitis. Here, we compare the innate immune responses of glial cells in RABV- and HSV-infected CNS, highlighting different viral strategies of neuroprotection or Neuroinflamm. in the context of viral encephalitis.


Subject(s)
Encephalitis, Viral/immunology , Herpes Simplex/immunology , Immunity, Innate , Inflammation , Rabies virus/immunology , Rabies/immunology , Simplexvirus/immunology , Animals , Astrocytes/immunology , Astrocytes/virology , Blood-Brain Barrier/virology , Central Nervous System/immunology , Central Nervous System/virology , Encephalitis, Viral/virology , Herpes Simplex/virology , Humans , Microglia/immunology , Microglia/virology , Neuroglia/immunology , Neuroglia/virology , Rabies/virology , Signal Transduction
12.
Sci Rep ; 11(1): 23569, 2021 12 07.
Article in English | MEDLINE | ID: mdl-34876649

ABSTRACT

Microvascular compression of the trigeminal root entry zone (TREZ) is the main cause of most primary trigeminal neuralgia (TN), change of glial plasticity was previously studied in the TREZ of TN rat model induced by chronic compression. To better understand the role of astrocytes and immune cells in the TREZ, different cell markers including glial fibrillary acidic protein (GFAP), complement C3, S100A10, CD45, CD11b, glutamate-aspartate transporter (GLAST), Iba-1 and TMEM119 were used in the TN rat model by immunohistochemistry and flow cytometry. On the post operation day 28, GFAP/C3-positive A1 astrocytes and GFAP/S100A10-positive A2 astrocytes were activated in the TREZ after compression injury, there were no statistical differences in the ratios of A1/A2 astrocytes between the sham and TN groups. There was no significant difference in Iba-1-positive cells between the two groups. The ratios of infiltrating lymphocytes (CD45+CD11b-) (p = 0.0075) and infiltrating macrophages (CD45highCD11b+) (p = 0.0388) were significantly higher than those of the sham group. In conclusion, different subtypes A1/A2 astrocytes in the TREZ were activated after compression injury, infiltrating macrophages and lymphocytes increased, these neuroimmune cells in the TREZ may participate in the pathogenesis of TN rat model.


Subject(s)
Trigeminal Neuralgia/immunology , Trigeminal Neuralgia/pathology , Animals , Annexin A2/metabolism , Astrocytes/immunology , Astrocytes/metabolism , Astrocytes/pathology , Complement C3/metabolism , Disease Models, Animal , Flow Cytometry , Glial Fibrillary Acidic Protein/metabolism , Lymphocytes/immunology , Lymphocytes/metabolism , Lymphocytes/pathology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Neuroglia/immunology , Neuroglia/metabolism , Neuroglia/pathology , Rats , Rats, Sprague-Dawley , S100 Proteins/metabolism , Trigeminal Nerve/immunology , Trigeminal Nerve/metabolism , Trigeminal Nerve/pathology
13.
Int J Mol Sci ; 22(23)2021 Nov 26.
Article in English | MEDLINE | ID: mdl-34884629

ABSTRACT

In recent years, much attention has been paid to the study of the therapeutic effect of the microelement selenium, its compounds, especially selenium nanoparticles, with a large number of works devoted to their anticancer effects. Studies proving the neuroprotective properties of selenium nanoparticles in various neurodegenerative diseases began to appear only in the last 5 years. Nevertheless, the mechanisms of the neuroprotective action of selenium nanoparticles under conditions of ischemia and reoxygenation remain unexplored, especially for intracellular Ca2+ signaling and neuroglial interactions. This work is devoted to the study of the cytoprotective mechanisms of selenium nanoparticles in the neuroglial networks of the cerebral cortex under conditions of ischemia/reoxygenation. It was shown for the first time that selenium nanoparticles dose-dependently induce the generation of Ca2+ signals selectively in astrocytes obtained from different parts of the brain. The generation of these Ca2+ signals by astrocytes occurs through the release of Ca2+ ions from the endoplasmic reticulum through the IP3 receptor upon activation of the phosphoinositide signaling pathway. An increase in the concentration of cytosolic Ca2+ in astrocytes leads to the opening of connexin Cx43 hemichannels and the release of ATP and lactate into the extracellular medium, which trigger paracrine activation of the astrocytic network through purinergic receptors. Incubation of cerebral cortex cells with selenium nanoparticles suppresses ischemia-induced increase in cytosolic Ca2+ and necrotic cell death. Activation of A2 reactive astrocytes exclusively after ischemia/reoxygenation, a decrease in the expression level of a number of proapoptotic and proinflammatory genes, an increase in lactate release by astrocytes, and suppression of the hyperexcitation of neuronal networks formed the basis of the cytoprotective effect of selenium nanoparticles in our studies.


Subject(s)
Astrocytes/cytology , Calcium/metabolism , Gliosis/drug therapy , Nanoparticles/administration & dosage , Neuroprotective Agents/administration & dosage , Reperfusion Injury/prevention & control , Selenium/administration & dosage , Animals , Antioxidants/administration & dosage , Antioxidants/chemistry , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/metabolism , Calcium Signaling , Gliosis/immunology , Gliosis/metabolism , Gliosis/pathology , Nanoparticles/chemistry , Neurons/drug effects , Neurons/immunology , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/chemistry , Rats , Reperfusion Injury/etiology , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Selenium/chemistry
14.
J Neuroinflammation ; 18(1): 252, 2021 Nov 02.
Article in English | MEDLINE | ID: mdl-34727934

ABSTRACT

OBJECTIVE: Astrocytes participate in the local innate immune response of the central nervous system. In response to stress such as ischemia, activated cells release endogenous factors known as damage-associated molecular patterns (DAMPs). Self-extracellular RNA (eRNA) is such a ubiquitous alarm signal. However, it is unclear whether eRNA is involved in the early acute phase of cerebral ischemia and is sufficient to sensitize astrocytes towards a DAMP or PAMP (pathogen-associated molecular pattern) reaction. METHODS: Pro-inflammatory activation upon eRNA stimulation was characterized in primary murine astrocyte cultures. In vivo, an experimental stroke model was used to localize and quantify eRNA in murine brain sections. Using primary cortical neurons and the mouse hippocampal neuronal cell line HT-22, neuronal RNA release upon stress conditions related to cerebral hypoxia/ischemia was analyzed. RESULTS: While low-dose eRNA alone did not promote pro-inflammatory activation of astrocytes in culture, it strongly enhanced the expression of pro-inflammatory cytokines in the presence of either Pam2CSK4, a synthetic PAMP molecule that mimics bacterial infection, or high mobility group box 1 (HMGB1), a prominent DAMP. Synergism of eRNA/Pam2CSK4 and eRNA/HMGB1 was prevented by blockage of the astroglial toll-like receptor (TLR)-2. Inhibition of NF-κB- and mitogen-activated protein kinase-dependent signaling pathways hampered eRNA/Pam2CSK4-mediated pro-inflammatory activation of astrocytes. In vivo, the amount of non-nuclear, presumably extracellular ribosomal RNA in close proximity to neurons significantly accumulated across the infarct core and peri-infarct areas that was accompanied by transcriptional up-regulation of various pro-inflammatory factors. Accordingly, the exposure of neurons to hypoxic/ischemic stress in vitro resulted in the release of eRNA, partly mediated by active cellular processes dependent on the cytosolic calcium level. CONCLUSION: The DAMP signal eRNA can sensitize astrocytes as active players in cerebral innate immunity towards exogenous and endogenous activators of inflammation (PAMPs and DAMPs) in a synergistic manner via TLR2-NF-κB-dependent signaling mechanisms. These findings provide new insights into the pathogenesis of ischemic stroke and other inflammatory neurological disorders. Further studies will clarify whether administration of RNase in vivo may serve as an effective treatment for inflammatory brain pathologies.


Subject(s)
Alarmins/immunology , Astrocytes/immunology , Inflammation/immunology , RNA/immunology , Stroke/immunology , Animals , Mice , Stroke/pathology
15.
J Neuroimmunol ; 360: 577718, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34600199

ABSTRACT

Autoimmune glial fibrillary acidic protein (GFAP) astrocytopathy is a recently defined autoimmune meningoencephalomyelitis, associated with GFAP-IgG antibody. A pooled analysis of 324 cases from published literature and a retrospective single-center study were performed, firstly reveals the possibility that patients with myelitic lesions respond better to initial immunotherapy, but are prone to relapse, suggesting a more aggressive and long-term immunosuppressive medication for them. Moreover, our results showed using tacrolimus at maintenance stage exhibited a less tendency to relapse, providing a possibly new choice to future clinical treatments.


Subject(s)
Astrocytes/immunology , Autoantigens/immunology , Autoimmune Diseases of the Nervous System/epidemiology , Glial Fibrillary Acidic Protein/immunology , Adolescent , Adrenal Cortex Hormones/therapeutic use , Adult , Aged , Aged, 80 and over , Astrocytes/pathology , Autoimmune Diseases of the Nervous System/diagnostic imaging , Autoimmune Diseases of the Nervous System/drug therapy , Child , Child, Preschool , China/epidemiology , Encephalomyelitis/diagnosis , Encephalomyelitis/drug therapy , Encephalomyelitis/epidemiology , Encephalomyelitis/immunology , Female , Follow-Up Studies , Humans , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/therapeutic use , Maintenance Chemotherapy , Male , Meningoencephalitis/diagnosis , Meningoencephalitis/drug therapy , Meningoencephalitis/epidemiology , Meningoencephalitis/immunology , Middle Aged , Prognosis , Recurrence , Retrospective Studies , Tacrolimus/therapeutic use , Young Adult
16.
Int Immunopharmacol ; 101(Pt A): 108181, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34607229

ABSTRACT

Demyelinating diseases of the central nervous system are characterized by recurrent demyelination and progressive neurodegeneration, but there are no clinical drugs targeting myelin regeneration or improving functional disability in the treatment of multiple sclerosis. Total flavone of Epimedium (TFE) is the main active components of Epimedium, which exhibits the beneficial biological activities in the treatment of diseases, but there is no report in the treatment of demyelinating disorder. The purpose of this study was to explore the therapeutic potential and possible mechanism of TFE in the treatment of demyelination. The results showed that TFE efficiently improved the behavioural performance and histological demyelination in cuprizone (CPZ)-induced demyelinating model. In terms of action, TFE increased astrocytes enrichment in corpus callosum, striatum and cortex, and promoted astrocytes to express neurotrophic factors. Furthermore, the expression of platelet-activating factor receptor (PAFR) in astrocytes was induced by CPZ feeding and LPS stimulation, accompanied by the increase of inflammatory cytokines TNF-α,IL-6 and IL-1ß. TFE declined the expression of PAFR, and inhibited inflammatory response. At the same time, TFE also antagonized PAFR activation and inflammatory response triggered by PAF, which further confirmed that TFE, as a new PAFR antagonist, inhibited the astrocyte-derived inflammatory response by antagonizing PAFR-neuroinflammation axis, thus contributing to myelin protection and regeneration.


Subject(s)
Demyelinating Diseases/drug therapy , Epimedium/chemistry , Neuroinflammatory Diseases/drug therapy , Plant Extracts/pharmacology , Platelet Membrane Glycoproteins/antagonists & inhibitors , Receptors, G-Protein-Coupled/antagonists & inhibitors , Administration, Oral , Animals , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/metabolism , Cuprizone/administration & dosage , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Demyelinating Diseases/immunology , Demyelinating Diseases/pathology , Disease Models, Animal , Flavones/pharmacology , Flavones/therapeutic use , Humans , Male , Mice , Myelin Sheath/drug effects , Myelin Sheath/immunology , Myelin Sheath/pathology , Neuroinflammatory Diseases/chemically induced , Neuroinflammatory Diseases/immunology , Neuroinflammatory Diseases/pathology , Plant Extracts/therapeutic use
17.
J Neuroimmunol ; 361: 577748, 2021 12 15.
Article in English | MEDLINE | ID: mdl-34653948

ABSTRACT

We report two patients with meningoencephalomyelitis without evidence of extra central nervous system (CNS) involvement. Brain MRI showed linear perivascular radial gadolinium enhancement patterns and spinal cord MRI showed longitudinal extensive T2-hyperintensity lesions. Pathological findings from brain biopsies were angiocentric T-cell predominant lymphoid infiltrates that lacked Epstein-Barr virus-positive atypical B cells. The patients were initially suspected to have isolated CNS-lymphomatoid granulomatosis (LYG). Thereafter, glial fibrillary acidic protein (GFAP)-immunoglobulin G were detected in their cerebrospinal fluid. This finding suggested autoimmune GFAP astrocytopathy. We speculate there is a link between isolated CNS-LYG and autoimmune GFAP astrocytopathy.


Subject(s)
Astrocytes/pathology , Autoimmune Diseases of the Nervous System/diagnosis , Glial Fibrillary Acidic Protein/immunology , Lymphomatoid Granulomatosis/diagnosis , Adrenal Cortex Hormones/therapeutic use , Aged , Antibody Specificity , Astrocytes/immunology , Autoantibodies/cerebrospinal fluid , Autoimmune Diseases of the Nervous System/cerebrospinal fluid , Autoimmune Diseases of the Nervous System/immunology , Autoimmune Diseases of the Nervous System/pathology , Brain/diagnostic imaging , Brain/immunology , Brain/pathology , Diagnosis, Differential , Humans , Immunoglobulin G/cerebrospinal fluid , Immunoglobulins, Intravenous/therapeutic use , Magnetic Resonance Imaging , Male , Meningoencephalitis/etiology , Middle Aged , Myelitis/etiology , Neuroimaging , Retrospective Studies , Spinal Cord/diagnostic imaging , Spinal Cord/immunology , Spinal Cord/pathology
18.
Neurosci Lett ; 765: 136284, 2021 11 20.
Article in English | MEDLINE | ID: mdl-34624394

ABSTRACT

Depression is a kind of common mental disorder associated with neuroinflammation, and astrocytes play a vital role in regulating and mediating neuroinflammation in central nervous system. Scutellarin has significant anti-inflammatory and neuroprotective effects. However, whether scutellarin exerts antidepressant effect remains unknown. In present study, it was found that scutellarin suppressed LPS-induced neuroinflammation in the hippocampus and alleviated depression-like behaviors in mice. In addition, scutellarin inhibited LPS-induced elevation of TNFα, IL-1ß, IL-6 and iNOS, and reversed the downregulation of IL-4 and BDNF in astrocytes in vitro. Furthermore, the activated TLR4/NF-κB pathway in LPS-treated astrocytes was suppressed by scutellarin. Collectively, these results suggest that scutellarin ameliorates depression-like behaviors induced by neuroinflammation partially through inhibiting the TLR4/NF-κB pathway in astrocytes.


Subject(s)
Apigenin/pharmacology , Astrocytes/drug effects , Astrocytes/immunology , Depression/immunology , Glucuronates/pharmacology , Neuroinflammatory Diseases/immunology , Animals , Anti-Inflammatory Agents/pharmacology , Hippocampus/drug effects , Lipopolysaccharides/immunology , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL
19.
Acta Neuropathol ; 142(5): 899-915, 2021 11.
Article in English | MEDLINE | ID: mdl-34487221

ABSTRACT

Multiple sclerosis (MS) is an inflammatory demyelinating disease of the central nervous system (CNS) characterized by varying degrees of secondary neurodegeneration. Retinal ganglion cells (RGC) are lost in MS in association with optic neuritis but the mechanisms of neuronal injury remain unclear. Complement component C3 has been implicated in retinal and cerebral synaptic pathology that may precede neurodegeneration. Herein, we examined post-mortem MS retinas, and then used a mouse model, experimental autoimmune encephalomyelitis (EAE), to examine the role of C3 in the pathogenesis of RGC loss associated with optic neuritis. First, we show extensive C3 expression in astrocytes (C3+/GFAP+ cells) and significant RGC loss (RBPMS+ cells) in post-mortem retinas from people with MS compared to retinas from non-MS individuals. A patient with progressive MS with a remote history of optic neuritis showed marked reactive astrogliosis with C3 expression in the inner retina extending into deeper layers in the affected eye more than the unaffected eye. To study whether C3 mediates retinal degeneration, we utilized global C3-/- EAE mice and found that they had less RGC loss and partially preserved neurites in the retina compared with C3+/+ EAE mice. C3-/- EAE mice had fewer axonal swellings in the optic nerve, reflecting reduced axonal injury, but had no changes in demyelination or T cell infiltration into the CNS. Using a C3-tdTomato reporter mouse line, we show definitive evidence of C3 expression in astrocytes in the retina and optic nerves of EAE mice. Conditional deletion of C3 in astrocytes showed RGC protection replicating the effects seen in the global knockouts. These data implicate astrocyte C3 expression as a critical mediator of retinal neuronal pathology in EAE and MS, and are consistent with recent studies showing C3 gene variants are associated with faster rates of retinal neurodegeneration in human disease.


Subject(s)
Complement C3/metabolism , Multiple Sclerosis/pathology , Neuroinflammatory Diseases/pathology , Retinal Ganglion Cells/pathology , Animals , Astrocytes/immunology , Astrocytes/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Humans , Mice , Multiple Sclerosis/immunology , Nerve Degeneration/immunology , Nerve Degeneration/pathology , Neuroinflammatory Diseases/immunology , Optic Nerve/pathology , Optic Neuritis/immunology , Optic Neuritis/pathology
20.
J Neuroinflammation ; 18(1): 208, 2021 Sep 16.
Article in English | MEDLINE | ID: mdl-34530847

ABSTRACT

Neuromyelitis optica (NMO) is a chronic inflammatory autoimmune disease of the central nervous system (CNS) characterized by acute optic neuritis (ON) and transverse myelitis (TM). NMO is caused by a pathogenic serum IgG antibody against the water channel aquoporin 4 (AQP4) in the majority of patients. AQP4-antibody (AQP4-ab) presence is highly specific, and differentiates NMO from multiple sclerosis. It binds to AQP4 channels on astrocytes, triggering activation of the classical complement cascade, causing granulocyte, eosinophil, and lymphocyte infiltration, culminating in injury first to astrocyte, then oligodendrocytes followed by demyelination and neuronal loss. NMO spectrum disorder (NMOSD) has recently been defined and stratified based on AQP4-ab serology status. Most NMOSD patients experience severe relapses leading to permanent neurologic disability, making suppression of relapse frequency and severity, the primary objective in disease management. The most common treatments used for relapses are steroids and plasma exchange.Currently, long-term NMOSD relapse prevention includes off-label use of immunosuppressants, particularly rituximab. In the last 2 years however, three pivotal clinical trials have expanded the spectrum of drugs available for NMOSD patients. Phase III studies have shown significant relapse reduction compared to placebo in AQP4-ab-positive patients treated with satralizumab, an interleukin-6 receptor (IL-6R) inhibitor, inebilizumab, an antibody against CD19+ B cells; and eculizumab, an antibody blocking the C5 component of complement. In light of the new evidence on NMOSD pathophysiology and of preliminary results from ongoing trials with new drugs, we present this descriptive review, highlighting promising treatment modalities as well as auspicious preclinical and clinical studies.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Autoantibodies/metabolism , Immunosuppressive Agents/therapeutic use , Neuromyelitis Optica/drug therapy , Neuromyelitis Optica/metabolism , Animals , Antibodies, Monoclonal, Humanized/pharmacology , Aquaporin 4/immunology , Aquaporin 4/metabolism , Astrocytes/drug effects , Astrocytes/immunology , Astrocytes/metabolism , Autoantibodies/drug effects , Autoantibodies/immunology , Azathioprine/pharmacology , Azathioprine/therapeutic use , Clinical Trials, Phase III as Topic/methods , Humans , Immunosuppressive Agents/pharmacology , Neuromyelitis Optica/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...