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
1.
J Infect Dis ; 229(2): 376-383, 2024 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-37565807

RESUMO

BACKGROUND: The United States has experienced a resurgence of pertussis following the introduction of acellular pertussis (aP) vaccines. This is likely due to the failure of aP vaccines to induce durable immunity and prevent infection, carriage, and transmission. METHODS: To evaluate the impact of aP vaccination on the immune response to infection and test the ability of infection to reprogram aP-imprinted immune responses, we challenged unvaccinated and aP-vaccinated baboons with Bordetella pertussis multiple times and accessed the immune responses and outcomes of infections after each exposure. RESULTS: Multiple infections were required to elicit T-helper 17 responses and protection in aP-vaccinated animals comparable to responses seen in unvaccinated animals after a single challenge. Even after 3 challenges, T-helper 1 responses were not observed in aP-vaccinated animals. Immunoglobulin G responses to vaccine and nonvaccine antigens were not negatively affected in aP-vaccinated animals. CONCLUSIONS: Our results indicate that it is possible to retrain aP-primed immune responses, but it will likely require an optimal booster and multiple doses. Our results in the baboon model suggest that circulation of B. pertussis in aP-vaccinated populations is concentrated in the younger age bands of the population, providing information that can guide improved modeling of B. pertussis epidemiology in aP-vaccinated populations.


Assuntos
Coqueluche , Animais , Coqueluche/prevenção & controle , Bordetella pertussis , Papio , Anticorpos Antibacterianos , Vacina contra Coqueluche , Vacinas Acelulares
2.
Curr Opin Immunol ; 59: 72-78, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31078081

RESUMO

Despite high vaccine coverage, reported cases of pertussis have increased steadily over the last twenty years. This resurgence has stimulated interest in host responses to pertussis infection and vaccination with the goal of developing more effective next-generation vaccines and vaccination strategies. Optimal protection against Bordetella pertussis appears to be multifactorial requiring both humoral and cellular responses. Natural infection and whole-cell pertussis vaccination induce Th1 and Th17-dominated responses. In contrast, acellular vaccines induce Th2-dominated responses. Available immunological data indicate that while antibodies provide protection against disease, Th1 and Th17-mediated immune responses are required for bacterial clearance and long-lasting protection. The nature of the priming in children appears to be important in modulating bias and durability of immune responses required to provide protection against B. pertussis. This review summarizes the current understanding of differences in immune responses and their role in protection against B. pertussis following infection or vaccination.


Assuntos
Bordetella pertussis/fisiologia , Vacina contra Coqueluche/imunologia , Células Th1/imunologia , Células Th17/imunologia , Coqueluche/imunologia , Animais , Anticorpos Antibacterianos/metabolismo , Criança , Humanos , Imunidade Celular , Imunidade Humoral , Memória Imunológica , Ativação Linfocitária , Vacinação , Vacinas Acelulares
3.
J Infect Dis ; 217(8): 1231-1236, 2018 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-29346585

RESUMO

Background: Bordetella pertussis is a human pathogen responsible for serious respiratory illness. The disease is most severe in infants too young to be vaccinated with most hospitalizations and deaths occurring within this age group. The Advisory Committee on Immunization Practices recommended immunization of pregnant women to protect infants from birth until their first vaccination at 6-8 weeks of age. We previously demonstrated that maternal vaccination with licensed acellular pertussis vaccines protected newborn baboons from disease. We hypothesized that protection was due to toxin-neutralizing, maternal anti-pertussis toxin antibodies and predicted that maternal vaccination with a pertussis toxoid (PTx)-only vaccine would protect newborns from disease. Methods: Infant baboons born to unvaccinated mothers or mothers vaccinated with a PTx-only vaccine were challenged with B. pertussis at 5 weeks of age and followed for infection and signs of disease. Results: Although all challenged infants were heavily colonized, the infant baboons born to mothers vaccinated with PTx-only vaccine were free from clinical disease following exposure to B. pertussis. In contrast, disease was observed in infants born to unvaccinated mothers. Conclusions: Our results demonstrated that maternal vaccination with a PTx-only vaccine is sufficient to protect newborn baboons from disease following exposure to pertussis.


Assuntos
Imunidade Materno-Adquirida/imunologia , Vacina contra Coqueluche/imunologia , Coqueluche/prevenção & controle , Animais , Carga Bacteriana , Bordetella parapertussis , Modelos Animais de Doenças , Feminino , Papio , Gravidez
4.
J Virol ; 89(18): 9299-312, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26136579

RESUMO

UNLABELLED: Myd88 signaling is critical to the control of numerous central nervous system (CNS) infections by promoting both innate and adaptive immune responses. Nevertheless, the extent to which Myd88 regulates type I interferon (IFN) versus proinflammatory factors and T cell function, as well as the anatomical site of action, varies extensively with the pathogen. CNS infection by neurotropic coronavirus with replication confined to the brain and spinal cord induces protective IFN-α/ß via Myd88-independent activation of melanoma differentiation-associated gene 5 (MDA5). However, a contribution of Myd88-dependent signals to CNS pathogenesis has not been assessed. Infected Myd88(-/-) mice failed to control virus, exhibited enhanced clinical disease coincident with increased demyelination, and succumbed to infection within 3 weeks. The induction of IFN-α/ß, as well as of proinflammatory cytokines and chemokines, was impaired early during infection. However, defects in both IFN-α/ß and select proinflammatory factors were rapidly overcome prior to T cell recruitment. Myd88 deficiency also specifically blunted myeloid and CD4 T cell recruitment into the CNS without affecting CD8 T cells. Moreover, CD4 T cells but not CD8 T cells were impaired in IFN-γ production. Ineffective virus control indeed correlated most prominently with reduced antiviral IFN-γ in the CNS of Myd88(-/-) mice. The results demonstrate a crucial role for Myd88 both in early induction of innate immune responses during coronavirus-induced encephalomyelitis and in specifically promoting protective CD4 T cell activation. In the absence of these responses, functional CD8 T cells are insufficient to control viral spread within the CNS, resulting in severe demyelination. IMPORTANCE: During central nervous system (CNS) infections, signaling through the adaptor protein Myd88 promotes both innate and adaptive immune responses. The extent to which Myd88 regulates antiviral type I IFN, proinflammatory factors, adaptive immunity, and pathology is pathogen dependent. These results reveal that Myd88 protects from lethal neurotropic coronavirus-induced encephalomyelitis by accelerating but not enhancing the induction of IFN-α/ß, as well as by promoting peripheral activation and CNS accumulation of virus-specific CD4 T cells secreting IFN-γ. By controlling both early innate immune responses and CD4 T cell-mediated antiviral IFN-γ, Myd88 signaling limits the initial viral dissemination and is vital for T cell-mediated control of viral loads. Uncontrolled viral replication in the absence of Myd88 leads to severe demyelination and pathology despite overall reduced inflammatory responses. These data support a vital role of Myd88 signaling in protective antimicrobial functions in the CNS by promoting proinflammatory mediators and T cell-mediated IFN-γ production.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Infecções por Coronavirus/imunologia , Encefalite Viral/imunologia , Imunidade Celular , Imunidade Inata , Vírus Elberfeld do Camundongo/imunologia , Fator 88 de Diferenciação Mieloide/imunologia , Animais , Linfócitos T CD4-Positivos/patologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/patologia , Infecções por Coronavirus/genética , Infecções por Coronavirus/patologia , Encefalite Viral/genética , Encefalite Viral/patologia , Interferon-alfa/genética , Interferon-alfa/imunologia , Interferon beta/genética , Interferon beta/imunologia , Vírus Elberfeld do Camundongo/genética , Camundongos , Camundongos Knockout , Fator 88 de Diferenciação Mieloide/genética
5.
J Immunol ; 193(1): 285-94, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24890725

RESUMO

IL-27 is a pleiotropic member of the IL-6 and IL-12 cytokine family composed of the IL-27p28 and the EBV-induced gene 3. IL-27 and its receptor mRNA are both upregulated in the CNS during acute encephalomyelitis induced by the JHM strain of mouse hepatitis virus (JHMV) and sustained during viral persistence. Contributions of IL-27 to viral pathogenesis were evaluated by infection of IL-27Rα-chain-deficient (IL-27Rα(-/-)) mice. The absence of IL-27 signaling accelerated virus control within the CNS associated with increased IFN-γ secreting virus-specific CD4+ and CD8+ T cells. Abrogation of IL-27 signaling did not affect virus-specific CD8+ T cell-mediated IL-10 production or cytolytic activity or Foxp3+ regulatory T cell populations. However, IL-10 production by virus-specific CD4+ T cells was reduced significantly. Despite increased T cell-mediated antiviral function in IL-27Rα(-/-) mice, the virus persisted in the CNS at similar levels as in wild-type mice. Nevertheless, IL-27Rα(-/-) mice exhibited decreased clinical disease during persistence, coincident with less severe demyelination, the hallmark tissue damage associated with JHMV infection. Overall, these data demonstrate that in contrast to viral infections at other sites, IL-27 does not play a proinflammatory role during JHMV-induced encephalomyelitis. Rather, it limits CNS inflammation and impairs control of CNS virus replication via induction of IL-10 in virus-specific CD4+ T cells. Furthermore, in contrast to its protective role in limiting CNS autoimmunity and preventing immunopathology, these data define a detrimental role of IL-27 in promoting demyelination by delaying viral control.


Assuntos
Sistema Nervoso Central/imunologia , Infecções por Coronavirus/imunologia , Encefalomielite Aguda Disseminada/imunologia , Interleucina-10/imunologia , Interleucinas/imunologia , Vírus da Hepatite Murina/imunologia , Transdução de Sinais/imunologia , Animais , Sistema Nervoso Central/patologia , Infecções por Coronavirus/genética , Infecções por Coronavirus/patologia , Doenças Desmielinizantes , Encefalomielite Aguda Disseminada/genética , Encefalomielite Aguda Disseminada/patologia , Interleucina-10/genética , Interleucinas/genética , Camundongos , Camundongos Knockout , Transdução de Sinais/genética , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/patologia
6.
J Neuroimmunol ; 270(1-2): 1-12, 2014 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-24642385

RESUMO

Double-stranded RNA-dependent protein kinase (PKR) regulates antiviral activity, immune responses, apoptosis and neurotoxicity. Gliatropic coronavirus infection induced PKR activation in infected as well uninfected cells within the central nervous system (CNS). However, PKR deficiency only modestly increased viral replication and did not affect IFN-α/ß or IL-1ß expression. Despite reduced Il-6, Ccl5, and Cxcl10 mRNA, protein levels remained unaltered. Furthermore, PKR deficiency selectively reduced IL-10 production in CD4, but not CD8 T cells, without affecting CNS pathology. The results demonstrate the ability of PKR to balance neuroinflammation by selectively modulating key cytokines and chemokines in CNS resident and CD4 T cells.


Assuntos
Infecções por Coronavirus/imunologia , Encefalite Viral/imunologia , Interleucina-10/imunologia , eIF-2 Quinase/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Infecções por Coronavirus/metabolismo , Encefalite Viral/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-10/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Reação em Cadeia da Polimerase em Tempo Real , eIF-2 Quinase/metabolismo
7.
J Virol ; 88(2): 1051-64, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24198415

RESUMO

Type I interferons (IFN-α/ß) limit viral dissemination prior to the emergence of adaptive immune responses through the concerted action of interferon-stimulated genes (ISGs). Although IFN-α/ß induction by coronaviruses is modest, it effectively limits viral spread within the central nervous system (CNS) and protects against mortality. The protective roles of specific ISGs against the mouse hepatitis virus (MHV) members of the coronaviruses are largely unknown. This study demonstrates a protective role of the ISG Ifit2 in encephalitis induced by the dual hepato- and neurotropic MHV-A59. Contrasting the mild encephalitis and 100% survival of MHV-A59-infected wild-type (wt) mice, nearly 60% of infected Ifit2(-/-) mice exhibited severe encephalitis and succumbed between 6 and 8 days postinfection. Increased clinical disease in Ifit2(-/-) mice coincided with higher viral loads and enhanced viral spread throughout the CNS parenchyma. Ifit2(-/-) mice also expressed significantly reduced IFN-α/ß and downstream ISG mRNAs Ifit1, Isg15, and Pkr, while expression of proinflammatory cytokines and chemokines was only modestly affected in the CNS. Impaired IFN-α/ß induction in the absence of Ifit2 was confirmed by ex vivo mRNA analysis of microglia and macrophages, the prominent cell types producing IFN-α/ß following MHV CNS infection. Furthermore, both IFN-α/ß mRNA and protein production were significantly reduced in MHV-infected Ifit2(-/-) relative to wt bone marrow-derived macrophages. Collectively, the data implicate Ifit2 as a positive regulator of IFN-α/ß expression, rather than direct antiviral mediator, during MHV-induced encephalitis.


Assuntos
Sistema Nervoso Central/virologia , Encefalite/veterinária , Interferon-alfa/genética , Interferon beta/genética , Macrófagos/imunologia , Vírus da Hepatite Murina/fisiologia , Proteínas/imunologia , Doenças dos Roedores/imunologia , Animais , Proteínas Reguladoras de Apoptose , Sistema Nervoso Central/imunologia , Encefalite/genética , Encefalite/imunologia , Encefalite/virologia , Feminino , Interferon-alfa/imunologia , Interferon beta/imunologia , Macrófagos/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Hepatite Murina/genética , Proteínas/genética , Proteínas de Ligação a RNA , Doenças dos Roedores/genética , Doenças dos Roedores/virologia , Tropismo Viral
8.
Am J Pathol ; 182(5): 1696-704, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23499374

RESUMO

Although myeloid cell activation is requisite for an optimal innate immune response, this process must be tightly controlled to prevent collateral host tissue damage. Kruppel-like factor 2 (KLF2) is a potent regulator of myeloid cell proinflammatory activation. As an approximately 30% to 50% reduction in KLF2 levels has been observed in human subjects with acute or chronic inflammatory disorders, we studied the biological response to inflammation in KLF2(+/-) mice. Herein, we show that partial deficiency of KLF2 modulates the in vivo response to acute (sepsis) and subacute (skin) inflammatory challenge. Mechanistically, we link the anti-inflammatory effects of KLF2 to the inhibition of NF-κB transcriptional activity. Collectively, the observations provide biologically relevant insights into KLF2-mediated modulation of these inflammatory processes that could potentially be manipulated for therapeutic gain.


Assuntos
Inflamação/genética , Inflamação/patologia , Fatores de Transcrição Kruppel-Like/metabolismo , Transcrição Gênica , Doença Aguda , Animais , Carragenina , Linhagem Celular , Doença Crônica , Modelos Animais de Doenças , Progressão da Doença , Edema/genética , Edema/patologia , Regulação da Expressão Gênica , Hemizigoto , Humanos , Fatores de Transcrição Kruppel-Like/deficiência , Lipopolissacarídeos , Camundongos , NF-kappa B/metabolismo , Sepse/genética , Sepse/patologia , Pele/patologia
9.
Arterioscler Thromb Vasc Biol ; 32(12): 2836-8, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23065827

RESUMO

OBJECTIVE: To investigate the role of Krüppel-like factor 4 (KLF4), an essential transcriptional regulator of macrophage polarization (M1/M2), in the pathogenesis of atherosclerosis. METHODS AND RESULTS: Despite the acknowledged importance of macrophages in atherosclerosis, the role of M1 (classically activated or proinflammatory) versus M2 (alternatively activated or anti-inflammatory) macrophages in this process remains incompletely understood. We recently identified KLF4 as a regulator of macrophage subset specification; that is, KLF4 promotes M2 and inhibits M1 phenotype. Here, we provide evidence that KLF4-deficient macrophages exhibit enhanced proinflammatory activation and foam cell formation in response to oxidized lipids. In vivo, myeloid KLF4-deficient mice (ApoE(-/-) background) develop significantly more vascular inflammation and atherosclerotic lesion formation. CONCLUSIONS: Our findings identify myeloid KLF4 as an essential regulator of vascular inflammation and experimental atherogenesis.


Assuntos
Apolipoproteínas E/deficiência , Aterosclerose/fisiopatologia , Fatores de Transcrição Kruppel-Like/deficiência , Fatores de Transcrição Kruppel-Like/fisiologia , Animais , Apolipoproteínas E/genética , Apolipoproteínas E/fisiologia , Aterosclerose/patologia , Modelos Animais de Doenças , Células Espumosas/patologia , Células Espumosas/fisiologia , Fator 4 Semelhante a Kruppel , Fatores de Transcrição Kruppel-Like/genética , Macrófagos/patologia , Macrófagos/fisiologia , Camundongos , Camundongos Knockout , Fenótipo
10.
Glia ; 60(10): 1555-66, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22736486

RESUMO

Type I interferons (IFNα/ß) provide a primary defense against infection. Nevertheless, the dynamics of IFNα/ß induction and responsiveness by central nervous system (CNS) resident cells in vivo in response to viral infections are poorly understood. Mice were infected with a neurotropic coronavirus with tropism for oligodendroglia and microglia to probe innate antiviral responses during acute encephalomyelitis. Expression of genes associated with the IFNα/ß pathways was monitored in microglia and oligodendroglia purified from naïve and infected mice by fluorescent activated cell sorting. Compared with microglia, oligodendroglia were characterized by low basal expression of mRNA encoding viral RNA sensing pattern recognition receptors (PRRs), IFNα/ß receptor chains, interferon sensitive genes (ISG), as well as kinases and transcription factors critical in IFNα/ß signaling. Although PRRs and ISGs were upregulated by infection in both cell types, the repertoire and absolute mRNA levels were more limited in oligodendroglia. Furthermore, although oligodendroglia harbored higher levels of viral RNA compared with microglia, Ifnα/ß was only induced in microglia. Stimulation with the double stranded RNA analogue poly I:C also failed to induce Ifnα/ß in oligodendroglia, and resulted in reduced and delayed induction of ISGs compared with microglia. The limited antiviral response by oligodendroglia was associated with a high threshold for upregulation of Ikkε and Irf7 transcripts, both central to amplifying IFNα/ß responses. Overall, these data reveal that oligodendroglia from the adult CNS are poor sensors of viral infection and suggest they require exogenous IFNα/ß to establish an antiviral state.


Assuntos
Interferon Tipo I/metabolismo , Oligodendroglia/metabolismo , Animais , Proteínas de Ligação a Calmodulina/genética , Proteínas de Ligação a Calmodulina/metabolismo , Proteína DEAD-box 58 , RNA Helicases DEAD-box/genética , RNA Helicases DEAD-box/metabolismo , Encefalomielite/genética , Encefalomielite/metabolismo , Encefalomielite/patologia , Citometria de Fluxo , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Indutores de Interferon/farmacologia , Helicase IFIH1 Induzida por Interferon , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Proteína Proteolipídica de Mielina/genética , Proteína Proteolipídica de Mielina/metabolismo , Oligodendroglia/efeitos dos fármacos , Poli I-C/farmacologia , RNA Mensageiro/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Virais/genética , Proteínas Virais/metabolismo
11.
J Virol ; 85(13): 6136-47, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21507985

RESUMO

Immunoglobulin in cerebral spinal fluid and antibody secreting cells (ASC) within the central nervous system (CNS) parenchyma are common hallmarks of microbial infections and autoimmune disorders. However, the signals directing ASC migration into the inflamed CNS are poorly characterized. This study demonstrates that CXCR3 mediates CNS accumulation of ASC during neurotropic coronavirus-induced encephalomyelitis. Expansion of CXCR3-expressing ASC in draining lymph nodes prior to accumulation within the CNS was consistent with their recruitment by sustained expression of CXCR3 ligands during viral persistence. Both total and virus-specific ASC were reduced greater than 80% in the CNS of infected CXCR3(-/-) mice. Similar T cell CNS recruitment and local T cell-dependent antiviral activity further indicated that the ASC migration defect was T cell independent. Furthermore, in contrast to the reduction of ASC in the CNS, neither virus-specific ASC trafficking to bone marrow nor antiviral serum antibody was reduced relative to levels in control mice. Impaired ASC recruitment into the CNS of infected CXCR3(-/-) mice coincided with elevated levels of persisting viral RNA, sustained infectious virus, increased clinical disease, and mortality. These results demonstrate that CXCR3 ligands are indispensable for recruitment of activated ASC into the inflamed CNS and highlight their local protective role during persistent infection.


Assuntos
Movimento Celular/imunologia , Sistema Nervoso Central/imunologia , Encefalomielite/imunologia , Encefalomielite/virologia , Vírus da Hepatite Murina/imunologia , Plasmócitos/fisiologia , Receptores CXCR3/metabolismo , Animais , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Vírus da Hepatite Murina/patogenicidade , Plasmócitos/citologia , Receptores CXCR3/genética
12.
PLoS Pathog ; 5(10): e1000602, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19798426

RESUMO

IFN-alpha/beta plays a critical role in limiting viral spread, restricting viral tropism and protecting mice from neurotropic coronavirus infection. However, the IFN-alpha/beta dependent mechanisms underlying innate anti-viral functions within the CNS are poorly understood. The role of RNase L in viral encephalomyelitis was explored based on its functions in inhibiting translation, inducing apoptosis, and propagating the IFN-alpha/beta pathway through RNA degradation intermediates. Infection of RNase L deficient (RL(-/-)) mice with a sub-lethal, demyelinating mouse hepatitis virus variant revealed that the majority of mice succumbed to infection by day 12 p.i. However, RNase L deficiency did not affect overall control of infectious virus, or diminish IFN-alpha/beta expression in the CNS. Furthermore, increased morbidity and mortality could not be attributed to altered proinflammatory signals or composition of cells infiltrating the CNS. The unique phenotype of infected RL(-/-) mice was rather manifested in earlier onset and increased severity of demyelination and axonal damage in brain stem and spinal cord without evidence for enhanced neuronal infection. Increased tissue damage coincided with sustained brain stem infection, foci of microglia infection in grey matter, and increased apoptotic cells. These data demonstrate a novel protective role for RNase L in viral induced CNS encephalomyelitis, which is not reflected in overall viral control or propagation of IFN-alpha/beta mediated signals. Protective function is rather associated with cell type specific and regional restriction of viral replication in grey matter and ameliorated neurodegeneration and demyelination.


Assuntos
Encéfalo/patologia , Infecções por Coronavirus/enzimologia , Doenças Desmielinizantes/patologia , Encefalite Viral/imunologia , Encefalite Viral/metabolismo , Endorribonucleases/metabolismo , Animais , Apoptose/fisiologia , Encéfalo/enzimologia , Encéfalo/imunologia , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/patologia , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/virologia , Encefalite Viral/patologia , Endorribonucleases/genética , Endorribonucleases/imunologia , Citometria de Fluxo , Imunofluorescência , Inflamação/enzimologia , Inflamação/genética , Inflamação/imunologia , Interferon-alfa/imunologia , Interferon-alfa/metabolismo , Interferon beta/imunologia , Interferon beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Vírus da Hepatite Murina , Degeneração Neural/enzimologia , Degeneração Neural/imunologia , Degeneração Neural/patologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Medula Espinal/enzimologia , Medula Espinal/imunologia , Medula Espinal/patologia
13.
J Virol ; 83(12): 5978-86, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19339350

RESUMO

The relative contributions of interleukin-12 (IL-12) and IL-23 to viral pathogenesis have not been extensively studied. IL-12p40 mRNA rapidly increases after neurotropic coronavirus infection. Infection of mice defective in both IL-12 and IL-23 (p40(-/-)), in IL-12 alone (p35(-/-)), and in IL-23 alone (p19(-/-)) revealed that the symptoms of coronavirus-induced encephalitis are regulated by IL-12. IL-17-producing cells never exceeded background levels, supporting a redundant role of IL-23 in pathogenesis. Viral control, tropism, and demyelination were all similar in p35(-/-), p19(-/-), and wild-type mice. Reduced morbidity in infected IL-12 deficient mice was also not associated with altered recruitment or composition of inflammatory cells. However, gamma interferon (IFN-gamma) levels and virus-specific IFN-gamma-secreting CD4 and CD8 T cells were all reduced in the central nervous systems (CNS) of infected p35(-/-) mice. Transcription of the proinflammatory cytokines IL-1beta and IL-6, but not tumor necrosis factor, were initially reduced in infected p35(-/-) mice but increased to wild-type levels during peak inflammation. Furthermore, although transforming growth factor beta mRNA was not affected, IL-10 was increased in the CNS in the absence of IL-12. These data suggest that IL-12 does not contribute to antiviral function within the CNS but enhances morbidity associated with viral encephalitis by increasing the ratio of IFN-gamma to protective IL-10.


Assuntos
Infecções por Coronavirus/imunologia , Encefalite Viral/imunologia , Subunidade p40 da Interleucina-12/deficiência , Interleucina-23/deficiência , Animais , Células Cultivadas , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/virologia , Doenças Desmielinizantes/imunologia , Doenças Desmielinizantes/virologia , Interferon gama/imunologia , Interleucina-10/imunologia , Subunidade p40 da Interleucina-12/imunologia , Interleucina-23/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Vírus da Hepatite Murina/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...