Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
1.
Nat Microbiol ; 9(3): 751-762, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38326571

RESUMO

Infection with Lassa virus (LASV) can cause Lassa fever, a haemorrhagic illness with an estimated fatality rate of 29.7%, but causes no or mild symptoms in many individuals. Here, to investigate whether human genetic variation underlies the heterogeneity of LASV infection, we carried out genome-wide association studies (GWAS) as well as seroprevalence surveys, human leukocyte antigen typing and high-throughput variant functional characterization assays. We analysed Lassa fever susceptibility and fatal outcomes in 533 cases of Lassa fever and 1,986 population controls recruited over a 7 year period in Nigeria and Sierra Leone. We detected genome-wide significant variant associations with Lassa fever fatal outcomes near GRM7 and LIF in the Nigerian cohort. We also show that a haplotype bearing signatures of positive selection and overlapping LARGE1, a required LASV entry factor, is associated with decreased risk of Lassa fever in the Nigerian cohort but not in the Sierra Leone cohort. Overall, we identified variants and genes that may impact the risk of severe Lassa fever, demonstrating how GWAS can provide insight into viral pathogenesis.


Assuntos
Febre Lassa , Humanos , Febre Lassa/genética , Febre Lassa/diagnóstico , Febre Lassa/epidemiologia , Estudo de Associação Genômica Ampla , Estudos Soroepidemiológicos , Vírus Lassa/genética , Febre , Genética Humana
2.
PLoS Negl Trop Dis ; 16(11): e0010882, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36441765

RESUMO

INTRODUCTION: Lassa virus is a priority pathogen for vaccine research and development, however the duration of cellular immunity and protection in Lassa fever (LF) survivors remains unclear. METHODS: We investigated Lassa virus specific CD8+ T cell responses in 93 LF survivors. Peripheral blood mononuclear cells from these individuals were infected with recombinant vesicular stomatitis virus encoding Lassa virus antigens and virus specific T cell responses were measured after 18-hour incubation. Participants who had undetectable CD8+ T cell response underwent further analysis using a 10-day T cell proliferation assays to evaluate for low T cell precursor frequency. RESULTS: Forty-five of the 93 LF survivors did not have a Lassa virus specific CD8+ T cell response. Of those with responses and a known date of onset of LF (N = 11), 9 had LF within the last ten years. Most participants without a measurable CD8+ T cell response were more than 10 years removed from a clinical history of LF (N = 14/16). Fourteen of 21 patients (67%) with undetectable CD8+ T cell response had a measurable Lassa virus specific CD8+ T cell response with the 10-day assay. DISCUSSION: Despite reports of strong CD8+ T cell responses during acute Lassa virus infection, circulating Lassa virus-specific CD8+ T cells declined to undetectable levels in most Lassa fever survivors after ten years when evaluated with an 18-hour T cell stimulation. However, when Lassa virus-specific T cells were expanded prior to restimulation, a Lassa virus-specific CD8+ T cell response could be detected in many if the samples that were negative in the 18-hour stimulation assay, suggesting that prolonged cellular immunity does exist in Lassa fever survivors at low frequencies.


Assuntos
Febre Lassa , Células Precursoras de Linfócitos T , Humanos , Vírus Lassa , Leucócitos Mononucleares , Imunidade , Linfócitos T CD8-Positivos
3.
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
4.
Viruses ; 13(6)2021 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-34205512

RESUMO

Mature male mice produce a particularly high concentration of major urinary proteins (MUPs) in their scent marks that provide identity and status information to conspecifics. Darcin (MUP20) is inherently attractive to females and, by inducing rapid associative learning, leads to specific attraction to the individual male's odour and location. Other polymorphic central MUPs, produced at much higher abundance, bind volatile ligands that are slowly released from a male's scent marks, forming the male's individual odour that females learn. Here, we show that infection of C57BL/6 males with LCMV WE variants (v2.2 or v54) alters MUP expression according to a male's infection status and ability to clear the virus. MUP output is substantially reduced during acute adult infection with LCMV WE v2.2 and when males are persistently infected with LCMV WE v2.2 or v54. Infection differentially alters expression of darcin and, particularly, suppresses expression of a male's central MUP signature. However, following clearance of acute v2.2 infection through a robust virus-specific CD8 cytotoxic T cell response that leads to immunity to the virus, males regain their normal mature male MUP pattern and exhibit enhanced MUP output by 30 days post-infection relative to uninfected controls. We discuss the likely impact of these changes in male MUP signals on female attraction and mate selection. As LCMV infection during pregnancy can substantially reduce embryo survival and lead to lifelong infection in surviving offspring, we speculate that females use LCMV-induced changes in MUP expression both to avoid direct infection from a male and to select mates able to develop immunity to local variants that will be inherited by their offspring.


Assuntos
Coriomeningite Linfocítica/complicações , Vírus da Coriomeningite Linfocítica/patogenicidade , Proteínas/metabolismo , Animais , Feminino , Peptídeos e Proteínas de Sinalização Intercelular/genética , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Odorantes , Feromônios/metabolismo , Proteínas/análise , Proteínas/genética
5.
Viruses ; 13(7)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34206262

RESUMO

During chronic viral infections, CD8 T cells rapidly lose antiviral and immune-stimulatory functions in a sustained program termed exhaustion. In addition to this loss of function, CD8 T cells with the highest affinity for viral antigen can be physically deleted. Consequently, treatments designed to restore function to exhausted cells and control chronic viral replication are limited from the onset by the decreased breadth of the antiviral T cell response. Yet, it remains unclear why certain populations of CD8 T cells are deleted while others are preserved in an exhausted state. We report that CD8 T cell deletion during chronic viral infection can be prevented by therapeutically lowering viral replication early after infection. The initial resistance to deletion enabled long-term maintenance of antiviral cytolytic activity of the otherwise deleted high-affinity CD8 T cells. In combination with decreased virus titers, CD4 T cell help and prolonged interactions with costimulatory molecules B7-1/B7-2 were required to prevent CD8 T cell deletion. Thus, therapeutic strategies to decrease early virus replication could enhance virus-specific CD8 T cell diversity and function during chronic infection.


Assuntos
Infecções por Arenaviridae/imunologia , Linfócitos T CD8-Positivos/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Infecção Persistente/imunologia , Imunidade Adaptativa , Animais , Antivirais/uso terapêutico , Infecções por Arenaviridae/tratamento farmacológico , Infecções por Arenaviridae/virologia , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Linfócitos T CD4-Positivos/imunologia , Vírus da Coriomeningite Linfocítica/efeitos dos fármacos , Vírus da Coriomeningite Linfocítica/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Nucleocapsídeo/imunologia , Infecção Persistente/tratamento farmacológico , Infecção Persistente/virologia , Ribavirina/uso terapêutico , Carga Viral , Replicação Viral/efeitos dos fármacos
6.
EMBO Mol Med ; 13(1): e13533, 2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33326687

RESUMO

The sphingosine-1-phosphate (S1P) is a lysophospholipid signaling molecule with important functions in many physiological and pathological conditions, including viral infection. In this issue of EMBO Molecular Medicine, Marfia et al present a risk stratification based on S1P serum level as a novel prognostic indicator for COVID-19 severity.


Assuntos
COVID-19 , Humanos , Lisofosfolipídeos , Medição de Risco , SARS-CoV-2 , Esfingosina/análogos & derivados
7.
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
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.
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
10.
Cell Rep ; 29(10): 3293-3302.e3, 2019 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-31801090

RESUMO

Inhibitory receptors (IRs) function as critical regulators of immune responses by tempering T cell activity. In humans, several persisting viruses as well as cancers exploit IR signaling by upregulating IR ligands, resulting in suppression of T cell function (i.e., exhaustion). This allows escape from immune surveillance and continuation of disease. Here, we report the design, implementation, and results of a phenotypic high-throughput screen for molecules that modulate CD8+ T cell activity. We identify 19 compounds from the ReFRAME drug-repurposing collection that restore cytokine production and enhance the proliferation of exhausted T cells. Analysis of our top hit, ingenol mebutate, a protein kinase C (PKC) inducing diterpene ester, reveals a role for this molecule in overriding the suppressive signaling cascade mediated by IR signaling on T cells. Collectively, these results demonstrate a disease-relevant methodology for identifying modulators of T cell function and reveal new targets for immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Proliferação de Células/efeitos dos fármacos , Chlorocebus aethiops , Receptores Coestimuladores e Inibidores de Linfócitos T/metabolismo , Citocinas/metabolismo , Imunoterapia/métodos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Quinase C/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Vero
11.
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
12.
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
13.
PLoS One ; 13(9): e0203118, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30199535

RESUMO

Viruses have long been implicated in the pathogenesis of autoimmunity, yet their contribution remains circumstantial partly due to the lack of well-documented information on infections prior to autoimmune disease onset. Here, we used the lymphocytic choriomeningitis virus (LCMV) as a model to mechanistically dissect the impact of viral infection on lupus-like autoimmunity. Virus persistence strongly enhanced disease in mice with otherwise weak genetic predisposition but not in highly predisposed or non-autoimmune mice, indicating a synergistic interplay between genetic susceptibility and virus infection. Moreover, endosomal Toll-like receptors (TLRs) and plasmacytoid dendritic cells (pDCs) were both strictly required for disease acceleration, even though LCMV also induces strong TLR-independent type I interferon (IFN-I) production via RNA helicases and MAVS in conventional DCs. These results suggest that LCMV enhances systemic autoimmunity primarily by providing stimulatory nucleic acids for endosomal TLR engagement, whereas overstimulation of the MAVS-dependent cytosolic pathway in the absence of endosomal TLR signaling is insufficient for disease induction.


Assuntos
Autoimunidade , Endossomos/imunologia , Lúpus Eritematoso Sistêmico/imunologia , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica , Receptores Toll-Like/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Autoimunidade/genética , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/virologia , Modelos Animais de Doenças , Endossomos/virologia , Feminino , Predisposição Genética para Doença , Interferon Tipo I/metabolismo , Lúpus Eritematoso Sistêmico/genética , Lúpus Eritematoso Sistêmico/virologia , Coriomeningite Linfocítica/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Transdução de Sinais
14.
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
15.
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
16.
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
18.
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
19.
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
20.
Annu Rev Pathol ; 11: 1-19, 2016 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-26514062

RESUMO

This odyssey is mine from early junior high school, where my dreams for adventure were shaped by Arthur Conan Doyle's Sherlock Holmes, Percival Christopher Wren's Beau Geste, and best of all the remarkable explorers in Paul de Kruif's Microbe Hunters. My birth site was in Manhattan (my mother was a Vogue model and my father worked in retail), and I traveled to college at the University of Alabama, Tuscaloosa, where my love of history and English literature was shaped along with a sufficient exposure to biology, chemistry, and genetics to meet requirements for entering medical school. By the second year at the University of Maryland School of Medicine, through expert teachers such as Theodore (Ted) Woodward and Sheldon (Shelly) Greisman in medicine and Charles Weissmann in virology and microbiology, I found that understanding why and how people became ill was more my cup of tea than identifying and treating their illnesses. Although I was becoming competent in diagnosis and treatment, I left medical school at the end of my sophomore year to seek a more basic understanding of biology and chemistry. I achieved this by working toward a PhD in biochemistry at Johns Hopkins McCollum-Pratt Institute combined with study of rickettsial toxin at Maryland. This was a very important time in my life, because it convinced me that addressing biologic and medical questions in a disciplined scientific manner was what my life voyage should be. That voyage led me initially, through Woodward's contact, to work a summer in Joe Smadel's unit at Walter Reed (Smadel being one of the deans of American virology) and to meet several times with Carleton Gajdusek and then John Enders at Harvard, who pointed me to Frank Dixon at Scripps in La Jolla, California, for postdoctoral training. Dixon was among the founders of modern immunology and a pathfinder for immunopathology. Training by and association with Dixon and his other postdoctoral fellows, my independent position at Scripps, early polishing by Karl Habel (a superb senior virologist who left the National Institutes of Health and came to Scripps), and the gifted postdoctoral fellows who joined my laboratory over four decades form the log of my scientific voyage. The strong friendships and collaborations developed with other young but growing experimentalists like Bernie Fields and Abner Notkins are the fabric of the tale I will weave and were pivotal in the establishment of viral pathogenesis as a discipline.


Assuntos
Viroses/história , Viroses/virologia , Fenômenos Fisiológicos Virais , Vírus/patogenicidade , Bioquímica , História do Século XX , História do Século XXI , Humanos , Faculdades de Medicina , Viroses/metabolismo , Vírus/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA