Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
J Neurosci ; 2024 Aug 28.
Article in English | MEDLINE | ID: mdl-39197941

ABSTRACT

Abnormal neuronal morphological features, such as dendrite branching, axonal branching, and spine density, is thought to contribute to the symptoms of depression and anxiety. However, the role and molecular mechanisms of aberrant neuronal morphology in the regulation of mood disorders remain poorly characterized. Here, we show that Neuritin, an activity-dependent protein, regulates the axonal morphology of serotonin neurons. Male neuritin knockout mice harbored impaired axonal branches of serotonin neurons in the medial prefrontal cortex and basolateral region of the amygdala (BLA), and male neuritin knockout mice exhibited depressive and anxiety-like behaviors. We also observed that the expression of Neuritin was decreased by unpredictable chronic stress (UCS) in the male mouse brain and that decreased expression of neuritin was associated with reduced axonal branching of serotonin neurons in the brain and with depressive and anxiety behaviors in mice. Furthermore, the stress-mediated impairments in axonal branching and depressive behaviors were reversed by the overexpression of Neuritin in the BLA. The ability of neuritin to increase axonal branching in serotonin neurons involves FGF signaling, and neuritin contributes to FGF-2-mediated axonal branching regulation in vitro Finally, the oral administration of an FGF inhibitor reduced the axonal branching of serotonin neurons in the brain and caused depressive and anxiety behaviors in male mice. Our results support the involvement of neuritin in models of stress-induced depression and suggest that neuronal morphological plasticity may play a role in controlling animal behavior.Significance statement Axonal atrophy of serotonin neurons is one of the representative neuroanatomical features of depression. We found that the secreted/membrane-anchored neurotrophic factor Neuritin regulated axonal branch formation, which is involved in the development of depression and anxiety. In addition, Neuritin and the secreted signaling protein fibroblast growth factor 2 (FGF-2) cooperate to promote axonal branching in serotonin neurons. Furthermore, the inhibition of FGF signaling promoted axonal branching impairments and depressive behavior in mice. Taken together, these findings suggest that Neuritin regulates axonal branching in serotonin neurons and that the loss of neuritin is related to the development of depression. FGF signaling is involved in the neuritin-mediated axonal branching of serotonin neurons.

2.
Dev Med Child Neurol ; 2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38641898

ABSTRACT

AIM: To modify the antibody prevalence in epilepsy (APE) score of children with suspected autoimmune central nervous system disease with seizures. METHODS: We retrospectively analysed the cerebrospinal fluid of 157 children (aged 0-18 years) with suspected autoimmune central nervous system disease for antineuronal antibodies in our laboratory from 2016 to 2023. Participants were randomly divided into the development cohort (n = 79, 35 females; median 7 years, SD 4 years 7 months, range 4-11 years) and validation cohort (n = 78, 28 females; median 7 years, SD 4 years 5 months, range 4-12 years). A paediatric antibody prevalence in seizure (PAPS) score was created for one cohort and evaluated in the other. Seven variables were selected through univariate and multivariate analysis to create a PAPS score. RESULTS: One hundred and fifty-seven children who fulfilled the inclusion criteria were enrolled; 49 tested positive for antineuronal antibodies. The sensitivity and specificity of an APE score of 4 and greater were 92% and 22.2% respectively; the sensitivity and specificity of a PAPS score of 2.5 and greater were 83.3% and 77.8% respectively. The area under the curve was 0.832 (95% confidence interval = 0.743-0.921), which was significantly better than that for the APE score (p < 0.001). INTERPRETATION: The APE score had high sensitivity but low specificity in children. The PAPS score may be useful for determining the need for antineuronal antibody testing.

3.
Brain Sci ; 14(3)2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38539632

ABSTRACT

Hypoxic-ischemic brain injury induces metabolic dysfunction that ultimately leads to neuronal cell death. Astrocytes, a type of glial cell, play a key role in brain metabolism; however, their response to hypoxic-ischemic brain injury is not fully understood. Microglia were removed from murine primary mixed glial cultures to enrich astrocytes. Next, we explored genes whose expression is altered following oxygen-glucose deprivation using a microarray. Microarray analysis revealed that the expression of Nr4a1 and Nr4a3 is markedly increased in astrocyte-enriched cultures after 15 h of oxygen-glucose deprivation. The expression of both Nr4a1 and Nr4a3 was regulated by HIF-1α. At the protein level, NR4A1 was translocated from the nucleus to the cytoplasm following oxygen-glucose deprivation and co-localized with mitochondria in apoptotic cells; however, its localization was restored to the nucleus after reoxygenation. Oxygen-glucose deprivation causes an increase in NR4A1 mRNA in astrocytes as well as its nuclear to cytoplasmic transfer. Furthermore, reoxygenation enhances NR4A1 transcription and promotes its nuclear translocation.

4.
Brain Res ; 1815: 148463, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37328088

ABSTRACT

AIMS: Glutamate-induced excitotoxicity is mainly mediated by neuronal NMDA receptors; however, it is unclear how astrocytes are involved in this phenomenon. This study aimed to explore the effects of excess glutamate on astrocytes both in vitro and in vivo. METHODS: We used astrocyte-enriched cultures (AECs), in which microglia were removed from mixed glial cultures, to investigate the effects of extracellular glutamate on these cells by microarray, quantitative PCR, ELISA, and immunostaining. We also examined the production of lipocalin-2 (Lcn2) by immunohistochemistry in the brains of mice after status epilepticus induced by pilocarpine and by ELISA in the cerebrospinal fluid (CSF) of patients characterised by status epilepticus. RESULTS: Microarray analysis identified Lcn2 as a factor upregulated in AECs by excess glutamate; glutamate addition increased Lcn2 in the cytoplasm of astrocytes and AECs released Lcn2 in a concentration-dependent manner. Lcn2 production was reduced by chemical inhibition of metabotropic glutamate receptor or siRNA knockdown of metabotropic glutamate receptor 3. Furthermore, Lcn2 was increased in the astrocytes of a status epilepticus mouse model and in the CSF of human patients. CONCLUSION: These results indicate that astrocytes stimulate Lcn2 production via metabotropic glutamate receptor 3 in response to high concentrations of glutamate.


Subject(s)
Astrocytes , Status Epilepticus , Humans , Mice , Animals , Lipocalin-2/genetics , Glutamic Acid/pharmacology , Neuroglia , Status Epilepticus/chemically induced
5.
Front Neurosci ; 17: 1014071, 2023.
Article in English | MEDLINE | ID: mdl-36816137

ABSTRACT

Background: The autoantibody to myelin oligodendrocyte glycoprotein (MOG), a component of the central nervous system myelin, has been identified in a subset of demyelinating diseases. However, there is no convincing evidence to support the direct pathogenic contribution of this autoantibody. Objective: To elucidate the role of anti-MOG autoantibodies in human demyelinating disorders, we assessed the effect of autoantibodies on MOG-expressing cells. Methods: Mammalian cells expressing the human MOG protein reacted with human anti-MOG autoantibodies in the presence or absence of complement. Sera from 86 patients and 11 healthy sera were used. We analyzed anti-MOG antibody titers, IgG subclass, and their cytotoxic ability in sera from patients with various neurological diseases. Membrane attack complex (MAC) formation was examined by detection of complement C9 or C9neo with western blot or flow cytometry. Results: Among 86 patients, 40 were determined to be MOG-IgG-positive and 46 were negative. Anti-MOG-positive sera, but not -negative sera, caused cell death in MOG-expressing cells. This cytotoxic effect was disappeared after heat inactivation of sera. Importantly, anti-MOG IgG and externally added complement were necessary for sufficient cytotoxic effects. Anti-MOG autoantibodies were histologically colocalized with complement and formed a membrane attack complex consisting of anti-MOG IgG and complement factors. Conclusion: The human MOG antibody specifically killed MOG-expressing cells in vitro in the presence of externally added complement. Membrane attack complexes were formed on the cells, indicating that this autoantibody activated complement-mediated cytotoxicity. Further studies in larger numbers of patients are needed to characterize the role of complement in MOGAD.

6.
Neurology ; 96(16): e2070-e2077, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33653900

ABSTRACT

OBJECTIVE: To evaluate the validity of the 2016 clinical diagnostic criteria proposed for probable anti-NMDA receptor (NMDAR) encephalitis in children, we tested the criteria in a Japanese pediatric cohort. METHODS: We retrospectively reviewed clinical information of patients with neurologic symptoms whose CSF was analyzed for NMDAR antibodies (NMDAR-Abs) in our laboratory from January 1, 2015, to March 31, 2019. RESULTS: Overall, 137 cases were included. Of the 41 cases diagnosed as probable anti-NMDAR encephalitis (criteria-positive) according to the 2016 criteria, 13 were positive and 28 were negative for anti-NMDAR-Abs. Of the 96 criteria-negative cases, 3 were positive and 93 were negative for anti-NMDAR-Abs. The sensitivity of the criteria was 81.2%, specificity was 76.9%, positive predictive value (PPV) was 31.7%, and negative predictive value was 96.9%. Compared with the true-positive group, the false-positive group contained more male than female patients (male:female, 4:9 in the true-positive vs 19:9 in the false-positive group, p = 0.0425). The majority of the cases with false-positive diagnoses were associated with neurologic autoimmunity. CONCLUSION: The clinical diagnostic criteria are reliable for deciding to start immunomodulatory therapy in the criteria-positive cases. Low PPV may be caused by a lower prevalence of NMDAR encephalitis or lower level of suspicion for encephalitis in the pediatric population. Physicians should therefore continue differential diagnosis, focusing especially on other forms of encephalitis. CLASSIFICATION OF EVIDENCE: This study provides Class IV evidence that the proposed diagnostic criteria for anti-NMDAR encephalitis in children has a sensitivity of 81.2% and a specificity of 76.9%.


Subject(s)
Anti-N-Methyl-D-Aspartate Receptor Encephalitis/diagnosis , Adolescent , Autoantibodies/immunology , Autoantigens/immunology , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Infant, Newborn , Japan , Male , Retrospective Studies , Sensitivity and Specificity
7.
Eur J Immunol ; 50(2): 205-219, 2020 02.
Article in English | MEDLINE | ID: mdl-31549730

ABSTRACT

The NLRP3 inflammasome is a molecular complex that translates signals from pathogens and tissue damage into inflammatory responses, and plays crucial roles in numerous neurological diseases. Activation of the NLRP3 inflammasome leads to caspase-1 dependent cleavage of pro-IL-1ß to form mature IL-1ß. By acting on the P2X7 purinergic receptor, extracellular ATP is one of the major stimuli that activates the NLRP3 inflammasome. Although microglia express multiple purinergic receptors, their roles in inflammasome-mediated inflammation are largely unknown. We studied the role of the P2Y12 receptor, a metabotropic P2Y receptor enriched in microglia, on inflammation in vitro. Inhibition of the microglial P2Y12 receptor by PSB0739 or siRNA knockdown suppressed IL-1ß release. P2Y12 receptor-deficient microglia displayed markedly attenuated IL-1ß mRNA expression and release. P2Y12 receptor blockade also suppressed IL-6 production. Both IL-1ß and IL-6 responses were augmented by extracellular ADP or ADP-ßS and were abrogated by PSB0739. Mechanistically, ADP-ßS potentiated NF-κB activation. In addition, ADP altered mitochondrial membrane potential in combination with ATP and increased the number of caspase-1 positive cells through the P2Y12 receptor. These results elucidate a novel inflammatory mechanism by which extracellular ADP acts on the P2Y12 receptor to activate NF-κB and the NLRP3 inflammasome to enhance microglial inflammation.


Subject(s)
Adenosine Diphosphate/metabolism , Inflammasomes/metabolism , NF-kappa B/metabolism , Receptors, Purinergic P2Y12/metabolism , Animals , Caspase 1/metabolism , Cell Line , Cytokines/metabolism , Gene Expression Regulation/physiology , Inflammation/metabolism , Inflammation Mediators/metabolism , Interleukin-1beta/metabolism , Macrophages/metabolism , Mice , Reactive Oxygen Species/metabolism , Signal Transduction/physiology
8.
Immunotargets Ther ; 4: 27-34, 2015.
Article in English | MEDLINE | ID: mdl-27471709

ABSTRACT

Alzheimer's disease (AD) is characterized by progressive neurodegeneration and is the most common cause of dementia. Immunotherapy has recently been regarded as a potential treatment for AD. This stems from the fact that the clinical and pathological findings from the active AD vaccine trial suggests that such vaccine therapy may be effective for AD. However, this trial was halted because of the occurrence of meningoencephalitis in some patients. Avoiding excessive immune reaction is necessary for the success of vaccine therapy. For this purpose, adjuvant-free vaccine therapies (eg, passive immunization or DNA vaccines) are currently under investigation. However, the results of clinical trials employing both active and passive anti-amyloid-beta immunotherapy have been unsatisfactory. In this article, we will analyze the reasons for the limited efficacy of currently available immunotherapies and discuss the effectiveness of new vaccine therapies. Finally, we will speculate on the possibility of its clinical application.

9.
PLoS One ; 8(9): e75203, 2013.
Article in English | MEDLINE | ID: mdl-24086465

ABSTRACT

It has recently been determined that not only Aß oligomers, but also other Aß species and amyloidogenic peptides are neurotoxic in Alzheimer disease (AD) and play a pivotal role in AD pathogenesis. In the present study, we attempted to develop new DNA vaccines targeting a wide range of Aß species. For this purpose, we first performed in vitro assays with newly developed vaccines to evaluate Aß production and Aß secretion abilities and then chose an IgL-Aßx4-Fc-IL-4 vaccine (designated YM3711) for further studies. YM3711 was vaccinated to mice, rabbits and monkeys to evaluate anti-Aß species antibody-producing ability and Aß reduction effects. It was found that YM3711 vaccination induced significantly higher levels of antibodies not only to Aß1-42 but also to AD-related molecules including AßpE3-42, Aß oligomers and Aß fibrils. Importantly, YM3711 significantly reduced these Aß species in the brain of model mice. Binding and competition assays using translated YM3711 protein products clearly demonstrated that a large part of antibodies induced by YM3711 vaccination are directed at conformational epitopes of the Aß complex and oligomers. Taken together, we demonstrate that YM3711 is a powerful DNA vaccine targeting a wide range of AD-related molecules and is worth examining in preclinical and clinical trials.


Subject(s)
Alzheimer Disease/prevention & control , Alzheimer Vaccines/immunology , Amyloid beta-Peptides/metabolism , Amyloidogenic Proteins/metabolism , Antibodies/immunology , Brain/metabolism , Vaccines, DNA/immunology , Alzheimer Vaccines/metabolism , Amyloid beta-Peptides/immunology , Animals , Blotting, Western , Brain/pathology , Immunohistochemistry , Macaca fascicularis , Mice , Rabbits , Statistics, Nonparametric , Vaccines, DNA/metabolism
10.
Neuropathology ; 33(6): 612-20, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23581743

ABSTRACT

Matrix metalloproteinases (MMPs) that are secreted by activated T cells play a significant role in degradation of the extracellular matrix around the blood vessels and facilitate autoimmune neuroinflammation; however, it remains unclear how MMPs act in lesion formation and whether MMP-targeted therapies are effective in disease suppression. In the present study, we attempted to treat experimental autoimmune encephalomyelitis (EAE) by administration of small interfering RNAs (siRNAs) for MMP-2, MMP-9, and minocycline, all of which have MMP-inhibiting functions. Minocycline, but not siRNAs, significantly suppressed disease development. In situ zymography revealed that gelatinase activities were almost completely suppressed in the spinal cords of minocycline-treated animals, while significant gelatinase activities were measured in the EAE lesions of control animals. However, MMP-2 and MMP-9 mRNAs and proteins in the spinal cords of treated rats were unexpectedly upregulated. At the same time, mRNA for tissue inhibitors of MMPs (TIMP)-1 and -2 were also upregulated. The EnzChek Gelatinase/Collagenase assay using tissue containing native MMPs and TIMPs demonstrated that gelatinase activity levels in the spinal cords of treated rats were suppressed to the same level as those in normal spinal cord tissues. Finally, double immunofluorescent staining demonstrated that MMP-9 immunoreactivities of treated rats were almost the same as those of control rats and that MMP-9 and TIMP-1 immunoreactivities were colocalized in the spinal cord. These findings suggest that minocycline administration does not suppress MMPs at mRNA and protein levels but that it suppresses gelatinase activities by upregulating TIMPs. Thus, MMP-targeted therapies should be designed after the mechanisms of candidate drugs have been considered.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/enzymology , Enzyme Inhibitors/pharmacology , Minocycline/pharmacology , Spinal Cord/enzymology , Tissue Inhibitor of Metalloproteinases/biosynthesis , Animals , Gelatinases/biosynthesis , Matrix Metalloproteinases/biosynthesis , RNA, Small Interfering , Rats , Rats, Inbred Lew , Real-Time Polymerase Chain Reaction , Spinal Cord/drug effects , Up-Regulation
11.
J Neuroimmunol ; 254(1-2): 39-45, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-22989513

ABSTRACT

Gene silencing with siRNAs is important as a therapeutic tool in autoimmune diseases. In this study, we administered siRNAs specific for cytokines that may be involved in pathogenesis of experimental autoimmune encephalomyelitis (EAE). siRNA specific for IL-23p19 (siRNA-IL-23) suppressed EAE almost completely, whereas siRNA-IL-17A did not modulate the clinical course. Flow cytometric analysis revealed that siRNA-IL-23 significantly reduced the proportion of both IFN-γ(+)IL-17(-) Th1 and IFN-γ(-)IL-17(+) Th17 cells in the spinal cord. Consistent with this finding, siRNA-IL-23 treatment downregulated IL-12, IL-17 and IL-23 mRNAs. These findings indicate that IL-23, but not IL-17, play an important role in the development of EAE.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/therapy , Interleukin-12/metabolism , Interleukin-17/metabolism , RNA, Small Interfering/therapeutic use , Analysis of Variance , Animals , Cytokines/genetics , Cytokines/metabolism , Disease Models, Animal , Drug Delivery Systems , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Flow Cytometry , Freund's Adjuvant/adverse effects , Interleukin-12/genetics , Interleukin-17/genetics , Myelin Basic Protein/immunology , Myelin Basic Protein/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Inbred Lew , Severity of Illness Index , Spinal Cord/drug effects , Spinal Cord/metabolism , Statistics, Nonparametric , Th1 Cells/drug effects , Th1 Cells/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism , Time Factors
12.
Neurochem Int ; 62(3): 271-80, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23266396

ABSTRACT

Microsomal prostaglandin synthetase-1 (mPGES-1) is an inducible terminal enzyme required for prostaglandin E(2) (PGE(2)) biosynthesis. In this study, we examined the role of mPGES-1 in the inflammation and demyelination observed in experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS). We induced EAE with myelin oligodendrocyte glycoprotein(35-55) peptide in mPGES-1-deficient (mPGES-1(-/-)) and wild-type (WT) mice. First, we examined the histopathology in the early and late phases of EAE progression. Next, we measured the concentration of PGE(2) in the spinal cord and investigated the expression of mPGES-1 using immunohistochemistry. In addition, we examined the progression of the severity of EAE using an EAE score to investigate a correlation between pathological features and paralysis. In this paper, we demonstrate that WT mice showed extensive inflammation and demyelination, whereas mPGES-1(-/-) mice exhibited significantly smaller and more localized changes in the perivascular area. The mPGES-1 protein was induced in vascular endothelial cells and microglia around inflammatory foci, and PGE(2) production was increased in WT mice but not mPGES-1(-/-) mice. Furthermore, mPGES-1(-/-) mice showed a significant reduction in the maximum EAE score and improved locomotor activity. These results suggest that central PGE(2) derived from non-neuronal mPGES-1 aggravates the disruption of the vessel structure, leading to the spread of inflammation and local demyelination in the spinal cord, which corresponds to the symptoms of EAE. The inhibition of mPGES-1 may be useful for the treatment of human MS.


Subject(s)
Demyelinating Diseases/physiopathology , Disease Models, Animal , Inflammation/physiopathology , Intramolecular Oxidoreductases/metabolism , Microsomes/enzymology , Multiple Sclerosis/physiopathology , Animals , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Female , Immunoenzyme Techniques , Immunohistochemistry , Mice , Mice, Inbred C57BL , Prostaglandin-E Synthases
13.
Cell Immunol ; 279(1): 70-7, 2012 Sep.
Article in English | MEDLINE | ID: mdl-23099153

ABSTRACT

Due to poor proliferation abilities of cardiomyocytes, the repair process in the heart after insults is often associated with fibrosis formation. In this study, we characterized inflammation and/or fibrosis-related molecules in the heart with experimental autoimmune carditis. Immunohistochemical examinations reveled that expression of tenascin-C (TNC), matrix metalloproteinase-9 (MMP-9), transforming growth factorß1 (TGF-ß1), connective tissue growth factor (CTGF) and α smooth muscle cell actin (αSMA) peaked at 2 weeks post-immunization but only TGF-ß1 expression was sustained at 8 weeks. Administration of siRNAs for MMP-2 (siMMP-2) and for MMP-9 (siMMP-9) alone did not modulate inflammation and fibrosis. In contrast, simultaneous administration of siMMP-2 and siMMP-9 significantly reduced inflammation and fibrosis. Of note, siRNA treatment for TGF-ß1, which is an anti-inflammatory cytokine, increased inflammation and decreased fibrosis. These findings suggest that in case of diseases characterized by initial inflammation and subsequent fibrosis, immunotherapies should target inflammation, not fibrosis, because the latter therapies exacerbate inflammation.


Subject(s)
Autoimmune Diseases/immunology , Myocarditis/immunology , Myocardium/immunology , RNA Interference/immunology , Actins/immunology , Actins/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Carrier Proteins/chemistry , Carrier Proteins/immunology , Connective Tissue Growth Factor/genetics , Connective Tissue Growth Factor/immunology , Connective Tissue Growth Factor/metabolism , Female , Fibrosis , HEK293 Cells , Humans , Immunohistochemistry , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/immunology , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/immunology , Matrix Metalloproteinase 9/metabolism , Muscle, Smooth/chemistry , Myocarditis/genetics , Myocarditis/metabolism , Myocardium/metabolism , Myocardium/pathology , Peptide Fragments/immunology , Rats , Rats, Inbred Lew , Tenascin/genetics , Tenascin/immunology , Tenascin/metabolism , Time Factors , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/immunology , Transforming Growth Factor beta1/metabolism
14.
Neuropathology ; 31(2): 101-11, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20573031

ABSTRACT

It is important to find biomarkers for autoimmune inflammation and demyelination in the CNS to monitor disease status in patients with multiple sclerosis (MS). For this purpose, we determined the titers of antibodies (Ab) reacting with native myelin oligodendrocyte glycoprotein (MOG)-expressing cells to evaluate the disease activity of chronic experimental autoimmune encephalomyelitis (EAE) in rats and the relationship between anti-MOGcme (cell membrane-expressed MOG), Ab titers and clinical and pathological parameters were evaluated. Consequently, we found that elevation of anti-MOGcme Ab titers was associated with clinical severity, except for some cases in very late stages and with severe and widespread demyelination but with dominant inflammation. In contrast, antibodies detected by standard ELISA using recombinant MOG were elevated in both symptomatic and asymptomatic rats and were not associated with parameters such as inflammation and demyelination. Longitudinal examination of anti-MOGcme Ab titers in individual rats revealed that Ab titers accurately reflect disease activity. Furthermore, anti-MOGcme Ab titer was not elevated in acute EAE without demyelination. These findings suggest that autoantibodies reacting with native and glycosylated MOG play an important role in the progression of demyelinating diseases and could be biomarkers for monitoring the status of patients with MS.


Subject(s)
Autoantibodies/blood , Demyelinating Diseases/pathology , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Myelin-Associated Glycoprotein/immunology , Animals , Autoantigens/immunology , Biomarkers/blood , Demyelinating Diseases/immunology , Encephalomyelitis, Autoimmune, Experimental/blood , Enzyme-Linked Immunosorbent Assay , Inflammation/immunology , Inflammation/pathology , Myelin Proteins , Myelin-Oligodendrocyte Glycoprotein , Rats
15.
Neuropathology ; 31(4): 392-400, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21175863

ABSTRACT

Although intravenous immunoglobulin (IVIG) has been reported to improve the status of expanded disability status scale (EDSS) of multiple sclerosis (MS) patients and reduce the annual relapse rate, some studies did not find its beneficial effects. In the present study, using an animal model for MS, we found that prophylactic, but not therapeutic, treatment successfully suppressed the disease development. During the search for factors involved in the disease suppression by IVIG, we obtained evidence suggesting that IVIG exerts its function, at least in part, by suppressing activation of matrix metalloproteinases (MMP)-2 and -9. Gelatin zymography revealed that gelatinase activities were suppressed by IVIG treatment in the spinal cord, but not in plasma. This finding raises the possibility that IVIG blocks MMP activities at the interface between the blood stream and CNS. With in situ zymography, we also observed that gelatinase activities were expressed mainly in astrocytes in the inflamed spinal cord of control rats and that this expression was attenuated by the treatment. These findings provide useful information to set optimal conditions for IVIG treatment of MS and to obtain more beneficial effects.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/prevention & control , Immunoglobulins, Intravenous/therapeutic use , Matrix Metalloproteinases/metabolism , Multiple Sclerosis/prevention & control , Spinal Cord/metabolism , Animals , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Encephalomyelitis, Autoimmune, Experimental/metabolism , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/prevention & control , Multiple Sclerosis/drug therapy , Multiple Sclerosis/metabolism , Rats , Treatment Outcome
16.
EMBO Mol Med ; 2(12): 504-15, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21064192

ABSTRACT

Apoptosis signal-regulating kinase 1 (ASK1) is an evolutionarily conserved mitogen-activated protein kinase (MAPK) kinase kinase which plays important roles in stress and immune responses. Here, we show that ASK1 deficiency attenuates neuroinflammation in experimental autoimmune encephalomyelitis (EAE), without affecting the proliferation capability of T cells. Moreover, we found that EAE upregulates expression of Toll-like receptors (TLRs) in activated astrocytes and microglia, and that TLRs can synergize with ASK1-p38 MAPK signalling in the release of key chemokines from astrocytes. Consequently, oral treatment with a specific small molecular weight inhibitor of ASK1 suppressed EAE-induced autoimmune inflammation in both spinal cords and optic nerves. These results suggest that the TLR-ASK1-p38 pathway in glial cells may serve as a valid therapeutic target for autoimmune demyelinating disorders including multiple sclerosis.


Subject(s)
Demyelinating Diseases/metabolism , MAP Kinase Kinase Kinase 5/metabolism , Signal Transduction , Toll-Like Receptors/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Brain Diseases/drug therapy , Brain Diseases/genetics , Brain Diseases/immunology , Brain Diseases/metabolism , Chemokines/immunology , Demyelinating Diseases/drug therapy , Demyelinating Diseases/genetics , Demyelinating Diseases/immunology , Disease Models, Animal , Encephalitis , Enzyme Inhibitors/administration & dosage , Female , Gene Expression Regulation , Hashimoto Disease/drug therapy , Hashimoto Disease/genetics , Hashimoto Disease/immunology , Hashimoto Disease/metabolism , Humans , MAP Kinase Kinase Kinase 5/antagonists & inhibitors , MAP Kinase Kinase Kinase 5/genetics , Male , Mice , Mice, Inbred C57BL , Neuroglia/metabolism , Rats , Rats, Sprague-Dawley , Toll-Like Receptors/genetics , p38 Mitogen-Activated Protein Kinases/genetics
17.
J Alzheimers Dis ; 22(4): 1351-61, 2010.
Article in English | MEDLINE | ID: mdl-20930304

ABSTRACT

We recently demonstrated that newly developed non-viral amyloid-ß (Aß) DNA vaccines are safe and effective in reducing Aß burdens in the brains of Alzheimer's disease (AD) model mice. The present study was undertaken to examine whether DNA vaccines effectively and safely reduce Aß deposition in the brain of rhesus monkeys. For this purpose, DNA vaccines or empty vector at a dose of 3 mg were injected intramuscularly on a biweekly basis into rhesus monkeys (15-18 years old). Before and during vaccination, blood was drawn once a month and used for hematological and biochemical examinations. Six months after the first vaccination, it was demonstrated that anti-Aß antibodies in plasma of vaccinated monkeys were significantly elevated than that of control monkeys. Immunohistochemical examinations revealed that DNA vaccination reduced the Aß burden to approximately 50% of that found in control monkeys (p=0.026). There was neither inflammation nor microhemorrhage in the brain and no significant changes in cytokine and chemokine levels in the blood throughout the observation period. Taken together, DNA vaccination to monkeys is safe and effective in Aß reduction and provides useful information for performing preclinical and clinical trials.


Subject(s)
Alzheimer Disease/prevention & control , Alzheimer Vaccines/adverse effects , Amyloid beta-Peptides/metabolism , Brain/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Alzheimer Vaccines/immunology , Amyloid beta-Peptides/immunology , Animals , Blotting, Western , Brain/drug effects , Brain/immunology , DNA, Viral/immunology , Enzyme-Linked Immunosorbent Assay , Female , Immunohistochemistry , Macaca mulatta , Male , Statistics, Nonparametric
18.
Arthritis Rheum ; 62(10): 3088-92, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20583106

ABSTRACT

OBJECTIVE: To substantiate a pathogenic role of cytotoxic CD8 T cells in the development of a murine polymyositis model, C protein-induced myositis (CIM). METHODS: Beta(2)-microglobulin-null mutant, perforin-null mutant, and wild-type (WT) C57BL/6 mice were immunized with skeletal muscle C protein fragments to provoke CIM. Regional lymph node CD8 or CD4 T cells stimulated with C protein-pulsed dendritic cells were transferred adoptively to naive mice. Inflammation and damage of the muscle tissues were evaluated histologically. RESULTS: The incidence of myositis development was significantly lower in ß2-microglobulin-null and perforin-null mutant mice compared with WT mice. Inflammation was less severe in mutant mice, and the incidence of muscle injury was reduced significantly. Adoptive transfer of lymph node T cells from mice with CIM induced myositis in naive recipient mice. The CD8 T cell-induced muscle injuries were significantly more severe than the CD4 T cell-induced muscle injuries. CONCLUSION: Perforin-mediated cytotoxicity by CD8 T cells is definitively responsible for muscle injury in CIM.


Subject(s)
Polymyositis/immunology , Pore Forming Cytotoxic Proteins/immunology , T-Lymphocytes, Cytotoxic/immunology , beta 2-Microglobulin/immunology , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
J Immunol ; 183(7): 4773-81, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19734212

ABSTRACT

Repeated or continuous inflammation of the heart is one of the initiation factors for dilated cardiomyopathy (DCM). In previous studies, we established a DCM animal model by immunizing rats with cardiac C protein. In the present study, we analyze the role of matrix metalloproteinases (MMPs) in experimental autoimmune carditis (EAC) and subsequent DCM to elucidate the pathomechanisms of this disease. In this model, inflammation begins approximately 9 days after immunization. At that time, MMP activities were detected by in situ zymography. Real-time PCR analysis revealed continuous up-regulation of MMP-2 mRNA from 2 wk and thereafter. MMP-9 mRNA, however, had only a transient increase at 2 wk. Double staining with in situ zymography and cell markers demonstrated that gelatinase (MMP-2 and MMP-9)-expressing cells are infiltrating macrophages during the early stage and cardiomyocytes at later stages. Minocycline, which inhibits MMP-9 activities more strongly than MMP-2, significantly suppressed EAC, but an MMP-2-specific inhibitor, TISAM, did not affect the course of the disease. Furthermore, immunohistochemical examination revealed that minocycline treatment suppressed T cell and macrophage infiltration strongly, whereas TISAM did not. These findings indicate that MMP-9, but not MMP-2, is involved in the pathogenesis of the acute phase of EAC, and further suggest that MMP-9 inhibitors, minocycline and its derivatives, may be useful therapies for EAC and DCM.


Subject(s)
Cardiomyopathy, Dilated/enzymology , Carrier Proteins/toxicity , Matrix Metalloproteinase 2 , Matrix Metalloproteinase 9/physiology , Myocarditis/enzymology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/enzymology , Autoimmune Diseases/pathology , Cardiomyopathy, Dilated/drug therapy , Cardiomyopathy, Dilated/immunology , Cardiomyopathy, Dilated/pathology , Disease Progression , Female , Inflammation Mediators/physiology , Male , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 2/physiology , Matrix Metalloproteinase 9/biosynthesis , Matrix Metalloproteinase Inhibitors , Myocarditis/drug therapy , Myocarditis/immunology , Myocarditis/pathology , Protease Inhibitors/therapeutic use , Rats , Rats, Inbred Lew
20.
Neurosci Lett ; 465(2): 160-4, 2009 Nov 13.
Article in English | MEDLINE | ID: mdl-19766171

ABSTRACT

Glaucoma, one of the leading causes of irreversible blindness, is characterized by progressive degeneration of retinal ganglion cells (RGCs) and optic nerves. Although glaucoma is often associated with elevated intraocular pressure, recent studies have shown a relatively high prevalence of normal tension glaucoma (NTG) in glaucoma patient populations. In the mammalian retina, glutamate/aspartate transporter (GLAST) is localized to Müller glial cells, whereas excitatory amino acid carrier 1 (EAAC1) is expressed in neural cells, including RGCs. Since the loss of GLAST or EAAC1 leads to retinal degeneration similar to that seen in NTG, we examined the effects of interleukin-1 (IL-1) on RGC death in GLAST- and EAAC1-deficient mice. IL-1 promoted increased glutamate uptake in Müller cells by suppressing intracellular Na(+) accumulation, which is necessary to counteract Na(+)-glutamate cotransport. The observed trends for the glutamate uptake increase in the wild-type (WT), GLAST- and EAAC1-deficient mice were similar; however, the baseline glutamate uptake and intracellular Na(+) concentration in the GLAST-deficient mice were significantly lower than those in the wild-type mice. Consistently, pretreatment with IL-1 exhibited no beneficial effects on glutamate-induced RGC degeneration in the GLAST-deficient mice. In contrast, IL-1 significantly increased glutamate uptake by Müller cells and the number of surviving RGCs in the wild-type and EAAC1-deficient mice. Our findings suggest that the use of IL-1 for enhancing the function of glutamate transporters may be useful for neuroprotection in retinal degenerative disorders including NTG.


Subject(s)
Excitatory Amino Acid Transporter 1/deficiency , Excitatory Amino Acid Transporter 3/deficiency , Interleukin-1/metabolism , Retina/physiopathology , Retinal Degeneration/physiopathology , Retinal Ganglion Cells/physiology , Animals , Cell Death/physiology , Cell Survival/physiology , Excitatory Amino Acid Transporter 1/genetics , Excitatory Amino Acid Transporter 1/metabolism , Excitatory Amino Acid Transporter 3/genetics , Excitatory Amino Acid Transporter 3/metabolism , Glaucoma/physiopathology , Glutamic Acid/metabolism , Intracellular Space/metabolism , Intraocular Pressure , Mice , Mice, Knockout , Neuroglia/physiology , Sodium/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL