Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 70
Filtrar
1.
Front Immunol ; 14: 1167972, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37153539

RESUMO

Theiler's murine encephalomyelitis virus (TMEV) establishes persistent viral infections in the central nervous system and induces chronic inflammatory demyelinating disease in susceptible mice. TMEV infects dendritic cells, macrophages, B cells, and glial cells. The state of TLR activation in the host plays a critical role in initial viral replication and persistence. The further activation of TLRs enhances viral replication and persistence, leading to the pathogenicity of TMEV-induced demyelinating disease. Various cytokines are produced via TLRs, and MDA-5 signals linked with NF-κB activation following TMEV infection. In turn, these signals further amplify TMEV replication and the persistence of virus-infected cells. The signals further elevate cytokine production, promoting the development of Th17 responses and preventing cellular apoptosis, which enables viral persistence. Excessive levels of cytokines, particularly IL-6 and IL-1ß, facilitate the generation of pathogenic Th17 immune responses to viral antigens and autoantigens, leading to TMEV-induced demyelinating disease. These cytokines, together with TLR2 may prematurely generate functionally deficient CD25-FoxP3+ CD4+ T cells, which are subsequently converted to Th17 cells. Furthermore, IL-6 and IL-17 synergistically inhibit the apoptosis of virus-infected cells and the cytolytic function of CD8+ T lymphocytes, prolonging the survival of virus-infected cells. The inhibition of apoptosis leads to the persistent activation of NF-κB and TLRs, which continuously provides an environment of excessive cytokines and consequently promotes autoimmune responses. Persistent or repeated infections of other viruses such as COVID-19 may result in similar continuous TLR activation and cytokine production, leading to autoimmune diseases.


Assuntos
COVID-19 , Doenças Desmielinizantes , Theilovirus , Camundongos , Animais , Interleucina-6 , Doenças Desmielinizantes/patologia , NF-kappa B , Virulência , Citocinas , Replicação Viral
2.
Int J Mol Sci ; 22(10)2021 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-34067536

RESUMO

Several virus-induced models were used to study the underlying mechanisms of multiple sclerosis (MS). The infection of susceptible mice with Theiler's murine encephalomyelitis virus (TMEV) establishes persistent viral infections and induces chronic inflammatory demyelinating disease. In this review, the innate and adaptive immune responses to TMEV are discussed to better understand the pathogenic mechanisms of viral infections. Professional (dendritic cells (DCs), macrophages, and B cells) and non-professional (microglia, astrocytes, and oligodendrocytes) antigen-presenting cells (APCs) are the major cell populations permissive to viral infection and involved in cytokine production. The levels of viral loads and cytokine production in the APCs correspond to the degrees of susceptibility of the mice to the TMEV-induced demyelinating diseases. TMEV infection leads to the activation of cytokine production via TLRs and MDA-5 coupled with NF-κB activation, which is required for TMEV replication. These activation signals further amplify the cytokine production and viral loads, promote the differentiation of pathogenic Th17 responses, and prevent cellular apoptosis, enabling viral persistence. Among the many chemokines and cytokines induced after viral infection, IFN α/ß plays an essential role in the downstream expression of costimulatory molecules in APCs. The excessive levels of cytokine production after viral infection facilitate the pathogenesis of TMEV-induced demyelinating disease. In particular, IL-6 and IL-1ß play critical roles in the development of pathogenic Th17 responses to viral antigens and autoantigens. These cytokines, together with TLR2, may preferentially generate deficient FoxP3+CD25- regulatory cells converting to Th17. These cytokines also inhibit the apoptosis of TMEV-infected cells and cytolytic function of CD8+ T lymphocytes (CTLs) and prolong the survival of B cells reactive to viral and self-antigens, which preferentially stimulate Th17 responses.


Assuntos
Doenças Desmielinizantes/imunologia , Esclerose Múltipla/imunologia , Theilovirus/fisiologia , Imunidade Adaptativa/imunologia , Animais , Células Apresentadoras de Antígenos/metabolismo , Astrócitos/metabolismo , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/metabolismo , Infecções por Cardiovirus/virologia , Citocinas , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Humanos , Imunidade Inata/imunologia , Camundongos , Microglia/metabolismo , Esclerose Múltipla/metabolismo , Oligodendroglia/metabolismo , Transdução de Sinais/imunologia , Theilovirus/patogenicidade
3.
J Neuroinflammation ; 17(1): 307, 2020 Oct 17.
Artigo em Inglês | MEDLINE | ID: mdl-33069239

RESUMO

BACKGROUND: Experimental autoimmune encephalitis (EAE) and virally induced demyelinating disease are two major experimental model systems used to study human multiple sclerosis. Although endothelin-1 level elevation was previously observed in the CNS of mice with EAE and viral demyelinating disease, the potential role of endothelin-1 in the development of these demyelinating diseases is unknown. METHODS AND RESULTS: In this study, the involvement of endothelin-1 in the development and progression of demyelinating diseases was investigated using these two experimental models. Administration of endothelin-1 significantly promoted the progression of both experimental diseases accompanied with elevated inflammatory T cell responses. In contrast, administration of specific endothelin-1 inhibitors (BQ610 and BQ788) significantly inhibited progression of these diseases accompanied with reduced T cell responses to the respective antigens. CONCLUSIONS: These results strongly suggest that the level of endothelin-1 plays an important role in the pathogenesis of immune-mediated CNS demyelinating diseases by promoting immune responses.


Assuntos
Infecções por Cardiovirus/metabolismo , Doenças Desmielinizantes/metabolismo , Endotelina-1/biossíntese , Theilovirus , Animais , Infecções por Cardiovirus/induzido quimicamente , Infecções por Cardiovirus/imunologia , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/imunologia , Endotelina-1/antagonistas & inibidores , Endotelina-1/toxicidade , Feminino , Camundongos , Oligopeptídeos/farmacologia , Linfócitos T/imunologia , Linfócitos T/metabolismo
4.
Int J Mol Sci ; 21(20)2020 Oct 19.
Artigo em Inglês | MEDLINE | ID: mdl-33086489

RESUMO

The infection of susceptible mice with Theiler's murine encephalomyelitis virus (TMEV) induces a T cell-mediated demyelinating disease. This system has been studied as a relevant infection model for multiple sclerosis (MS). Therefore, defining the type of T cell responses and their functions is critically important for understanding the relevant pathogenic mechanisms. In this study, we adoptively transferred naive VP2-specific TCR-Tg CD4+ T cells into syngeneic susceptible SJL mice and monitored the development of the disease and the activation and proliferation of CD4+ T cells during the early stages of viral infection. The preexisting VP2-specific naive CD4+ T cells promoted the pathogenesis of the disease in a dose-dependent manner. The transferred VP2-specific CD4+ T cells proliferated rapidly in the CNS starting at 2-3 dpi. High levels of FoxP3+CD4+ T cells were found in the CNS early in viral infection (3 dpi) and persisted throughout the infection. Activated VP2-specific FoxP3+CD4+ T cells inhibited the production of IFN-γ, but not IL-17, via the same VP2-specific CD4+ T cells without interfering in proliferation. Thus, the early presence of regulatory T cells in the CNS with viral infection may favor the induction of pathogenic Th17 cells over protective Th1 cells in susceptible mice, thereby establishing the pathogenesis of virus-induced demyelinating disease.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Infecções por Cardiovirus/imunologia , Infecções por Cardiovirus/virologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/virologia , Theilovirus/fisiologia , Transferência Adotiva , Animais , Proliferação de Células , Sistema Nervoso Central/patologia , Citocinas/biossíntese , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/virologia , Fatores de Transcrição Forkhead/metabolismo , Interferon gama/metabolismo , Interleucina-17/biossíntese , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos T/metabolismo , Especificidade da Espécie
5.
Cells ; 9(8)2020 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-32727036

RESUMO

Theiler's murine encephalomyelitis virus (TMEV) induces immune-mediated inflammatory demyelinating disease in susceptible mice that is similar to human multiple sclerosis (MS). In light of anti-CD20 therapies for MS, the susceptibility of B cells to TMEV infection is particularly important. In our study, direct viral exposure to macrophages and lymphocytes resulted in viral replication and cellular stimulation in the order of DCs, macrophages, B cells, and T cells. Notably, B cells produced viral proteins and expressed elevated levels of CD69, an activation marker. Similarly, the expression of major histocompatibility complex class II and costimulatory molecules in B cells was upregulated. Moreover, TMEV-infected B cells showed elevated levels of antigen-presenting function and antibody production. TMEV infection appeared to polyclonally activate B cells to produce autoantibodies and further T cell stimulation. Thus, the viral infection might potentially affect the outcome of autoimmune diseases, and/or the development of other chronic infections, including the protection and/or pathogenesis of TMEV-induced demyelinating disease.


Assuntos
Autoanticorpos/metabolismo , Linfócitos B/imunologia , Esclerose Múltipla/imunologia , Theilovirus/patogenicidade , Animais , Feminino , Camundongos
6.
PLoS One ; 14(2): e0212017, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30811446

RESUMO

Adult heart size is determined predominantly by the cardiomyocyte number and size. The cardiomyocyte number is determined primarily in the embryonic and perinatal period, as adult cardiomyocyte proliferation is restricted in comparison to that seen during the perinatal period. Recent evidence has implicated the mammalian Hippo kinase pathway as being critical in cardiomyocyte proliferation. Though the transcription factor, Tead1, is the canonical downstream transcriptional factor of the hippo kinase pathway in cardiomyocytes, the specific role of Tead1 in cardiomyocyte proliferation in the perinatal period has not been determined. Here, we report the generation of a cardiomyocyte specific perinatal deletion of Tead1, using Myh6-Cre deletor mice (Tead1-cKO). Perinatal Tead1 deletion was lethal by postnatal day 9 in Tead1-cKO mice due to dilated cardiomyopathy. Tead1-deficient cardiomyocytes have significantly decreased proliferation during the immediate postnatal period, when proliferation rate is normally high. Deletion of Tead1 in HL-1 cardiac cell line confirmed that cell-autonomous Tead1 function is required for normal cardiomyocyte proliferation. This was secondary to significant decrease in levels of many proteins, in vivo, that normally promote cell cycle in cardiomyocytes. Taken together this demonstrates the non-redundant critical requirement for Tead1 in regulating cell cycle proteins and proliferation in cardiomyocytes in the perinatal heart.


Assuntos
Cardiomiopatia Dilatada/mortalidade , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Miócitos Cardíacos/citologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Animais Recém-Nascidos , Cardiomiopatia Dilatada/genética , Proliferação de Células , Células Cultivadas , Feminino , Deleção de Genes , Genes Letais , Camundongos , Miócitos Cardíacos/metabolismo , Tamanho do Órgão , Gravidez , Transdução de Sinais , Fatores de Transcrição de Domínio TEA
7.
Artigo em Inglês | MEDLINE | ID: mdl-29410948

RESUMO

CXCL-1, also called keratinocyte-derived cytokine (KC), is a predominant chemokine produced in glial cells upon infection with Theiler's murine encephalomyelitis virus (TMEV). In this study, we assessed the role of KC in the development of TMEV-induced demyelinating disease by utilizing polyclonal anti-KC antibodies as well as KC-expressing recombinant TMEV. Our results indicate that the level of KC produced after infection with TMEV or stimulation with various TLRs is significantly higher in various cells from susceptible SJL mice compared to those in cells from resistant B6 mice. SJL mice treated with rabbit anti-KC antibodies displayed accelerated development of TMEV-induced demyelinating disease, elevated viral loads in the CNS and decreased antiviral T cell responses. In addition, infection of susceptible SJL mice with recombinant KC-TMEV produced biologically active KC, which resulted in the accelerated pathogenesis of demyelinating disease and elevated T cell responses to viral antigens compared to mice infected with control recombinant HEL-TMEV. These results strongly suggest that both the lack of KC during TMEV infection and the excessive presence of the chemokine promote the pathogenesis of demyelinating disease. Therefore, a balance in the level of KC during TMEV infection appears to be critically important in controlling the pathogenesis of demyelinating disease.


Assuntos
Infecções por Cardiovirus/complicações , Infecções por Cardiovirus/virologia , Quimiocina CXCL1/metabolismo , Doenças Desmielinizantes/etiologia , Doenças Desmielinizantes/metabolismo , Theilovirus/fisiologia , Animais , Anticorpos Monoclonais/farmacologia , Astrócitos/imunologia , Astrócitos/metabolismo , Astrócitos/virologia , Quimiocina CXCL1/antagonistas & inibidores , Doenças Desmielinizantes/diagnóstico , Doenças Desmielinizantes/tratamento farmacológico , Modelos Animais de Doenças , Feminino , Imunoglobulina G/farmacologia , Interferon gama/biossíntese , Queratinócitos/metabolismo , Queratinócitos/virologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/virologia , Camundongos , Fator 88 de Diferenciação Mieloide/metabolismo , Neutrófilos/imunologia , Neutrófilos/metabolismo , Neutrófilos/virologia , Medula Espinal/imunologia , Medula Espinal/metabolismo , Medula Espinal/patologia , Medula Espinal/virologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
8.
JCI Insight ; 2(17)2017 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-28878117

RESUMO

Heart disease remains the leading cause of death worldwide, highlighting a pressing need to identify novel regulators of cardiomyocyte (CM) function that could be therapeutically targeted. The mammalian Hippo/Tead pathway is critical in embryonic cardiac development and perinatal CM proliferation. However, the requirement of Tead1, the transcriptional effector of this pathway, in the adult heart is unknown. Here, we show that tamoxifen-inducible adult CM-specific Tead1 ablation led to lethal acute-onset dilated cardiomyopathy, associated with impairment in excitation-contraction coupling. Mechanistically, we demonstrate Tead1 is a cell-autonomous, direct transcriptional activator of SERCA2a and SR-associated protein phosphatase 1 regulatory subunit, Inhibitor-1 (I-1). Thus, Tead1 deletion led to a decrease in SERCA2a and I-1 transcripts and protein, with a consequent increase in PP1-activity, resulting in accumulation of dephosphorylated phospholamban (Pln) and decreased SERCA2a activity. Global transcriptomal analysis in Tead1-deleted hearts revealed significant changes in mitochondrial and sarcomere-related pathways. Additional studies demonstrated there was a trend for correlation between protein levels of TEAD1 and I-1, and phosphorylation of PLN, in human nonfailing and failing hearts. Furthermore, TEAD1 activity was required to maintain PLN phosphorylation and expression of SERCA2a and I-1 in human induced pluripotent stem cell-derived (iPS-derived) CMs. To our knowledge, taken together, this demonstrates a nonredundant, novel role of Tead1 in maintaining normal adult heart function.


Assuntos
Cardiomiopatia Dilatada/metabolismo , Proteínas de Ligação a DNA/fisiologia , Miócitos Cardíacos/citologia , Fatores de Transcrição/fisiologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Cardiomiopatia Dilatada/induzido quimicamente , Cardiomiopatia Dilatada/patologia , Proliferação de Células , Proteínas de Ligação a DNA/genética , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Camundongos , Camundongos Knockout , Miocárdio/enzimologia , Miocárdio/metabolismo , Fosforilação , Proteína Fosfatase 1/metabolismo , Retículo Sarcoplasmático/enzimologia , ATPases Transportadoras de Cálcio do Retículo Sarcoplasmático/metabolismo , Fatores de Transcrição de Domínio TEA , Tamoxifeno/farmacologia , Fatores de Transcrição/genética
9.
J Mol Diagn ; 19(5): 651-658, 2017 09.
Artigo em Inglês | MEDLINE | ID: mdl-28743024

RESUMO

Customized gene-panel tests, based on next-generation sequencing, have demonstrated their usefulness in a plethora of clinical settings. As with other clinical diagnostic techniques, gene-panel sequencing for clinical purposes requires precise quality control (QC) measures to ensure its reliability. Only detected variants are currently recorded in clinical reports; however, identifying whether a nondetected variant is a true or false negative is regarded essential in a clinical setting and, thus, a comprehensive QC measure is in demand. Conventional QC metrics, such as mean coverage and uniformity, are considered inadequate for such an evaluation. As such, a more specific measure focused on clinically important variants is herein proposed. In this study, we suggest a new scoring method for assessing the quality of clinical gene-panel sequencing data, specifically for the detection of a set of single-nucleotide variants. The performance of the method was analyzed using 2295 clinical samples (1012 formalin-fixed, paraffin-embedded and 1283 fresh-frozen tissues), and was shown to provide additional information that conventional methods do not show, such as mean depth and uniformity. Customized sequencing protocols, which include QC criteria, have been optimized by each genomic laboratory. The pass rate scoring method proposed in this study provides an appropriate QC response variable for the customized panel, which strengthens the reliability of calls on clinically relevant variants implicated in clinical reports.


Assuntos
Testes Genéticos/métodos , Testes Genéticos/normas , Polimorfismo de Nucleotídeo Único , Análise de Sequência de DNA/normas , Algoritmos , Alelos , Frequência do Gene , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Controle de Qualidade , Reprodutibilidade dos Testes
10.
PLoS One ; 12(4): e0176406, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28445497

RESUMO

Infection of various cells with Theiler's murine encephalomyelitis virus (TMEV) activates the TLR- and melanoma differentiation-associated gene 5 (MDA5)-dependent pathways, resulting in the production of IL-1ß via the activation of caspase-1 upon assembly of the node-like receptor protein 3 (NLRP3) inflammasome. The role of IL-1ß in the pathogenesis of TMEV-induced demyelinating disease was previously investigated. However, the signaling effects of prostaglandin E2 (PGE2) downstream of the NLRP3 inflammasome on the immune responses to viral determinants and the pathogenesis of demyelinating disease are unknown. In this study, we investigated the levels of intermediate molecules leading to PGE2 signaling and the effects of blocking PGE2 signaling on the immune response to TMEV infection, viral persistence and the development of demyelinating disease. We demonstrate here that TMEV infection activates the NLRP3 inflammasome and PGE2 signaling much more vigorously in dendritic cells (DCs) and CD11b+ cells from susceptible SJL mice than in cells from resistant B6 mice. Inhibition of virus-induced PGE2 signaling using AH23848 resulted in decreased pathogenesis of demyelinating disease and viral loads in the central nervous system (CNS). In addition, AH23848 treatment caused the elevation of protective early IFN-γ-producing CD4+ and CD8+ T cell responses. Because the levels of IFN-ß were lower in AH23848-treated mice but the level of IL-6 was similar, over-production of pathogenic IFN-ß was modulated and the generation of IFN-γ-producing T cell responses was enhanced by the inhibition of PGE2 signaling. These results strongly suggest that excessive activation of the NLRP3 inflammasome and downstream PGE2 signaling contribute to the pathogenesis of TMEV-induced demyelinating disease.


Assuntos
Infecções por Cardiovirus/complicações , Doenças Desmielinizantes/fisiopatologia , Dinoprostona/metabolismo , Theilovirus/fisiologia , Animais , Compostos de Bifenilo/farmacologia , Células da Medula Óssea/citologia , Encéfalo/metabolismo , Encéfalo/patologia , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/citologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/metabolismo , Infecções por Cardiovirus/fisiopatologia , Células Cultivadas , Doenças Desmielinizantes/virologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Feminino , Expressão Gênica/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Neuroglia/citologia , Neuroglia/metabolismo , Receptores de Prostaglandina E Subtipo EP4/antagonistas & inibidores , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais/efeitos dos fármacos , Medula Espinal/metabolismo , Medula Espinal/patologia , Carga Viral
11.
Int Immunol ; 28(12): 575-584, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27803063

RESUMO

Natalizumab, which is an antibody against α4 integrin, has been used for the treatment of multiple sclerosis. In the present study, we investigated both the role of α4 integrin and the therapeutic effect of HCA3551, a newly synthesized orally active small molecule α4 integrin antagonist, in the development of Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD). The mRNA levels of α4 integrins were significantly up-regulated in the central nervous system (CNS) of mice with TMEV-IDD as compared with naive mice (*P < 0.05). HCA3551 treatment in the effector phase significantly suppressed both the clinical and histological development of TMEV-IDD. The number of infiltrating mononuclear inflammatory cells in the CNS was significantly decreased in the mice treated with HCA3551 (**P < 0.01). The labeling indices for CD68 antigen and the absolute cell numbers of TNF-α-producing CD4+ T cells and IFN-γ-producing CD8+ T cells were significantly decreased in the CNS of mice treated with HCA3551 (*P < 0.05). HCA3551 treatment in the effector phase might inhibit the binding of α4 integrin to vascular cell adhesion molecule-1, thereby decreasing the number of mononuclear cells in the CNS.


Assuntos
Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/virologia , Integrina alfa4/metabolismo , Esclerose Múltipla/metabolismo , Theilovirus/patogenicidade , Animais , Modelos Animais de Doenças , Feminino , Integrina alfa4/genética , Integrina alfa4/imunologia , Camundongos , Camundongos Endogâmicos , Esclerose Múltipla/virologia
12.
J Neuroinflammation ; 13(1): 133, 2016 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-27250711

RESUMO

BACKGROUND: Chronic infection with Theiler's murine encephalomyelitis virus (TMEV) in susceptible SJL/J mice induces an immune-mediated demyelinating disease and has extensively been used as a relevant infectious model for multiple sclerosis (MS). Infection of the host with many other viruses also leads to acute or chronic inflammatory diseases in the central nervous system (CNS). Levels of viral load in the host often play a critical role in the pathogenesis of virus-induced diseases. Thus, the inhibition of viral replication in the host against a broad spectrum of similar viruses is critically important for preventing the viral pathogenicity. METHODS: P2/P3-expressing transgenic (B6 X SJL)F1 founders were generated and bred onto the C57BL/6 and SJL/J backgrounds. Differences in the development of demyelinating disease were compared. Viral persistence, cytokine production, and immune responses in the CNS of infected control and P2/P3-Tg mice were analyzed after infection using quantitative PCR, ELISA, and flow cytometry. Various cell types from the control and P2/P3-Tg mice, as well as cells transfected in vitro with the P2 and/or P3 regions, were also analyzed for viral replication and innate cytokine production. RESULTS: P2/P3-transgenic (P2/P3-Tg) mice carrying the viral non-structural protein genes displayed significantly reduced virus-specific T cell responses in the CNS against both the structural and non-structural proteins. Consequently, viral loads in the CNS were greater in the Tg mice during the chronic infection. However, P2/P3-Tg SJL mice exhibited reduced disease incidence and less severe clinical symptoms than did their non-transgenic littermates. Interestingly, P2/P3-Tg mice showed low viral loads in the CNS at a very early period after infection (1-3 days) with TMEV and related EMCV but not unrelated VSV. Cells from P2/P3-Tg mice and cells transfected with the P2 and/or P3 regions in vitro yielded also lower viral replication but higher IFN-α/ß production. CONCLUSIONS: This study demonstrates that the expression of viral non-structural genes in mice inhibits initial viral replication and suppresses sustaining pathogenic anti-viral immune responses to broad viral determinants. It appears that the elevation of innate immune cytokines produced in the cells expressing the non-structural viral genes upon viral infection is responsible for the inhibitions. The inhibition is partially virus-specific as it is more efficient for a related virus compared to an unrelated virus, suggesting a role for the similarity in the viral genome structures. Therefore, the expression of viral non-structural genes may serve as a useful new method to prevent a broadly virus-specific pathogenesis in the hosts.


Assuntos
Doenças Desmielinizantes/genética , Doenças Desmielinizantes/metabolismo , Regulação Viral da Expressão Gênica , Theilovirus/genética , Theilovirus/metabolismo , Replicação Viral/fisiologia , Animais , Células Cultivadas , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Linfócitos T/fisiologia
13.
Int Immunol ; 27(7): 333-44, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25721871

RESUMO

Dimethyl fumarate (DMF) is a modifier of the nuclear factor (erythroid-derived 2)-2 (Nrf2)-kelch-like ECH-associated protein 1 (Keap1) pathway. DMF treatment in the effector phase significantly suppressed the development of Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD) both clinically and histologically. DMF treatment leads to an enhanced Nrf2 antioxidant response in TMEV-IDD mice. DMF treatment in the effector phase significantly suppressed the level of IL-17A mRNA. DMF is known to inhibit differentiation of T helper 17 (Th17) cells via suppressing NF-κB. Taken together, our data suggest that DMF treatment in the effector phase may suppress TMEV-IDD not only via enhancing the antioxidant response but also via suppressing IL-17A.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas do Citoesqueleto/metabolismo , Doenças Desmielinizantes/tratamento farmacológico , Fumarato de Dimetilo/farmacologia , Fator 2 Relacionado a NF-E2/metabolismo , Theilovirus/efeitos dos fármacos , Animais , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/virologia , Feminino , Proteína 1 Associada a ECH Semelhante a Kelch , Camundongos , Camundongos Endogâmicos
14.
J Virol ; 89(3): 1867-78, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25428872

RESUMO

UNLABELLED: Intracerebral infection with Theiler's murine encephalomyelitis virus (TMEV) induces immune-mediated demyelinating disease in susceptible SJL/J mice but not in resistant C57BL/6 mice. Previous studies have indicated that the major histocompatibility complex (MHC) genes play the most prominent role in the development of TMEV-induced demyelinating disease. In this study, we used C57BL/6.S (B6.S) congenic mice, which carry H-2(s) MHC genes instead of H-2(b) MHC genes in conjunction with the C57BL/6 (B6) background genes. Our data show that virus-infected B6.S mice are free from disease and have significantly lower viral loads than susceptible SJL mice, particularly in the spinal cord. A strong protective Th1-type T helper response with virtually no pathogenic Th17 response was detected in B6.S mice, in contrast to the reduced Th1- and robust Th17-type responses in SJL mice. Notably, lower levels of viral infectivity in B6.S antigen-presenting cells (APCs) correlated with the disease resistance and T-cell-type response. In vitro studies using APCs from B6.S and SJL mice show that TLR2, -3, -4, and -7, but not TLR9, signaling can replace viral infection and augment the effect of viral infection in the differentiation of the pathogenic Th17 cell type. Taken together, these results strongly suggest that the viral replication levels in APCs critically affect the induction of protective versus pathogenic Th cell types via the signaling of pattern recognition receptors for innate immune responses. Our current findings further imply that the levels of viral infectivity/replication and TLR-mediated signaling play critical roles in the pathogenesis of chronic viral diseases. IMPORTANCE: This study indicates that innate immune cytokines produced in antigen-presenting cells stimulating the T cell immune responses during early viral infection play a critical role in determining the susceptibility of mice to the development of demyelinating disease. The level of innate immune cytokines reflects the level of initial viral infection in the antigen-presenting cells, and the level determines the development of T cell types, which are either protective or pathogenic. The level of initial viral infection to the cells is controlled by a gene or genes that are not associated with the major histocompatibility antigen complex genes. This finding has an important implication in controlling not only chronic viral infections but also infection-induced autoimmune-like diseases, which are closely associated with the pathogenic type of T cell responses.


Assuntos
Células Apresentadoras de Antígenos/virologia , Infecções por Cardiovirus/patologia , Infecções por Cardiovirus/virologia , Doenças Desmielinizantes/patologia , Doenças Desmielinizantes/virologia , Theilovirus/isolamento & purificação , Carga Viral , Animais , Células Apresentadoras de Antígenos/imunologia , Infecções por Cardiovirus/imunologia , Doenças Desmielinizantes/imunologia , Feminino , Genes MHC Classe I , Camundongos Endogâmicos C57BL , Células Th1/imunologia , Células Th17/imunologia , Theilovirus/imunologia , Receptores Toll-Like/imunologia
15.
Bio Protoc ; 5(2)2015 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-27148559

RESUMO

Variety of immunological and biochemical studies associated with infection or inflammation in the central nervous system (CNS) utilize CNS-resident and/or infiltrating cells which were isolated from the CNS of naïve and affected mice in order to investigate the underlying mechanisms and the potential roles of the cell populations. Mechanical and enzyme-based single cell preparations of CNS cells are subjected to a density gradient to obtain functional single cells. In combination with cell-specific biomarkers, the function and/or status of resident microglia and infiltrating lymphocytes, including B and T cells as well as macrophages, can be characterized.

16.
J Neuroimmunol ; 274(1-2): 78-85, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25027060

RESUMO

Programmed death-1 (PD-1) belongs to the CD28 family of co-stimulatory and co-inhibitory molecules and regulates adaptive immunity. This molecule induces the development of regulatory T cells, T cell tolerance, or apoptosis. We examined the role of PD-1 pathway in Theiler's murine encephalomyelitis virus (TMEV)-induced demyelinating disease (TMEV-IDD) mice. Up-regulation of PD-1 and PD-1 ligand-1 (PD-L1) mRNA expression in bone marrow-derived dendritic cells were induced by TMEV infection in vitro. Furthermore, PD-1 and PD-L1 mRNA expression was increased in the spinal cords of the TMEV-infected mice in vivo. Treatment with a blocking monoclonal antibody (mAb) against PD-1, especially during the effector phase, resulted in significant deterioration of the TMEV-IDD both clinically and histologically. Flow cytometric analysis revealed a dramatically increase of CD4(+) T cells producing Th1 cytokines such as IFN-γ and TNF-α in the spinal cord of anti-PD-1 mAb-treated mice. These results indicate that the PD-1 pathway plays a pivotal regulatory role in the development of TMEV-IDD.


Assuntos
Antígeno B7-H1/imunologia , Doenças Desmielinizantes/imunologia , Receptor de Morte Celular Programada 1/imunologia , Medula Espinal/imunologia , Theilovirus/imunologia , Animais , Anticorpos Bloqueadores/imunologia , Anticorpos Bloqueadores/farmacologia , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linhagem Celular , Cricetinae , Doenças Desmielinizantes/metabolismo , Doenças Desmielinizantes/virologia , Células Dendríticas/imunologia , Feminino , Citometria de Fluxo , Expressão Gênica/imunologia , Interferon gama/imunologia , Interferon gama/metabolismo , Rim/citologia , Camundongos , Camundongos Endogâmicos , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Células Th1/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
17.
J Virol ; 88(15): 8479-89, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24829345

RESUMO

UNLABELLED: Interleukin-6 (IL-6) plays an important role in the development and progression of inflammatory responses, autoimmune diseases, and cancers. Many viral infections, including Theiler's murine encephalomyelitis virus (TMEV), result in the vigorous production of IL-6. However, the role of IL-6 in the development of virus-induced inflammatory responses is unclear. The infection of susceptible mice with TMEV induces the development of chronic demyelinating disease, which is considered a relevant infectious model for multiple sclerosis. In this study, we demonstrate that resistant C57BL/6 mice carrying an IL-6 transgene (IL-6 Tg) develop a TMEV-induced demyelinating disease accompanied by an increase in viral persistence and an elevated Th17 cell response in the central nervous system. Either IL-6 or IL-17 induced the expression of Bcl-2 and Bcl-xL at a high concentration. The upregulated expression of prosurvival molecules in turn inhibited target cell destruction by virus-specific CD8(+) T cells. More interestingly, IL-6 and IL-17 synergistically promoted the expression of these prosurvival molecules, preventing cellular apoptosis at a much lower (<5-fold) concentration. The signals involved in the synergy appear to include the activation of both STAT3 and NF-κB via distinct cytokine-dependent pathways. Thus, the excessive IL-6 promotes the generation of Th17 cells, and the resulting IL-6 and IL-17 synergistically promote viral persistence by protecting virus-infected cells from apoptosis and CD8(+) T cell-mediated target destruction. These results suggest that blocking both IL-6 and IL-17 functions are important considerations for therapies of chronic viral diseases, autoimmune diseases, and cancers. IMPORTANCE: This study indicates that an excessive level of IL-6 cytokine produced following viral infection promotes the development of IL-17-producing pathogenic helper T cells. We demonstrate here for the first time that IL-6 together with IL-17 synergistically enhances the expression of survival molecules to hinder critical host defense mechanisms removing virus-infected cells. This finding has an important implication in controlling not only chronic viral infections but also autoimmune diseases and cancers, which are associated with prolonged cell survival.


Assuntos
Apoptose , Evasão da Resposta Imune , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Linfócitos T Citotóxicos/imunologia , Theilovirus/imunologia , Theilovirus/fisiologia , Animais , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
18.
Int Immunol ; 26(7): 369-81, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24486565

RESUMO

Infection by Theiler's murine encephalomyelitis virus (TMEV) in the central nervous system (CNS) induces an immune-mediated demyelinating disease in susceptible mouse strains and serves as a relevant infection model for human multiple sclerosis. T-cell immunoglobulin and mucin domain-3 (TIM-3) has been demonstrated to play a crucial role in the maintenance of peripheral tolerance. In this study, we examined the regulatory role of the TIM-3 pathway in the development of TMEV-induced demyelinating disease (TMEV-IDD). The expression of TIM-3 was increased at both protein and mRNA levels in the spinal cords of mice with TMEV-IDD compared with naive controls. In addition, by utilizing a blocking mAb, we demonstrate that TIM-3 negatively regulates TMEV-specific ex vivo production of IFN-γ and IL-10 by CD4(+) T cells and IFN-γ by CD8(+) T cells from the CNS of mice with TMEV-IDD at 36 days post-infection (dpi). In vivo blockade of TIM-3 by using the anti-TIM-3 mAb resulted in significant exacerbation of the development of TMEV-IDD both clinically and histologically. The number of infiltrating mononuclear cells in the CNS was also increased in mice administered with anti-TIM-3 mAb both at the induction phase (10 dpi) and at the effector phase (36 dpi). Flow cytometric analysis of intracellular cytokines revealed that the number of CD4(+) T cells producing TNF, IL-4, IL-10 and IL-17 was significantly increased at the effector phase in the CNS of anti-TIM-3 mAb-treated mice. These results suggest that the TIM-3 pathway plays a critical role in the regulation of TMEV-IDD.


Assuntos
Poliomielite/genética , RNA Mensageiro/imunologia , Receptores Virais/imunologia , Medula Espinal/imunologia , Theilovirus/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Receptor Celular 2 do Vírus da Hepatite A , Humanos , Interferon gama/biossíntese , Interferon gama/metabolismo , Interleucina-10/biossíntese , Interleucina-10/metabolismo , Interleucina-17/biossíntese , Interleucina-17/metabolismo , Interleucina-4/biossíntese , Interleucina-4/metabolismo , Camundongos , Esclerose Múltipla , Tolerância Periférica , Poliomielite/imunologia , Poliomielite/patologia , Poliomielite/virologia , RNA Mensageiro/genética , Receptores Virais/genética , Transdução de Sinais , Medula Espinal/patologia , Theilovirus/patogenicidade
19.
J Neuroimmunol ; 268(1-2): 25-34, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24434077

RESUMO

We examined the regulatory role of αv integrins in the development of Theiler's murine encephalomyelitis virus-induced demyelinating disease (TMEV-IDD), a model of multiple sclerosis (MS). Blockade of αv integrins by anti-αv integrin monoclonal antibody (mAb) in the effector phase significantly suppressed the development of TMEV-IDD both clinically and histologically. The number of infiltrating mononuclear cells (MNCs) in the CNS was significantly decreased in mice treated with anti-αv integrin mAb. Flow cytometric analysis of cytokine staining revealed that absolute numbers of IFN-γ- and IL-17-producing CD4+ and IFN-γ-producing CD8+ T cells were significantly decreased in the CNS of mice treated with anti-αv integrin mAb. These data suggest that αv integrins may play important roles in the development of TMEV-IDD.


Assuntos
Anticorpos Monoclonais/farmacologia , Infecções por Cardiovirus/imunologia , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/imunologia , Integrina alfaV/imunologia , Animais , Infecções por Cardiovirus/patologia , Doenças Autoimunes Desmielinizantes do Sistema Nervoso Central/patologia , Feminino , Citometria de Fluxo , Imunofluorescência , Camundongos , Reação em Cadeia da Polimerase em Tempo Real , Theilovirus
20.
J Virol ; 87(21): 11538-51, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23966393

RESUMO

Infection with Theiler's murine encephalomyelitis virus (TMEV) in the central nervous system (CNS) of susceptible mice results in an immune-mediated demyelinating disease which is considered a relevant viral model of human multiple sclerosis. We previously demonstrated that the expression of positive costimulatory molecules (CD40, CD80, and CD86) is higher on the microglia of TMEV-resistant C57BL/6 (B6) mice than the microglia of TMEV-susceptible SJL/J (SJL) mice. In this study, we analyzed the expression levels of the negative costimulatory molecules PD-1 and PDL-1 in the CNS of TMEV-infected SJL mice and B6 mice. Our results indicated that TMEV infection induces the expression of both PD-1 and PDL-1 on microglia and macrophages in the CNS but not in the periphery. The expression of PD-1 only on CNS-infiltrating macrophages and not on resident microglia was considerably higher (>4-fold) in TMEV-infected SJL mice than TMEV-infected B6 mice. We further demonstrated that interleukn-6 (IL-6) is necessary to induce the maximal expression of PDL-1 but not PD-1 after TMEV infection using IL-6-deficient mice and IL-6-transgenic mice in conjunction with recombinant IL-6. In addition, cells from type I interferon (IFN) receptor knockout mice failed to upregulate PD-1 and PDL-1 expression after TMEV infection in vitro, indicating that type I IFN signaling is associated with the upregulation. However, other IFN signaling may also participate in the upregulation. Taken together, these results strongly suggest that the expression of PD-1 and PDL-1 in the CNS is primarily upregulated following TMEV infection via type I IFN signaling and the maximal expression of PDL-1 additionally requires IL-6 signaling.


Assuntos
Antígeno B7-H1/metabolismo , Infecções por Cardiovirus/virologia , Sistema Nervoso Central/virologia , Interleucina-6/imunologia , Microglia/virologia , Receptor de Morte Celular Programada 1/metabolismo , Theilovirus/imunologia , Animais , Infecções por Cardiovirus/imunologia , Feminino , Macrófagos/virologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...