Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
J Hum Genet ; 69(5): 177-183, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38351237

RESUMO

Truncus Arteriosus (TA) is a congenital heart disease characterized by a single common blood vessel emerging from the right and left ventricles instead of the main pulmonary artery and aorta. TA accounts for 4% of all critical congenital heart diseases. The most common cause of TA is 22q11.2 deletion syndrome, accounting for 12-35% of all TA cases. However, no major causes of TA other than 22q11.2 deletion have been reported. We performed whole-genome sequencing of 11 Japanese patients having TA without 22q11.2 deletion. Among five patients, we identified pathogenic variants in TMEM260; the biallelic loss-of-function variants of which have recently been associated with structural heart defects and renal anomalies syndrome (SHDRA). In one patient, we identified a de novo pathogenic variant in GATA6, and in another patient, we identified a de novo probably pathogenic variant in NOTCH1. Notably, we identified a prevalent variant in TMEM260 (ENST00000261556.6), c.1617del (p.Trp539Cysfs*9), in 8/22 alleles among the 11 patients. The c.1617del variant was estimated to occur approximately 23 kiloyears ago. Based on the allele frequency of the c.1617del variant in the Japanese population (0.36%), approximately 26% of Japanese patients afflicted with TA could harbor homozygous c.1617del variants. This study highlights TMEM260, especially c.1617del, as a major genetic cause of TA in the Japanese population.


Assuntos
Síndrome de DiGeorge , Proteínas de Membrana , Feminino , Humanos , Masculino , Alelos , Síndrome de DiGeorge/genética , População do Leste Asiático/genética , Japão/epidemiologia , Proteínas de Membrana/genética , Receptor Notch1/genética , Tronco Arterial/patologia , Sequenciamento Completo do Genoma
3.
Brain Dev ; 45(8): 456-461, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37246116

RESUMO

BACKGROUND: A few case reports have described patients with myelin oligodendrocyte glycoprotein antibody (MOG-Ab)-associated demyelinating syndrome who presented with symptoms of aseptic meningitis. All such patients required immunotherapy. We report a patient with MOG-Ab-associated disorder (MOGAD) who presented with symptoms of aseptic meningitis and improved without treatment. CASE: A 13-year-old girl presented with fever, headache, decreased appetite, and neck stiffness. Cerebrospinal fluid (CSF) analysis revealed pleocytosis and magnetic resonance imaging (MRI) showed leptomeningeal enhancement. The patient was diagnosed with aseptic meningitis at admission. However, there were no signs of recovery 4 days after admission (i.e., 8 days after disease onset). Therefore, we performed extensive investigations to identify the cause of the underlying infection and inflammation. On day 14 after admission, the serum MOG-Ab test performed at admission came back positive (1:128) and she was diagnosed with MOGAD. She was discharged on day 18 after admission, because her symptoms, CSF pleocytosis, and MRI findings had improved. About 6 weeks after discharge, MRI revealed hyperintensity without gadolinium enhancement. However, her serum MOG-Ab test was negative. We did follow-ups for 11 months but found no new neurological symptoms. DISCUSSION AND CONCLUSION: To the best of our knowledge, this is the first ever report of a pediatric patient with MOGAD experiencing spontaneous remission with no demyelinating symptoms during an extended follow-up period.


Assuntos
Meningite Asséptica , Feminino , Humanos , Autoanticorpos , Meios de Contraste , Gadolínio , Leucocitose , Glicoproteína Mielina-Oligodendrócito , Remissão Espontânea , Adolescente
4.
Front Immunol ; 9: 2870, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30619258

RESUMO

Theiler's murine encephalomyelitis virus (TMEV) induces different diseases in the central nervous system (CNS) and heart, depending on the mouse strains and time course, with cytokines playing key roles for viral clearance and immune-mediated pathology (immunopathology). In SJL/J mice, TMEV infection causes chronic TMEV-induced demyelinating disease (TMEV-IDD) in the spinal cord about 1 month post-inoculation (p.i.). Unlike other immunopathology models, both pro- and anti-inflammatory cytokines can play dual roles in TMEV-IDD. Pro-inflammatory cytokines play beneficial roles in viral clearance while they are also detrimental in immune-mediated demyelination. Anti-inflammatory cytokines suppress not only protective anti-viral immune responses but also detrimental autoreactive immune responses. Conversely, in C3H mice, TMEV infection induces a non-CNS disease, myocarditis, with three distinctive phases: phase I, viral pathology with interferon and chemokine responses; phase II, immunopathology mediated by acquired immune responses; and phase III, cardiac fibrosis. Although the exact mechanism(s) by which a single virus, TMEV, induces these different diseases in different organs is unclear, our bioinformatics approaches, especially principal component analysis (PCA) of transcriptome data, allow us to identify the key factors contributing to organ-specific immunopathology. The PCA demonstrated that in vitro infection of a cardiomyocyte cell line reproduced the transcriptome profile of phase I in TMEV-induced myocarditis; distinct interferon/chemokine-related responses were induced in vitro in TMEV-infected cardiomyocytes, but not in infected neuronal cells. In addition, the PCA of the in vivo CNS transcriptome data showed that decreased lymphatic marker expressions were weakly associated with inflammation in TMEV infection. Here, dysfunction of lymphatic vessels is shown to potentially contribute to immunopathology by delaying the clearance of cytokines and immune cells from the inflammatory site, although this can also confine the virus at these sites, preventing virus spread via lymphatic vessels. On the other hand, in the heart, dysfunction of lymphatics was associated with reduced lymphatic muscle contractility provoked by pro-inflammatory cytokines. Therefore, TMEV infection may induce different patterns of cytokine expressions as well as lymphatic vessel dysfunction by rather different mechanisms between the CNS and heart, which might explain observed patterns of organ-specific immunopathology.


Assuntos
Infecções por Cardiovirus/imunologia , Citocinas/imunologia , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/imunologia , Miocardite/imunologia , Theilovirus/imunologia , Animais , Infecções por Cardiovirus/virologia , Linhagem Celular , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/metabolismo , Citocinas/genética , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/virologia , Modelos Animais de Doenças , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Vasos Linfáticos/imunologia , Vasos Linfáticos/metabolismo , Camundongos , Camundongos Endogâmicos/imunologia , Miocardite/virologia , Miocárdio/imunologia , Miocárdio/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Análise de Componente Principal
5.
Sci Rep ; 7(1): 10496, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28874814

RESUMO

Intracerebral Theiler's murine encephalomyelitis virus (TMEV) infection in mice induces inflammatory demyelination in the central nervous system. Although C57BL/6 mice normally resistant to TMEV infection with viral clearance, we have previously demonstrated that RORγt-transgenic (tg) C57BL/6 mice, which have Th17-biased responses due to RORγt overexpression in T cells, became susceptible to TMEV infection with viral persistence. Here, using T-bet-tg C57BL/6 mice and Gata3-tg C57BL/6 mice, we demonstrated that overexpression of T-bet, but not Gata3, in T cells was detrimental in TMEV infection. Unexpectedly, T-bet-tg mice died 2 to 3 weeks after infection due to failure of viral clearance. Here, TMEV infection induced splenic T cell depletion, which was associated with lower anti-viral antibody and T cell responses. In contrast, Gata3-tg mice remained resistant, while Gata3-tg mice had lower IFN-γ and higher IL-4 production with increased anti-viral IgG1 responses. Thus, our data identify how overexpression of T-bet and Gata3 in T cells alters anti-viral immunity and confers susceptibility to TMEV infection.


Assuntos
Fator de Transcrição GATA3/genética , Expressão Gênica , Poliomielite/genética , Poliomielite/virologia , Proteínas com Domínio T/genética , Theilovirus/fisiologia , Animais , Biomarcadores , Citocinas/metabolismo , Doenças Desmielinizantes/genética , Suscetibilidade a Doenças , Fator de Transcrição GATA3/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Imuno-Histoquímica , Camundongos , Camundongos Transgênicos , Poliomielite/imunologia , Poliomielite/metabolismo , Baço/imunologia , Baço/metabolismo , Proteínas com Domínio T/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Virulência , Replicação Viral
6.
J Neuroimmunol ; 278: 174-84, 2015 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-25434008

RESUMO

Theiler's murine encephalomyelitis virus (TMEV) can induce demyelination or myocarditis in susceptible mouse strains. A deficiency of NKT cells exacerbated TMEV-induced demyelinating disease (TMEV-IDD) in SJL/J and BALB/c mice. In C57BL/6 background, however, NKT-cell-deficient Jα18 KO mice remained as resistant to TMEV-IDD as wild-type mice. Echocardiography and histology showed that Jα18 KO mice developed more severe myocarditis (greater T cell infiltration and fibrosis) than wild-type mice, suggesting a protective role of NKT cells in myocarditis in C57BL/6 mice. Jα18 KO mice had higher cardiac viral RNA and anti-viral antibody titers, but had lower lymphoproliferation and IL-4 and IL-10 production.


Assuntos
Infecções por Cardiovirus/complicações , Doenças Desmielinizantes/etiologia , Miocardite/etiologia , Células T Matadoras Naturais/fisiologia , Fatores Etários , Animais , Anticorpos/sangue , Anticorpos/farmacologia , Infecções por Cardiovirus/patologia , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Sistema Nervoso Central/patologia , Sistema Nervoso Central/virologia , Citocinas/metabolismo , Doenças Desmielinizantes/virologia , Fibrose/patologia , Linfócitos/efeitos dos fármacos , Linfócitos/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocardite/patologia , Miocardite/virologia , Receptores de Antígenos de Linfócitos T alfa-beta/deficiência , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Especificidade da Espécie , Theilovirus/genética , Theilovirus/imunologia , Theilovirus/patogenicidade , Fatores de Tempo
7.
Brain Behav Immun ; 43: 86-97, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25046854

RESUMO

In a viral model for multiple sclerosis (MS), Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD), both immune-mediated tissue damage (immunopathology) and virus persistence have been shown to cause pathology. T helper (Th) 17 cells are a Th cell subset, whose differentiation requires the transcription factor retinoic acid-related orphan receptor (ROR) γt, secrete pro-inflammatory cytokines, including IL-17, and can antagonize Th1 cells. Although Th17 cells have been shown to play a pathogenic role in immune-mediated diseases or a protective role in bacterial and fungal infections, their role in viral infections is unclear. Using newly established Th17-biased RORγt Tg mice, we tested whether Th17 cells could play a pathogenic or protective role in TMEV-IDD by contributing to immunopathology and/or by modulating anti-viral Th1 immune responses. While TMEV-infected wild-type littermate C57BL/6 mice are resistant to TMEV-IDD, RORγt Tg mice developed inflammatory demyelinating lesions with virus persistence in the spinal cord. TMEV-infected RORγt Tg mice had higher levels of IL-17, lower levels of interferon-γ, and fewer CD8(+) T cells, without alteration in overall levels of anti-viral lymphoproliferative and antibody responses, compared with TMEV-infected wild-type mice. This suggests that a Th17-biased "gain-of-function" mutation could increase susceptibility to virus-mediated demyelinating diseases.


Assuntos
Modelos Animais de Doenças , Esclerose Múltipla/imunologia , Medula Espinal/imunologia , Células Th17/imunologia , Theilovirus/imunologia , Animais , Suscetibilidade a Doenças , Camundongos , Camundongos Transgênicos , Esclerose Múltipla/patologia , Esclerose Múltipla/virologia , Medula Espinal/patologia
8.
Cell Immunol ; 292(1-2): 85-93, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25460083

RESUMO

We established a novel model of myocarditis induced with Theiler's murine encephalomyelitis virus (TMEV), which has been used as a viral model for multiple sclerosis and seizure/epilepsy. Following TMEV infection, C3H mice developed severe myocarditis with T cell infiltration, while C57BL/6 mice had mild lesions and SJL/J mice had no inflammation in the heart. In C3H mice, myocarditis was divided into three phases: acute viral, subacute immune, and chronic fibrotic phases. Using toll-like receptor (TLR) 4-deficient C3H mice, we found that interleukin (IL)-6, IL-17, TLR4, and anti-viral immune responses were associated with myocarditis susceptibility.


Assuntos
Miocardite/imunologia , Linfócitos T/imunologia , Theilovirus/fisiologia , Replicação Viral , Animais , Fibrose/imunologia , Cinética , Camundongos Endogâmicos , Miocardite/patologia , Miocardite/virologia
9.
J Neuroimmunol ; 276(1-2): 142-9, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25288300

RESUMO

Th17 cells play an important role in multiple sclerosis (MS) and its autoimmune model, experimental autoimmune encephalomyelitis (EAE). However, studies have not addressed how enhanced Th17 immune responses can affect demyelinating diseases. We induced EAE with MOG in RORγt transgenic C57BL/6 mice that overexpress a Th17 inducing transcription factor. RORγt transgenic mice developed more severe EAE than wild-type mice with more robust anti-MOG Th17 immune responses. In contrast, mice overexpressing T-bet, a Th1-inducing transcription factor, were resistant to EAE. Therefore, a genetic bias toward Th17 immune responses could contribute to CNS immunopathology.


Assuntos
Sistema Nervoso Central/patologia , Esclerose Múltipla/genética , Esclerose Múltipla/patologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sistema Nervoso Central/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/genética , Citometria de Fluxo , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Esclerose Múltipla/induzido quimicamente , Proteínas com Domínio T/genética , Células Th1/patologia
10.
Circ Cardiovasc Genet ; 7(4): 444-54, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25031303

RESUMO

BACKGROUND: Myocarditis is an inflammatory disease of the cardiac muscle and is mainly caused by viral infections. Viral myocarditis has been proposed to be divided into 3 phases: the acute viral phase, the subacute immune phase, and the chronic cardiac remodeling phase. Although individualized therapy should be applied depending on the phase, no clinical or experimental studies have found biomarkers that distinguish between the 3 phases. Theiler's murine encephalomyelitis virus belongs to the genus Cardiovirus and can cause myocarditis in susceptible mouse strains. METHODS AND RESULTS: Using this novel model for viral myocarditis induced with Theiler's murine encephalomyelitis virus, we conducted multivariate analysis including echocardiography, serum troponin and viral RNA titration, and microarray to identify the biomarker candidates that can discriminate the 3 phases. Using C3H mice infected with Theiler's murine encephalomyelitis virus on 4, 7, and 60 days post infection, we conducted bioinformatics analyses, including principal component analysis and k-means clustering of microarray data, because our traditional cardiac and serum assays, including 2-way comparison of microarray data, did not lead to the identification of a single biomarker. Principal component analysis separated heart samples clearly between the groups of 4, 7, and 60 days post infection. Representative genes contributing to the separation were as follows: 4 and 7 days post infection, innate immunity-related genes, such as Irf7 and Cxcl9; 7 and 60 days post infection, acquired immunity-related genes, such as Cd3g and H2-Aa; and cardiac remodeling-related genes, such as Mmp12 and Gpnmb. CONCLUSIONS: Sets of molecules, not single molecules, identified by unsupervised principal component analysis, were found to be useful as phase-specific biomarkers.


Assuntos
Biomarcadores/sangue , Miocardite/patologia , Theilovirus/patogenicidade , Imunidade Adaptativa/genética , Animais , Análise por Conglomerados , Biologia Computacional , Modelos Animais de Doenças , Ecocardiografia , Imunidade Inata/genética , Camundongos , Camundongos Endogâmicos C3H , Análise Multivariada , Miocardite/imunologia , Miocardite/virologia , Análise de Sequência com Séries de Oligonucleotídeos , Análise de Componente Principal , RNA Viral/sangue , Reação em Cadeia da Polimerase em Tempo Real , Theilovirus/genética , Troponina/sangue
11.
Int J Mol Sci ; 15(2): 1700-18, 2014 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-24463292

RESUMO

T helper (Th)2 cells have been proposed to play a neuroprotective role in multiple sclerosis (MS). This is mainly based on "loss-of-function" studies in an animal model for MS, experimental autoimmune encephalomyelitis (EAE), using blocking antibodies against Th2 related cytokines, and knockout mice lacking Th2-related molecules. We tested whether an increase of Th2 responses ("gain-of-function" approach) could alter EAE, the approach of novel GATA binding protein 3 (GATA3)-transgenic (tg) mice that overexpress GATA3, a transcription factor required for Th2 differentiation. In EAE induced with myelin oligodendrocyte glycoprotein (MOG)35-55 peptide, GATA3-tg mice had a significantly delayed onset of disease and a less severe maximum clinical score, compared with wild-type C57BL/6 mice. Histologically, GATA3-tg mice had decreased levels of meningitis and demyelination in the spinal cord, and anti-inflammatory cytokine profiles immunologically, however both groups developed similar levels of MOG-specific lymphoproliferative responses. During the early stage, we detected higher levels of interleukin (IL)-4 and IL-10, with MOG and mitogen stimulation of regional lymph node cells in GATA3-tg mice. During the late stage, only mitogen stimulation induced higher IL-4 and lower interferon-γ and IL-17 production in GATA3-tg mice. These results suggest that a preexisting bias toward a Th2 immune response may reduce the severity of inflammatory demyelinating diseases, including MS.


Assuntos
Fator de Transcrição GATA3/genética , Esclerose Múltipla/genética , Esclerose Múltipla/imunologia , Células Th2/imunologia , Células Th2/metabolismo , Animais , Complexo CD3/metabolismo , Citocinas/metabolismo , Doenças Desmielinizantes/genética , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/metabolismo , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Fator de Transcrição GATA3/metabolismo , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Transgênicos , Esclerose Múltipla/metabolismo , Mutação , Glicoproteína Mielina-Oligodendrócito/imunologia , Medula Espinal/imunologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
12.
Brain Pathol ; 24(5): 436-51, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24417588

RESUMO

Multiple sclerosis (MS) has been proposed to be an immune-mediated disease in the central nervous system (CNS) that can be triggered by virus infections. In Theiler's murine encephalomyelitis virus (TMEV) infection, during the first week (acute stage), mice develop polioencephalomyelitis. After 3 weeks (chronic stage), mice develop immune-mediated demyelination with virus persistence, which has been used as a viral model for MS. Regulatory T cells (Tregs) can suppress inflammation, and have been suggested to be protective in immune-mediated diseases, including MS. However, in virus-induced inflammatory demyelination, although Tregs can suppress inflammation, preventing immune-mediated pathology, Tregs may also suppress antiviral immune responses, leading to more active viral replication and/or persistence. To determine the role and potential translational usage of Tregs in MS, we treated TMEV-infected mice with ex vivo generated induced Tregs (iTregs) on day 0 (early) or during the chronic stage (therapeutic). Early treatment worsened clinical signs during acute disease. The exacerbation of acute disease was associated with increased virus titers and decreased immune cell recruitment in the CNS. Therapeutic iTreg treatment reduced inflammatory demyelination during chronic disease. Immunologically, iTreg treatment increased interleukin-10 production from B cells, CD4(+) T cells and dendritic cells, which may contribute to the decreased CNS inflammation.


Assuntos
Esclerose Múltipla/imunologia , Linfócitos T Reguladores/imunologia , Animais , Encéfalo/imunologia , Encéfalo/patologia , Infecções por Cardiovirus/imunologia , Encefalite/imunologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Esclerose Múltipla/virologia , Poliomielite/imunologia , Medula Espinal/imunologia , Medula Espinal/patologia , Theilovirus/imunologia
13.
Future Virol ; 7(6): 593-608, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23024699

RESUMO

In immune-mediated diseases, Treg and proinflammatory Th17 cells have been suggested to play either suppressor (beneficial) or effector (detrimental) roles, respectively. Tissue damage in viral infections can be caused by direct viral replication or immunopathology. Viral replication can be enhanced by anti-inflammatory responses and suppressed by proinflammatory responses. However, Tregs could suppress proinflammatory responses, reducing immunopathology, while Th17 cell-induced inflammation may enhance immunopathology. Here, the roles of Treg and Th17 cells depend on whether tissue damage is caused by direct virus replication or immunopathology, which differ depending on the virus, disease stage and host immune background. Although the precise mechanisms of tissue damage in multiple sclerosis and myocarditis are unclear, both viral replication and immune effector cells have been proposed to cause pathogenesis. Personalized medicine that alters the balance between Treg and Th17 cells may ameliorate viral pathology during infections.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...