Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
Mais filtros

País/Região como assunto
Intervalo de ano de publicação
1.
Cell ; 164(3): 349-52, 2016 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-26824652

RESUMO

Type I interferon (IFN-I) elicits a complex cascade of events in response to microbial infection. Here, we review recent developments illuminating the large number of IFN-I species and describing their unique biologic functions.


Assuntos
Infecções Bacterianas/imunologia , Interferon Tipo I/metabolismo , Viroses/imunologia , Animais , Infecções Bacterianas/microbiologia , Humanos , Interferon Tipo I/química , Interferon Tipo I/imunologia , Receptor de Interferon alfa e beta/metabolismo , Viroses/virologia
2.
Annu Rev Biochem ; 82: 637-62, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23527695

RESUMO

The sphingosine-1-phosphate (S1P) receptor signaling system has biological and medical importance and is the first lipid G protein-coupled receptor (GPCR) structure to be solved to 2.8-Å resolution. S1P binds to five high-affinity GPCRs generating multiple downstream signals that play essential roles in vascular development and endothelial integrity, control of cardiac rhythm, and routine oral treatment of multiple sclerosis. Genetics, chemistry, and now structural biology have advanced this integrated biochemical system. The S1P receptors have a novel N-terminal fold that occludes access to the binding pocket from the extracellular environment as well as orthosteric and bitopic ligands with very different physicochemical properties. S1P receptors and metabolizing enzymes have been deleted, inducibly deleted, and knocked in as tagged or altered receptors in mice. An array of genetic models allows analysis of integrated receptor function in vivo. We can now directly understand causal relationships among protein expression, signal, and control points in physiology and pathology.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Receptores de Lisoesfingolipídeo/metabolismo , Transdução de Sinais/fisiologia , Animais , Ligantes , Camundongos , Receptores Acoplados a Proteínas G/química , Receptores de Lisoesfingolipídeo/química , Receptores de Lisoesfingolipídeo/genética
3.
Cell ; 146(6): 980-91, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21925319

RESUMO

Cytokine storm during viral infection is a prospective predictor of morbidity and mortality, yet the cellular sources remain undefined. Here, using genetic and chemical tools to probe functions of the S1P(1) receptor, we elucidate cellular and signaling mechanisms that are important in initiating cytokine storm. Whereas S1P(1) receptor is expressed on endothelial cells and lymphocytes within lung tissue, S1P(1) agonism suppresses cytokines and innate immune cell recruitment in wild-type and lymphocyte-deficient mice, identifying endothelial cells as central regulators of cytokine storm. Furthermore, our data reveal immune cell infiltration and cytokine production as distinct events that are both orchestrated by endothelial cells. Moreover, we demonstrate that suppression of early innate immune responses through S1P(1) signaling results in reduced mortality during infection with a human pathogenic strain of influenza virus. Modulation of endothelium with a specific agonist suggests that diseases in which amplification of cytokine storm is a significant pathological component could be chemically tractable.


Assuntos
Citocinas/imunologia , Células Endoteliais/imunologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Influenza Humana/imunologia , Infecções por Orthomyxoviridae/imunologia , Animais , Modelos Animais de Doenças , Humanos , Interferons/imunologia , Pulmão/citologia , Pulmão/imunologia , Pulmão/virologia , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Infecções por Orthomyxoviridae/patologia , Infecções por Orthomyxoviridae/virologia , Receptores de Lisoesfingolipídeo/agonistas , Transdução de Sinais
4.
Nat Immunol ; 14(8): 849-57, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23812097

RESUMO

Follicular helper T cells (T(FH) cells) provide critical help to B cells during humoral immune responses. Here we report that mice with T cell-specific deletion of the miR-17∼92 family of microRNAs (miRNAs) had substantially compromised T(FH) differentiation, germinal-center formation and antibody responses and failed to control chronic viral infection. Conversely, mice with T cell-specific expression of a transgene encoding miR-17∼92 spontaneously accumulated T(FH) cells and developed a fatal immunopathology. Mechanistically, the miR-17∼92 family controlled the migration of CD4(+) T cells into B cell follicles by regulating signaling intensity from the inducible costimulator ICOS and kinase PI(3)K by suppressing expression of the phosphatase PHLPP2. Our findings demonstrate an essential role for the miR-17∼92 family in T(FH) differentiation and establish PHLPP2 as an important mediator of their function in this process.


Assuntos
Linfócitos B/imunologia , Diferenciação Celular/imunologia , Centro Germinativo/imunologia , MicroRNAs/imunologia , Proteínas Nucleares/imunologia , Fosfoproteínas Fosfatases/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Citometria de Fluxo , Centro Germinativo/citologia , Imunidade Humoral/imunologia , Imuno-Histoquímica , Proteína Coestimuladora de Linfócitos T Induzíveis/imunologia , Coriomeningite Linfocítica/imunologia , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Fosfatidilinositol 3-Quinases/imunologia , Transdução de Sinais/imunologia , Organismos Livres de Patógenos Específicos , Linfócitos T Auxiliares-Indutores/citologia , Linfócitos T Auxiliares-Indutores/enzimologia
5.
Proc Natl Acad Sci U S A ; 119(3)2022 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-35022243

RESUMO

Recent studies have identified a critical role for B cell-produced cytokines in regulating both humoral and cellular immunity. Here, we show that B cells are an essential source of interleukin-27 (IL-27) during persistent lymphocytic choriomeningitis virus (LCMV) clone 13 (Cl-13) infection. By using conditional knockout mouse models with specific IL-27p28 deletion in B cells, we observed that B cell-derived IL-27 promotes survival of virus-specific CD4 T cells and supports functions of T follicular helper (Tfh) cells. Mechanistically, B cell-derived IL-27 promotes CD4 T cell function, antibody class switch, and the ability to control persistent LCMV infection. Deletion of IL-27ra in T cells demonstrated that T cell-intrinsic IL-27R signaling is essential for viral control, optimal CD4 T cell responses, and antibody class switch during persistent LCMV infection. Collectively, our findings identify a cellular mechanism whereby B cell-derived IL-27 drives antiviral immunity and antibody responses through IL-27 signaling on T cells to promote control of LCMV Cl-13 infection.


Assuntos
Linfócitos B/metabolismo , Interleucina-27/metabolismo , Coriomeningite Linfocítica/imunologia , Imunidade Adaptativa , Animais , Anticorpos Antivirais , Linfócitos T CD4-Positivos/imunologia , Citocinas/metabolismo , Imunidade Celular , Interleucina-27/genética , Interleucinas , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
PLoS Pathog ; 16(3): e1008352, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32142546

RESUMO

Lassa virus infects hundreds of thousands of people each year across rural West Africa, resulting in a high number of cases of Lassa fever (LF), a febrile disease associated with high morbidity and significant mortality. The lack of approved treatments or interventions underscores the need for an effective vaccine. At least four viral lineages circulate in defined regions throughout West Africa with substantial interlineage nucleotide and amino acid diversity. An effective vaccine should be designed to elicit Lassa virus specific humoral and cell mediated immunity across all lineages. Most current vaccine candidates use only lineage IV antigens encoded by Lassa viruses circulating around Sierra Leone, Liberia, and Guinea but not Nigeria where lineages I-III are found. As previous infection is known to protect against disease from subsequent exposure, we sought to determine whether LF survivors from Nigeria and Sierra Leone harbor memory T cells that respond to lineage IV antigens. Our results indicate a high degree of cross-reactivity of CD8+ T cells from Nigerian LF survivors to lineage IV antigens. In addition, we identified regions within the Lassa virus glycoprotein complex and nucleoprotein that contributed to these responses while T cell epitopes were not widely conserved across our study group. These data are important for current efforts to design effective and efficient vaccine candidates that can elicit protective immunity across all Lassa virus lineages.


Assuntos
Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/imunologia , Vírus Lassa/imunologia , África Ocidental , Reações Cruzadas , Feminino , Humanos , Masculino , Especificidade da Espécie
7.
Proc Natl Acad Sci U S A ; 116(36): 18001-18008, 2019 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-31427525

RESUMO

Lymphocytic choriomeningitis virus (LCMV) WE variant 2.2 (v2.2) generated a high level of the major mouse urinary protein: MUP. Mice infected with LCMV WE v54, which differed from v2.2 by a single amino acid in the viral glycoprotein, failed to generate MUP above baseline levels found in uninfected controls. Variant 54 bound at 2.5 logs higher affinity to the LCMV receptor α-dystroglycan (α-DG) than v2.2 and entered α-DG-expressing but not α-DG-null cells. Variant 2.2 infected both α-DG-null or -expressing cells. Variant 54 infected more dendritic cells, generated a negligible CD8 T cell response, and caused a persistent infection, while v2.2 generated cytotoxic T lymphocytes (CTLs) and cleared virus within 10 days. By 20 days postinfection and through the 80-day observation period, significantly higher amounts of MUP were found in v2.2-infected mice. Production of MUP was dependent on virus-specific CTL as deletion of such cells aborted MUP production. Furthermore, MUP production was not elevated in v2.2 persistently infected mice unless virus was cleared following transfer of virus-specific CTL.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Regulação da Expressão Gênica/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Proteínas/imunologia , Animais , Distroglicanas/imunologia , Coriomeningite Linfocítica/patologia , Camundongos
8.
J Virol ; 94(12)2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32269122

RESUMO

Early and robust T cell responses have been associated with survival from Lassa fever (LF), but the Lassa virus-specific memory responses have not been well characterized. Regions within the virus surface glycoprotein (GPC) and nucleoprotein (NP) are the main targets of the Lassa virus-specific T cell responses, but, to date, only a few T cell epitopes within these proteins have been identified. We identified GPC and NP regions containing T cell epitopes and HLA haplotypes from LF survivors and used predictive HLA-binding algorithms to identify putative epitopes, which were then experimentally tested using autologous survivor samples. We identified 12 CD8-positive (CD8+) T cell epitopes, including epitopes common to both Nigerian and Sierra Leonean survivors. These data should be useful for the identification of dominant Lassa virus-specific T cell responses in Lassa fever survivors and vaccinated individuals as well as for designing vaccines that elicit cell-mediated immunity.IMPORTANCE The high morbidity and mortality associated with clinical cases of Lassa fever, together with the lack of licensed vaccines and limited and partially effective interventions, make Lassa virus (LASV) an important health concern in its regions of endemicity in West Africa. Previous infection with LASV protects from disease after subsequent exposure, providing a framework for designing vaccines to elicit similar protective immunity. Multiple major lineages of LASV circulate in West Africa, and therefore, ideal vaccine candidates should elicit immunity to all lineages. We therefore sought to identify common T cell epitopes between Lassa fever survivors from Sierra Leone and Nigeria, where distinct lineages circulate. We identified three such epitopes derived from highly conserved regions within LASV proteins. In this process, we also identified nine other T cell epitopes. These data should help in the design of an effective pan-LASV vaccine.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Epitopos de Linfócito T/química , Febre Lassa/imunologia , Vírus Lassa/imunologia , Nucleoproteínas/imunologia , Proteínas do Envelope Viral/imunologia , Adolescente , Sequência de Aminoácidos , Animais , Anticorpos Antivirais/biossíntese , Antígenos Virais/química , Antígenos Virais/genética , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/virologia , Criança , Epitopos de Linfócito T/genética , Epitopos de Linfócito T/imunologia , Feminino , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/imunologia , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/imunologia , Haplótipos , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Humanos , Soros Imunes/análise , Memória Imunológica , Febre Lassa/genética , Febre Lassa/patologia , Vírus Lassa/patogenicidade , Masculino , Nigéria , Nucleoproteínas/genética , Serra Leoa , Sobreviventes , Proteínas do Envelope Viral/genética , Adulto Jovem
9.
Proc Natl Acad Sci U S A ; 115(33): E7814-E7823, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-30061383

RESUMO

Understanding of T cell exhaustion and successful therapy to restore T cell function was first described using Clone (Cl) 13 variant selected from the lymphocytic choriomeningitis virus (LCMV) Armstrong (ARM) 53b parental strain. T cell exhaustion plays a pivotal role in both persistent infections and cancers of mice and humans. C57BL/6, BALB, SWR/J, A/J, 129, C3H, and all but one collaborative cross (CC) mouse strain following Cl 13 infection have immunosuppressed T cell responses, high PD-1, and viral titers leading to persistent infection and normal life spans. In contrast, the profile of FVB/N, NZB, PL/J, SL/J, and CC NZO mice challenged with Cl 13 is a robust T cell response, high titers of virus, PD-1, and Lag3 markers on T cells. These mice all die 7 to 9 d after Cl 13 infection. Death is due to enhanced pulmonary endothelial vascular permeability, pulmonary edema, collapse of alveolar air spaces, and respiratory failure. Pathogenesis involves abundant levels of Cl 13 receptor alpha-dystroglycan on endothelial cells, with high viral replication in such cells leading to immunopathologic injury. Death is aborted by blockade of interferon-1 (IFN-1) signaling or deletion of CD8 T cells.


Assuntos
Linfócitos T CD8-Positivos , Interferon Tipo I , Coriomeningite Linfocítica , Vírus da Coriomeningite Linfocítica/fisiologia , Replicação Viral/genética , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/patologia , Humanos , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Coriomeningite Linfocítica/genética , Coriomeningite Linfocítica/metabolismo , Coriomeningite Linfocítica/patologia , Camundongos , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Proteína do Gene 3 de Ativação de Linfócitos
10.
Proc Natl Acad Sci U S A ; 115(32): E7578-E7586, 2018 08 07.
Artigo em Inglês | MEDLINE | ID: mdl-30038008

RESUMO

The recent Ebola epidemic exemplified the importance of understanding and controlling emerging infections. Despite the importance of T cells in clearing virus during acute infection, little is known about Ebola-specific CD8+ T cell responses. We investigated immune responses of individuals infected with Ebola virus (EBOV) during the 2013-2016 West Africa epidemic in Sierra Leone, where the majority of the >28,000 EBOV disease (EVD) cases occurred. We examined T cell memory responses to seven of the eight Ebola proteins (GP, sGP, NP, VP24, VP30, VP35, and VP40) and associated HLA expression in survivors. Of the 30 subjects included in our analysis, CD8+ T cells from 26 survivors responded to at least one EBOV antigen. A minority, 10 of 26 responders (38%), made CD8+ T cell responses to the viral GP or sGP. In contrast, 25 of the 26 responders (96%) made response to viral NP, 77% to VP24 (20 of 26), 69% to VP40 (18 of 26), 42% (11 of 26) to VP35, with no response to VP30. Individuals making CD8+ T cells to EBOV VP24, VP35, and VP40 also made CD8+ T cells to NP, but rarely to GP. We identified 34 CD8+ T cell epitopes for Ebola. Our data indicate the immunodominance of the EBOV NP-specific T cell response and suggest that its inclusion in a vaccine along with the EBOV GP would best mimic survivor responses and help boost cell-mediated immunity during vaccination.


Assuntos
Anticorpos Antivirais/imunologia , Linfócitos T CD8-Positivos/imunologia , Ebolavirus/imunologia , Epidemias , Antígenos HLA/imunologia , Doença pelo Vírus Ebola/imunologia , Adolescente , Adulto , Anticorpos Antivirais/sangue , Antígenos Virais/imunologia , Epitopos de Linfócito T/imunologia , Feminino , Antígenos HLA/sangue , Doença pelo Vírus Ebola/sangue , Doença pelo Vírus Ebola/epidemiologia , Doença pelo Vírus Ebola/prevenção & controle , Humanos , Masculino , Nucleoproteínas/imunologia , Serra Leoa , Sobreviventes , Vacinação/métodos , Proteínas Virais/imunologia , Adulto Jovem
11.
J Infect Dis ; 222(9): 1488-1497, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32436943

RESUMO

BACKGROUND: Ebola virus (EBOV) disease has killed thousands of West and Central Africans over the past several decades. Many who survive the acute disease later experience post-Ebola syndrome, a constellation of symptoms whose causative pathogenesis is unclear. METHODS: We investigated EBOV-specific CD8+ and CD4+ T-cell responses in 37 Sierra Leonean EBOV disease survivors with (n = 19) or without (n = 18) sequelae of arthralgia and ocular symptoms. Peripheral blood mononuclear cells were infected with recombinant vesicular stomatitis virus encoding EBOV antigens. We also studied the presence of EBOV-specific immunoglobulin G, antinuclear antibodies, anti-cyclic citrullinated peptide antibodies, rheumatoid factor, complement levels, and cytokine levels in these 2 groups. RESULTS: Survivors with sequelae had a significantly higher EBOV-specific CD8+ and CD4+ T-cell response. No differences in EBOV-specific immunoglobulin G, antinuclear antibody, or anti-cyclic citrullinated peptide antibody levels were found. Survivors with sequelae showed significantly higher rheumatoid factor levels. CONCLUSION: EBOV-specific CD8+ and CD4+ T-cell responses were significantly higher in Ebola survivors with post-Ebola syndrome. These findings suggest that pathogenesis may occur as an immune-mediated disease via virus-specific T-cell immune response or that persistent antigen exposure leads to increased and sustained T-cell responses.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Ebolavirus/imunologia , Doença pelo Vírus Ebola/imunologia , Adulto , Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Feminino , Imunofluorescência , Doença pelo Vírus Ebola/patologia , Humanos , Imunidade Celular , Masculino , Serra Leoa/epidemiologia , Sobreviventes
12.
Proc Natl Acad Sci U S A ; 114(14): 3708-3713, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28325871

RESUMO

Blockade of IFN-α but not IFN-ß signaling using either an antibody or a selective S1PR1 agonist, CYM-5442, prevented type 1 diabetes (T1D) in the mouse Rip-LCMV T1D model. First, treatment with antibody or CYM-5442 limited the migration of autoimmune "antiself" T cells to the external boundaries around the islets and prevented their entry into the islets so they could not be positioned to engage, kill, and thus remove insulin-producing ß cells. Second, CYM-5442 induced an exhaustion signature in antiself T cells by up-regulating the negative immune regulator receptor genes Pdcd1, Lag3, Ctla4, Tigit, and Btla, thereby limiting their killing ability. By such means, insulin production was preserved and glucose regulation maintained, and a mechanism for S1PR1 immunomodulation described.


Assuntos
Diabetes Mellitus Tipo 1/prevenção & controle , Indanos/administração & dosagem , Interferon-alfa/metabolismo , Oxidiazóis/administração & dosagem , Estado Pré-Diabético/tratamento farmacológico , Receptores de Lisoesfingolipídeo/agonistas , Linfócitos T/efeitos dos fármacos , Animais , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Progressão da Doença , Indanos/farmacologia , Insulina/metabolismo , Células Secretoras de Insulina/imunologia , Ilhotas Pancreáticas/imunologia , Camundongos , Oxidiazóis/farmacologia , Estado Pré-Diabético/imunologia , Receptores Imunológicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptores de Esfingosina-1-Fosfato , Linfócitos T/imunologia
13.
J Infect Dis ; 219(8): 1338-1346, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30445431

RESUMO

INTRODUCTION: Live attenuated zoster vaccine (Zostavax) was used to test the hypothesis that constitutive level of interleukin 10 (IL-10), which may be high in elderly subjects, impairs vaccine efficacy. If constitutive IL-10 impairs vaccine efficacy, the effectiveness of viral vaccines might be improved by transient inhibition of IL-10 before vaccination. METHODS: Zostavax was given to 26 patients (age, 60-80 years). IL-10 and immunity to varicella zoster virus (VZV) were measured at baseline and after vaccination. Fluorescent antibody to membrane antigen (FAMA) assays and glycoprotein enzyme-linked immunosorbent assays (gpELISAs) were used to assess humoral immunity; anti-varicella virus T-cell responses were studied in a subset of subjects. In a prospective animal model, T-cell responses to chimeric vaccines against lymphocytic choriomeningitis virus (LCMV) were assessed in mice that express or lack IL-10. RESULTS: FAMA assays revealed significant boosting (by 4-fold) of humoral immunity, which occurred only in subjects (10 of 26) with a low constitutive IL-10 level (ie, <20 pg/mL); moreover, the Zostavax-induced FAMA and gpELISA responses were inversely related to the constitutive IL-10 level. Significant VZV-specific T-cell responses followed vaccination only in subjects with a low constitutive IL-10 level. Vaccine-induced LCMV-specific T-cell responses in mice lacking IL-10 were greater than in wild-type animals. CONCLUSIONS: A high constitutive IL-10 level adversely affects vaccine efficacy.


Assuntos
Vacina contra Herpes Zoster/imunologia , Herpesvirus Humano 3/imunologia , Interleucina-10/sangue , Idoso , Idoso de 80 Anos ou mais , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Imunofluorescência , Humanos , Imunidade Humoral/imunologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade
14.
PLoS Pathog ; 13(8): e1006588, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28837667

RESUMO

The multifunctional NS1 protein of influenza A viruses suppresses host cellular defense mechanisms and subverts other cellular functions. We report here on a new role for NS1 in modifying cell-cell signaling via the Hedgehog (Hh) pathway. Genetic epistasis experiments and FRET-FLIM assays in Drosophila suggest that NS1 interacts directly with the transcriptional mediator, Ci/Gli1. We further confirmed that Hh target genes are activated cell-autonomously in transfected human lung epithelial cells expressing NS1, and in infected mouse lungs. We identified a point mutation in NS1, A122V, that modulates this activity in a context-dependent fashion. When the A122V mutation was incorporated into a mouse-adapted influenza A virus, it cell-autonomously enhanced expression of some Hh targets in the mouse lung, including IL6, and hastened lethality. These results indicate that, in addition to its multiple intracellular functions, NS1 also modifies a highly conserved signaling pathway, at least in part via cell autonomous activities. We discuss how this new Hh modulating function of NS1 may influence host lethality, possibly through controlling cytokine production, and how these new insights provide potential strategies for combating infection.


Assuntos
Proteínas Hedgehog/metabolismo , Infecções por Orthomyxoviridae/metabolismo , Transdução de Sinais/fisiologia , Proteínas não Estruturais Virais/metabolismo , Animais , Drosophila , Humanos , Imuno-Histoquímica , Virus da Influenza A Subtipo H5N1/metabolismo , Camundongos , Camundongos Endogâmicos C57BL
15.
Proc Natl Acad Sci U S A ; 113(5): 1351-6, 2016 Feb 02.
Artigo em Inglês | MEDLINE | ID: mdl-26787880

RESUMO

Blunting immunopathology without abolishing host defense is the foundation for safe and effective modulation of infectious and autoimmune diseases. Sphingosine 1-phosphate receptor 1 (S1PR1) agonists are effective in treating infectious and multiple autoimmune pathologies; however, mechanisms underlying their clinical efficacy are yet to be fully elucidated. Here, we uncover an unexpected mechanism of convergence between S1PR1 and interferon alpha receptor 1 (IFNAR1) signaling pathways. Activation of S1PR1 signaling by pharmacological tools or endogenous ligand sphingosine-1 phosphate (S1P) inhibits type 1 IFN responses that exacerbate numerous pathogenic conditions. Mechanistically, S1PR1 selectively suppresses the type I IFN autoamplification loop in plasmacytoid dendritic cells (pDCs), a specialized DC subset, for robust type I IFN release. S1PR1 agonist suppression is pertussis toxin-resistant, but inhibited by an S1PR1 C-terminal-derived transactivating transcriptional activator (Tat)-fusion peptide that blocks receptor internalization. S1PR1 agonist treatment accelerates turnover of IFNAR1, suppresses signal transducer and activator of transcription 1 (STAT1) phosphorylation, and down-modulates total STAT1 levels, thereby inactivating the autoamplification loop. Inhibition of S1P-S1PR1 signaling in vivo using the selective antagonist Ex26 significantly elevates IFN-α production in response to CpG-A. Thus, multiple lines of evidence demonstrate that S1PR1 signaling sets the sensitivity of pDC amplification of IFN responses, thereby blunting pathogenic immune responses. These data illustrate a lipid G-protein coupled receptor (GPCR)-IFNAR1 regulatory loop that balances effective and detrimental immune responses and elevated endogenous S1PR1 signaling. This mechanism will likely be advantageous in individuals subject to a range of inflammatory conditions.


Assuntos
Células Dendríticas/metabolismo , Interferon-alfa/metabolismo , Receptor de Interferon alfa e beta/metabolismo , Receptores de Lisoesfingolipídeo/fisiologia , Animais , Camundongos , Camundongos Knockout , Proteólise , Receptor de Interferon alfa e beta/genética
16.
Proc Natl Acad Sci U S A ; 112(42): E5706-14, 2015 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-26438836

RESUMO

Endoplasmic reticulum (ER)-resident proteins are continually retrieved from the Golgi and returned to the ER by Lys-Asp-Glu-Leu (KDEL) receptors, which bind to an eponymous tetrapeptide motif at their substrate's C terminus. Mice and humans possess three paralogous KDEL receptors, but little is known about their functional redundancy, or if their mutation can be physiologically tolerated. Here, we present a recessive mouse missense allele of the prototypical mammalian KDEL receptor, KDEL ER protein retention receptor 1 (KDELR1). Kdelr1 homozygous mutants were mildly lymphopenic, as were mice with a CRISPR/Cas9-engineered frameshift allele. Lymphopenia was cell intrinsic and, in the case of T cells, was associated with reduced expression of the T-cell receptor (TCR) and increased expression of CD44, and could be partially corrected by an MHC class I-restricted TCR transgene. Antiviral immunity was also compromised, with Kdelr1 mutant mice unable to clear an otherwise self-limiting viral infection. These data reveal a nonredundant cellular function for KDELR1, upon which lymphocytes distinctly depend.


Assuntos
Retículo Endoplasmático/metabolismo , Predisposição Genética para Doença , Linfopenia/genética , Mutação , Receptores de Peptídeos/genética , Viroses/prevenção & controle , Animais , Doença Crônica , Feminino , Masculino , Camundongos , Camundongos Mutantes , Viroses/genética
17.
PLoS Pathog ; 11(1): e1004588, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25569216

RESUMO

Many persistent viral infections are characterized by a hypofunctional T cell response and the upregulation of negative immune regulators. These events occur days after the initiation of infection. However, the very early host-virus interactions that determine the establishment of viral persistence remain poorly uncharacterized. Here we show that to establish persistence, LCMV must counteract an innate anti-viral immune response within eight hours after infection. While the virus triggers cytoplasmic RNA sensing pathways soon after infection, LCMV counteracts this pathway through a rapid increase in viral titers leading to a dysfunctional immune response characterized by a high cytokine and chemokine expression profile. This altered immune environment allows for viral replication in the splenic white pulp as well as infection of immune cells essential to an effective anti-viral immune response. Our findings illustrate how early events during infection critically dictate the characteristics of the immune response to infection and facilitate either virus control and clearance or persistence.


Assuntos
Interações Hospedeiro-Patógeno/fisiologia , Coriomeningite Linfocítica/imunologia , Viroses , Animais , Células Cultivadas , Chlorocebus aethiops , Doença Crônica , Cricetinae , Tolerância Imunológica , Imunidade Inata , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Baço/imunologia , Fatores de Tempo , Células Vero , Viroses/imunologia , Viroses/virologia , Latência Viral/imunologia , Replicação Viral/imunologia
18.
Proc Natl Acad Sci U S A ; 111(10): 3799-804, 2014 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-24572573

RESUMO

During pathogenic influenza virus infection, robust cytokine production (cytokine storm), excessive inflammatory infiltrates, and virus-induced tissue destruction all contribute to morbidity and mortality. Earlier we reported that modulation of sphingosine-1-phosphate-1 receptor (S1P1R) signaling provided a chemically tractable approach for the effective blunting of cytokine storm, leading to the improvement of clinical and survival outcomes. Here, we show that S1P1R agonist treatment suppresses global cytokine amplification. Importantly, S1P1R agonist treatment was able to blunt cytokine/chemokine production and innate immune cell recruitment in the lung independently of endosomal and cytosolic innate sensing pathways. S1P1R signaling suppression of cytokine amplification was independent of multiple innate signaling adaptor pathways for myeloid differentiation primary response gene 88 (MyD88) and IFN-ß promoter stimulator-1 signaling, indicating a common pathway inhibition of cytokine storm. We identify the MyD88 adaptor molecule as responsible for the majority of cytokine amplification observed following influenza virus challenge.


Assuntos
Citocinas/imunologia , Imunidade Inata/imunologia , Infecções por Orthomyxoviridae/imunologia , Transdução de Sinais/imunologia , Animais , Transplante de Medula Óssea , Cães , Citometria de Fluxo , Células Madin Darby de Rim Canino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide , Infecções por Orthomyxoviridae/fisiopatologia , Receptores de Lisoesfingolipídeo/metabolismo
19.
Proc Natl Acad Sci U S A ; 111(24): 8925-30, 2014 Jun 17.
Artigo em Inglês | MEDLINE | ID: mdl-24889626

RESUMO

The outcome of a viral infection reflects the balance between virus virulence and host susceptibility. The clone 13 (Cl13) variant of lymphocytic choriomeningitis virus--a prototype of Old World arenaviruses closely related to Lassa fever virus--elicits in C57BL/6 and BALB/c mice abundant negative immunoregulatory molecules, associated with T-cell exhaustion, negligible T-cell-mediated injury, and high virus titers that persist. Conversely, here we report that in NZB mice, despite the efficient induction of immunoregulatory molecules and high viremia, Cl13 generated a robust cytotoxic T-cell response, resulting in thrombocytopenia, pulmonary endothelial cell loss, vascular leakage, and death within 6-8 d. These pathogenic events required type I IFN (IFN-I) signaling on nonhematopoietic cells and were completely abrogated by IFN-I receptor blockade. Thus, IFN-I may play a prominent role in hemorrhagic fevers and other acute virus infections associated with severe vascular pathology, and targeting IFN-I or downstream effector molecules may be an effective therapeutic approach.


Assuntos
Interferon Tipo I/metabolismo , Febre Lassa/virologia , Doenças Vasculares/virologia , Animais , Lavagem Broncoalveolar , Linhagem Celular , Cricetinae , Citocinas/metabolismo , Feminino , Vírus Lassa , Coriomeningite Linfocítica/virologia , Vírus da Coriomeningite Linfocítica/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NZB , Camundongos Transgênicos , Transdução de Sinais , Células-Tronco/química , Linfócitos T Citotóxicos/virologia , Ativação Viral
20.
Proc Natl Acad Sci U S A ; 110(11): 4180-3, 2013 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-23341590

RESUMO

Viruses have unique lifestyles. To describe the pathogenesis and significance of viral infection in terms of host responses, resultant injury, and therapy, we focused on two RNA viruses: lymphocytic choriomeningitis (LCMV) and influenza (Flu). Many of the currently established concepts and consequences about viruses and immunologic tolerance, virus-induced immunosuppression, virus-induced autoimmunity, immune complex disease, and virus-lymphocyte and virus-dendritic cell interactions evolved through studies of LCMV in its natural murine host. Similarly, the mechanisms, aftermath, and treatment of persistent RNA viruses emerged, in large part, from research on LCMV. Analysis of acute influenza virus infections uncovered the prominent direct role that cytokine storm plays in the pathogenesis, morbidity, and mortality from this disease. Cytokine storm of influenza virus infection is initiated via a pulmonary endothelial cell amplification loop involving IFN-producing cells and virus-infected pulmonary epithelial cells. Importantly, the cytokine storm is chemically treatable with specific agonist therapy directed to the sphingosphine 1 phosphate receptor 1, which is located on pulmonary endothelial cells, pointing to the endothelial cells as the gatekeepers of this hyperaggressive host immune response.


Assuntos
Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/fisiologia , Vírus da Coriomeningite Linfocítica/patogenicidade , Infecções por Orthomyxoviridae/imunologia , Orthomyxoviridae/fisiologia , Orthomyxoviridae/patogenicidade , Animais , Autoimunidade , Tolerância Imunológica , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA