Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Nature ; 533(7601): 100-4, 2016 May 05.
Artigo em Inglês | MEDLINE | ID: mdl-27147028

RESUMO

Despite the magnitude of the Ebola virus disease (EVD) outbreak in West Africa, there is still a fundamental lack of knowledge about the pathophysiology of EVD. In particular, very little is known about human immune responses to Ebola virus. Here we evaluate the physiology of the human T cell immune response in EVD patients at the time of admission to the Ebola Treatment Center in Guinea, and longitudinally until discharge or death. Through the use of multiparametric flow cytometry established by the European Mobile Laboratory in the field, we identify an immune signature that is unique in EVD fatalities. Fatal EVD was characterized by a high percentage of CD4(+) and CD8(+) T cells expressing the inhibitory molecules CTLA-4 and PD-1, which correlated with elevated inflammatory markers and high virus load. Conversely, surviving individuals showed significantly lower expression of CTLA-4 and PD-1 as well as lower inflammation, despite comparable overall T cell activation. Concomitant with virus clearance, survivors mounted a robust Ebola-virus-specific T cell response. Our findings suggest that dysregulation of the T cell response is a key component of EVD pathophysiology.


Assuntos
Ebolavirus/imunologia , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/fisiopatologia , Linfócitos T/imunologia , Antígeno CTLA-4/metabolismo , Feminino , Citometria de Fluxo , Guiné/epidemiologia , Doença pelo Vírus Ebola/mortalidade , Humanos , Mediadores da Inflamação/imunologia , Estudos Longitudinais , Ativação Linfocitária , Masculino , Alta do Paciente , Receptor de Morte Celular Programada 1/metabolismo , Sobreviventes , Linfócitos T/metabolismo , Carga Viral
2.
J Virol ; 92(17)2018 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-29950419

RESUMO

The flexible regulation of cellular metabolic pathways enables cellular adaptation to changes in energy demand under conditions of stress such as posed by a virus infection. To analyze such an impact on cellular metabolism, rubella virus (RV) was used in this study. RV replication under selected substrate supplementation with glucose, pyruvate, and glutamine as essential nutrients for mammalian cells revealed its requirement for glutamine. The assessment of the mitochondrial respiratory (based on the oxygen consumption rate) and glycolytic (based on the extracellular acidification rate) rate and capacity by respective stress tests through Seahorse technology enabled determination of the bioenergetic phenotype of RV-infected cells. Irrespective of the cellular metabolic background, RV infection induced a shift of the bioenergetic state of epithelial cells (Vero and A549) and human umbilical vein endothelial cells to a higher oxidative and glycolytic level. Interestingly there was a RV strain-specific, but genotype-independent demand for glutamine to induce a significant increase in metabolic activity. While glutaminolysis appeared to be rather negligible for RV replication, glutamine could serve as donor of its amide nitrogen in biosynthesis pathways for important metabolites. This study suggests that the capacity of RVs to induce metabolic alterations could evolve differently during natural infection. Thus, changes in cellular bioenergetics represent an important component of virus-host interactions and could complement our understanding of the viral preference for a distinct host cell population.IMPORTANCE RV pathologies, especially during embryonal development, could be connected with its impact on mitochondrial metabolism. With bioenergetic phenotyping we pursued a rather novel approach in virology. For the first time it was shown that a virus infection could shift the bioenergetics of its infected host cell to a higher energetic state. Notably, the capacity to induce such alterations varied among different RV isolates. Thus, our data add viral adaptation of cellular metabolic activity to its specific needs as a novel aspect to virus-host evolution. In addition, this study emphasizes the implementation of different viral strains in the study of virus-host interactions and the use of bioenergetic phenotyping of infected cells as a biomarker for virus-induced pathological alterations.


Assuntos
Metabolismo Energético , Glutamina/metabolismo , Glicólise/efeitos dos fármacos , Consumo de Oxigênio/fisiologia , Vírus da Rubéola/metabolismo , Células A549 , Células Endoteliais/metabolismo , Células Endoteliais/virologia , Glucose/metabolismo , Glucose/farmacologia , Glutamina/farmacologia , Homeostase , Humanos , Cinurenina/metabolismo , Redes e Vias Metabólicas/efeitos dos fármacos , Mitocôndrias/metabolismo , Nucleotídeos/biossíntese , Oxirredução , Estresse Oxidativo , Consumo de Oxigênio/efeitos dos fármacos , Fenótipo , Ácido Pirúvico/metabolismo , Ácido Pirúvico/farmacologia , Replicação Viral/efeitos dos fármacos
3.
Eur J Immunol ; 47(2): 345-352, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27859043

RESUMO

Influenza virus infection triggers an increase in the number of monocyte-derived dendritic cells (moDCs) in the respiratory tract, but the role of these cells during antiviral immunity is still unclear. Here we show that during influenza infection, moDCs dominate the late activation of CD8+ T cells and trigger the switch in immunodominance of the CD8+ T-cell response from acidic polymerase specificity to nucleoprotein specificity. Abrogation of monocyte recruitment or depletion of moDCs strongly compromised host resistance to secondary influenza challenge. These findings underscore a novel function of moDCs in the antiviral response to influenza virus, and have important implications for vaccine design.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Pulmão/imunologia , Monócitos/imunologia , Infecções por Orthomyxoviridae/imunologia , Especificidade do Receptor de Antígeno de Linfócitos T , Animais , Células Cultivadas , Células Dendríticas/virologia , Epitopos Imunodominantes/imunologia , Memória Imunológica , Pulmão/virologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Proteínas do Core Viral/imunologia
4.
PLoS Pathog ; 12(5): e1005656, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27191716

RESUMO

Lassa fever (LASF) is a highly severe viral syndrome endemic to West African countries. Despite the annual high morbidity and mortality caused by LASF, very little is known about the pathophysiology of the disease. Basic research on LASF has been precluded due to the lack of relevant small animal models that reproduce the human disease. Immunocompetent laboratory mice are resistant to infection with Lassa virus (LASV) and, to date, only immunodeficient mice, or mice expressing human HLA, have shown some degree of susceptibility to experimental infection. Here, transplantation of wild-type bone marrow cells into irradiated type I interferon receptor knockout mice (IFNAR-/-) was used to generate chimeric mice that reproduced important features of severe LASF in humans. This included high lethality, liver damage, vascular leakage and systemic virus dissemination. In addition, this model indicated that T cell-mediated immunopathology was an important component of LASF pathogenesis that was directly correlated with vascular leakage. Our strategy allows easy generation of a suitable small animal model to test new vaccines and antivirals and to dissect the basic components of LASF pathophysiology.


Assuntos
Modelos Animais de Doenças , Febre Lassa/imunologia , Febre Lassa/patologia , Animais , Citometria de Fluxo , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Quimera por Radiação
5.
J Infect Dis ; 213(6): 934-8, 2016 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-26531247

RESUMO

We studied the therapeutic potential of favipiravir (T-705) for Lassa fever, both alone and in combination with ribavirin. Favipiravir suppressed Lassa virus replication in cell culture by 5 log10 units. In a novel lethal mouse model, it lowered the viremia level and the virus load in organs and normalized levels of cell-damage markers. Treatment with 300 mg/kg per day, commenced 4 days after infection, when the viremia level had reached 4 log10 virus particles/mL, rescued 100% of Lassa virus-infected mice. We found a synergistic interaction between favipiravir and ribavirin in vitro and an increased survival rate and extended survival time when combining suboptimal doses in vivo.


Assuntos
Amidas/uso terapêutico , Antivirais/uso terapêutico , Febre Lassa/tratamento farmacológico , Pirazinas/uso terapêutico , Ribavirina/uso terapêutico , Amidas/administração & dosagem , Animais , Antivirais/administração & dosagem , Chlorocebus aethiops , Quimioterapia Combinada , Camundongos , Pirazinas/administração & dosagem , Ribavirina/administração & dosagem , Células Vero , Carga Viral , Replicação Viral
6.
J Infect Dis ; 214(suppl 3): S275-S280, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27521367

RESUMO

A number of previous studies have identified antigen-presenting cells (APCs) as key targets of Ebola virus (EBOV), but the role of APCs in human Ebola virus disease (EVD) is not known. We have evaluated the phenotype and kinetics of monocytes, neutrophils, and dendritic cells (DCs) in peripheral blood of patients for whom EVD was diagnosed by the European Mobile Laboratory in Guinea. Acute EVD was characterized by reduced levels of circulating nonclassical CD16+ monocytes with a poor activation profile. In survivors, CD16+ monocytes were activated during recovery, coincident with viral clearance, suggesting an important role of this cell subset in EVD pathophysiology.


Assuntos
Células Dendríticas/imunologia , Ebolavirus/imunologia , Doença pelo Vírus Ebola/imunologia , Monócitos/imunologia , Neutrófilos/imunologia , Receptores de IgG/imunologia , Células Dendríticas/virologia , Ebolavirus/isolamento & purificação , Feminino , Doença pelo Vírus Ebola/diagnóstico , Doença pelo Vírus Ebola/fisiopatologia , Doença pelo Vírus Ebola/virologia , Humanos , Cinética , Unidades Móveis de Saúde , Monócitos/virologia , Neutrófilos/virologia , Fenótipo
7.
J Infect Dis ; 214(suppl 3): S250-S257, 2016 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-27638946

RESUMO

BACKGROUND: A unit of the European Mobile Laboratory (EMLab) consortium was deployed to the Ebola virus disease (EVD) treatment unit in Guéckédou, Guinea, from March 2014 through March 2015. METHODS: The unit diagnosed EVD and malaria, using the RealStar Filovirus Screen reverse transcription-polymerase chain reaction (RT-PCR) kit and a malaria rapid diagnostic test, respectively. RESULTS: The cleaned EMLab database comprised 4719 samples from 2741 cases of suspected EVD from Guinea. EVD was diagnosed in 1231 of 2178 hospitalized patients (57%) and in 281 of 563 who died in the community (50%). Children aged <15 years had the highest proportion of Ebola virus-malaria parasite coinfections. The case-fatality ratio was high in patients aged <5 years (80%) and those aged >74 years (90%) and low in patients aged 10-19 years (40%). On admission, RT-PCR analysis of blood specimens from patients who died in the hospital yielded a lower median cycle threshold (Ct) than analysis of blood specimens from survivors (18.1 vs 23.2). Individuals who died in the community had a median Ct of 21.5 for throat swabs. Multivariate logistic regression on 1047 data sets revealed that low Ct values, ages of <5 and ≥45 years, and, among children aged 5-14 years, malaria parasite coinfection were independent determinants of a poor EVD outcome. CONCLUSIONS: Virus load, age, and malaria parasite coinfection play a role in the outcome of EVD.


Assuntos
Ebolavirus/isolamento & purificação , Epidemias , Infecções por Filoviridae/diagnóstico , Doença pelo Vírus Ebola/diagnóstico , Malária/complicações , Unidades Móveis de Saúde , Adolescente , Adulto , Idoso , Criança , Pré-Escolar , Serviços de Laboratório Clínico , Ebolavirus/genética , Feminino , Filoviridae , Infecções por Filoviridae/complicações , Infecções por Filoviridae/virologia , Guiné , Doença pelo Vírus Ebola/complicações , Doença pelo Vírus Ebola/virologia , Humanos , Lactente , Malária/parasitologia , Masculino , Pessoa de Meia-Idade , RNA Viral/sangue , Carga Viral , Adulto Jovem
8.
J Virol ; 89(8): 4700-4, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25673711

RESUMO

The development of treatments for Ebola virus disease (EVD) has been hampered by the lack of small-animal models that mimick human disease. Here we show that mice with transplanted human hematopoetic stem cells reproduce features typical of EVD. Infection with Ebola virus was associated with viremia, cell damage, liver steatosis, signs of hemorrhage, and high lethality. Our study provides a small-animal model with human components for the development of EVD therapies.


Assuntos
Modelos Animais de Doenças , Ebolavirus/imunologia , Transplante de Células-Tronco Hematopoéticas/métodos , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/transmissão , Xenoenxertos/imunologia , Camundongos Endogâmicos NOD , Animais , Fígado Gorduroso/patologia , Hemorragia/patologia , Doença pelo Vírus Ebola/patologia , Humanos , Estimativa de Kaplan-Meier , Camundongos , Microscopia de Fluorescência , Viremia/patologia
9.
J Immunol ; 193(3): 1324-32, 2014 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-24958904

RESUMO

Live-attenuated influenza vaccines (LAIVs) have the potential to generate CD8 T cell immunity that may limit the virulence of an antigenically shifted influenza strain in a population lacking protective Abs. However, current LAIVs exert limited T cell immunity restricted to the vaccine strains. One approach to improve LAIV-induced T cell responses is the use of specific adjuvants to enhance T cell priming by respiratory dendritic cells, but this hypothesis has not been addressed. In this study, we assessed the effect of the TLR3 ligand polyinosinic-polycytidylic acid (poly IC) on CD8 T cell immunity and protection elicited by LAIVs. Mucosal treatment with poly IC shortly after vaccination enhanced respiratory dendritic cell function, CD8 T cell formation, and production of neutralizing Abs. This adjuvant effect of poly IC was dependent on amplification of TLR3 signaling by nonhematopoietic radioresistant cells and enhanced mouse protection to homosubtypic, as well as heterosubtypic, virus challenge. Our findings indicate that mucosal TLR3 ligation may be used to improve CD8 T cell responses to replicating vaccines, which has implications for protection in the absence of pre-existing Ab immunity.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Mucosa Nasal/imunologia , Poli I-C/administração & dosagem , Poli I-C/uso terapêutico , Replicação Viral/imunologia , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/uso terapêutico , Animais , Linfócitos T CD8-Positivos/efeitos dos fármacos , Linfócitos T CD8-Positivos/virologia , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/virologia , Células HEK293 , Humanos , Imunidade Celular/efeitos dos fármacos , Vírus da Influenza A Subtipo H1N1/imunologia , Vacinas contra Influenza/uso terapêutico , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Poli I-C/imunologia , Regulação para Cima/imunologia , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Vacinas Atenuadas/uso terapêutico , Replicação Viral/efeitos dos fármacos
10.
Sci Immunol ; 8(84): eadd7446, 2023 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-37294749

RESUMO

The recruitment of monocytes and their differentiation into immunosuppressive cells is associated with the low efficacy of preclinical nonconformal radiotherapy (RT) for tumors. However, nonconformal RT (non-CRT) does not mimic clinical practice, and little is known about the role of monocytes after RT modes used in patients, such as conformal RT (CRT). Here, we investigated the acute immune response induced by after CRT. Contrary to non-CRT approaches, we found that CRT induces a rapid and robust recruitment of monocytes to the tumor that minimally differentiate into tumor-associated macrophages or dendritic cells but instead up-regulate major histocompatibility complex II and costimulatory molecules. We found that these large numbers of infiltrating monocytes are responsible for activating effector polyfunctional CD8+ tumor-infiltrating lymphocytes that reduce tumor burden. Mechanistically, we show that monocyte-derived type I interferon is pivotal in promoting monocyte accumulation and immunostimulatory function in a positive feedback loop. We also demonstrate that monocyte accumulation in the tumor microenvironment is hindered when RT inadvertently affects healthy tissues, as occurs in non-CRT. Our results unravel the immunostimulatory function of monocytes during clinically relevant modes of RT and demonstrate that limiting the exposure of healthy tissues to radiation has a positive therapeutic effect on the overall antitumor immune response.


Assuntos
Interferon Tipo I , Neoplasias , Humanos , Monócitos , Neoplasias/radioterapia , Diferenciação Celular , Interferon Tipo I/farmacologia , Linfócitos do Interstício Tumoral , Microambiente Tumoral
11.
Cell Rep ; 29(11): 3736-3750.e8, 2019 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-31825848

RESUMO

Plasmacytoid dendritic cells (pDCs) are sensor cells with diverse immune functions, from type I interferon (IFN-I) production to antigen presentation, T cell activation, and tolerance. Regulation of these functions remains poorly understood but could be mediated by functionally specialized pDC subpopulations. We address pDC diversity using a high-dimensional single-cell approach: mass cytometry (CyTOF). Our analysis uncovers a murine pDC-like population that specializes in antigen presentation with limited capacity for IFN-I production. Using a multifaceted cross-species comparison, we show that this pDC-like population is the definitive murine equivalent of the recently described human AXL+ DCs, which we unify under the name transitional DCs (tDCs) given their continuum of pDC and cDC2 characteristics. tDCs share developmental traits with pDCs, as well as recruitment dynamics during viral infection. Altogether, we provide a framework for deciphering the function of pDCs and tDCs during diseases, which has the potential to open new avenues for therapeutic design.


Assuntos
Apresentação de Antígeno , Células Dendríticas/imunologia , Interferon gama/metabolismo , Adulto , Animais , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/virologia , Feminino , Citometria de Fluxo/métodos , Humanos , Interferon gama/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Orthomyxoviridae/imunologia , Orthomyxoviridae/patogenicidade , Análise de Célula Única/métodos , Especificidade da Espécie , Transcriptoma
12.
JCI Insight ; 4(21)2019 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-31550241

RESUMO

Filoviruses of the genus Ebolavirus include 6 species with marked differences in their ability to cause disease in humans. From the highly virulent Ebola virus to the seemingly nonpathogenic Reston virus, case fatality rates can range between 0% and 90%. In order to understand the molecular basis of these differences, it is imperative to establish disease models that recapitulate human disease as faithfully as possible. Nonhuman primates (NHPs) are the gold-standard models for filovirus pathogenesis, but comparative studies are skewed by the fact that Reston virus infection can be lethal for NHPs. Here we used HLA-A2-transgenic, NOD-scid-IL-2γ receptor-knockout (NSG-A2) mice reconstituted with human hematopoiesis to compare Ebola virus and Reston virus pathogenesis in a human-like environment. While markedly less pathogenic than Ebola virus, Reston virus killed 20% of infected mice, a finding that was linked to exacerbated inflammation and viral replication in the liver. In addition, the case fatality ratios of different Ebolavirus species in humans were recapitulated in the humanized mice. Our findings point to humanized mice as a putative model to test the pathogenicity of newly discovered filoviruses, and suggest that further investigations on Reston virus pathogenesis in humans are warranted.


Assuntos
Doença pelo Vírus Ebola/patologia , Animais , Modelos Animais de Doenças , Ebolavirus/patogenicidade , Ebolavirus/fisiologia , Doença pelo Vírus Ebola/virologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mucosa/virologia , Carga Viral , Replicação Viral
13.
Sci Rep ; 7: 43776, 2017 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-28256637

RESUMO

Ebola virus (EBOV) causes severe systemic disease in humans and non-human primates characterized by high levels of viremia and virus titers in peripheral organs. The natural portals of virus entry are the mucosal surfaces and the skin where macrophages and dendritic cells (DCs) are primary EBOV targets. Due to the migratory properties of DCs, EBOV infection of these cells has been proposed as a necessary step for virus dissemination via draining lymph nodes and blood. Here we utilize chimeric mice with competent hematopoietic-driven immunity, to show that EBOV primarily infects CD11b+ DCs in non-lymphoid and lymphoid tissues, but spares the main cross-presenting CD103+ DC subset. Furthermore, depletion of CD8 and CD4 T cells resulted in loss of early control of virus replication, viremia and fatal Ebola virus disease (EVD). Thus, our findings point out at T cell function as a key determinant of EVD progress and outcome.


Assuntos
Ebolavirus/imunologia , Doença pelo Vírus Ebola/imunologia , Linfócitos T/imunologia , Replicação Viral/imunologia , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Antígeno CD11b/imunologia , Antígeno CD11b/metabolismo , Apresentação Cruzada/imunologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Dendríticas/virologia , Ebolavirus/fisiologia , Doença pelo Vírus Ebola/virologia , Interações Hospedeiro-Patógeno/imunologia , Cadeias alfa de Integrinas/imunologia , Cadeias alfa de Integrinas/metabolismo , Cinética , Tecido Linfoide/imunologia , Tecido Linfoide/metabolismo , Tecido Linfoide/virologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Viremia/imunologia , Viremia/virologia
14.
Genome Biol ; 18(1): 4, 2017 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-28100256

RESUMO

BACKGROUND: In 2014, Western Africa experienced an unanticipated explosion of Ebola virus infections. What distinguishes fatal from non-fatal outcomes remains largely unknown, yet is key to optimising personalised treatment strategies. We used transcriptome data for peripheral blood taken from infected and convalescent recovering patients to identify early stage host factors that are associated with acute illness and those that differentiate patient survival from fatality. RESULTS: The data demonstrate that individuals who succumbed to the disease show stronger upregulation of interferon signalling and acute phase responses compared to survivors during the acute phase of infection. Particularly notable is the strong upregulation of albumin and fibrinogen genes, which suggest significant liver pathology. Cell subtype prediction using messenger RNA expression patterns indicated that NK-cell populations increase in patients who survive infection. By selecting genes whose expression properties discriminated between fatal cases and survivors, we identify a small panel of responding genes that act as strong predictors of patient outcome, independent of viral load. CONCLUSIONS: Transcriptomic analysis of the host response to pathogen infection using blood samples taken during an outbreak situation can provide multiple levels of information on both disease state and mechanisms of pathogenesis. Host biomarkers were identified that provide high predictive value under conditions where other predictors, such as viral load, are poor prognostic indicators. The data suggested that rapid analysis of the host response to infection in an outbreak situation can provide valuable information to guide an understanding of disease outcome and mechanisms of disease.


Assuntos
Ebolavirus , Perfilação da Expressão Gênica , Doença pelo Vírus Ebola/genética , Doença pelo Vírus Ebola/virologia , Interações Hospedeiro-Patógeno/genética , Transcriptoma , Análise por Conglomerados , Coinfecção , Biologia Computacional/métodos , Resistência à Doença/genética , Resistência à Doença/imunologia , Guiné , Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Interferons/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Avaliação de Resultados da Assistência ao Paciente , Curva ROC , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Carga Viral
15.
PLoS Negl Trop Dis ; 11(5): e0005645, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28558022

RESUMO

BACKGROUND: Human Ebola infection is characterized by a paralysis of the immune system. A signature of αß T cells in fatal Ebola infection has been recently proposed, while the involvement of innate immune cells in the protection/pathogenesis of Ebola infection is unknown. Aim of this study was to analyze γδ T and NK cells in patients from the Ebola outbreak of 2014-2015 occurred in West Africa, and to assess their association with the clinical outcome. METHODOLOGY/PRINCIPAL FINDINGS: Nineteen Ebola-infected patients were enrolled at the time of admission to the Ebola Treatment Centre in Guinea. Patients were divided in two groups on the basis of the clinical outcome. The analysis was performed by using multiparametric flow cytometry established by the European Mobile Laboratory in the field. A low frequency of Vδ2 T-cells was observed during Ebola infection, independently from the clinical outcome. Moreover, Vδ2 T-cells from Ebola patients massively expressed CD95 apoptotic marker, suggesting the involvement of apoptotic mechanisms in Vδ2 T-cell loss. Interestingly, Vδ2 T-cells from survivors expressed an effector phenotype and presented a lower expression of the CTLA-4 exhaustion marker than fatalities, suggesting a role of effector Vδ2 T-cells in the protection. Furthermore, patients with fatal Ebola infection were characterized by a lower NK cell frequency than patients with non fatal infection. In particular, both CD56bright and CD56dim NK frequency were very low both in fatal and non fatal infections, while a higher frequency of CD56neg NK cells was associated to non-fatal infections. Finally, NK activation and expression of NKp46 and CD158a were independent from clinical outcome. CONCLUSIONS/SIGNIFICANCES: Altogether, the data suggest that both effector Vδ2 T-cells and NK cells may play a role in the complex network of protective response to EBOV infection. Further studies are required to characterize the protective effector functions of Vδ2 and NK cells.


Assuntos
Doença pelo Vírus Ebola/imunologia , Doença pelo Vírus Ebola/mortalidade , Células Matadoras Naturais/imunologia , Receptores de Antígenos de Linfócitos T gama-delta/imunologia , Subpopulações de Linfócitos T/imunologia , Biomarcadores/metabolismo , Antígeno CD56/metabolismo , Antígeno CTLA-4/metabolismo , Bases de Dados Factuais , Ebolavirus , Feminino , Citometria de Fluxo , Guiné/epidemiologia , Humanos , Ativação Linfocitária/imunologia , Masculino , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Receptores KIR2DL1/metabolismo , Carga Viral , Receptor fas/metabolismo
16.
J Vis Exp ; (100): e52803, 2015 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-26168339

RESUMO

Vaccines are one of the greatest achievements of mankind, and have saved millions of lives over the last century. Paradoxically, little is known about the physiological mechanisms that mediate immune responses to vaccines perhaps due to the overall success of vaccination, which has reduced interest into the molecular and physiological mechanisms of vaccine immunity. However, several important human pathogens including influenza virus still pose a challenge for vaccination, and may benefit from immune-based strategies. Although influenza reverse genetics has been successfully applied to the generation of live-attenuated influenza vaccines (LAIVs), the addition of molecular tools in vaccine preparations such as tracer components to follow up the kinetics of vaccination in vivo, has not been addressed. In addition, the recent generation of mouse models that allow specific depletion of leukocytes during kinetic studies has opened a window of opportunity to understand the basic immune mechanisms underlying vaccine-elicited protection. Here, we describe how the combination of reverse genetics and chimeric mouse models may help to provide new insights into how vaccines work at physiological and molecular levels, using as example a recombinant, cold-adapted, live-attenuated influenza vaccine (LAIV). We utilized laboratory-generated LAIVs harboring cell tracers as well as competitive bone marrow chimeras (BMCs) to determine the early kinetics of vaccine immunity and the main physiological mechanisms responsible for the initiation of vaccine-specific adaptive immunity. In addition, we show how this technique may facilitate gene function studies in single animals during immune responses to vaccines. We propose that this technique can be applied to improve current prophylactic strategies against pathogens for which urgent medical countermeasures are needed, for example influenza, HIV, Plasmodium, and hemorrhagic fever viruses such as Ebola virus.


Assuntos
Imunidade nas Mucosas/efeitos dos fármacos , Vacinas contra Influenza/administração & dosagem , Vacinas contra Influenza/imunologia , Animais , Quimera , Células Dendríticas/imunologia , Feminino , Células HEK293 , Humanos , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H2N2/imunologia , Vacinas contra Influenza/genética , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia
17.
Antiviral Res ; 105: 17-21, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24583123

RESUMO

Outbreaks of Ebola hemorrhagic fever in sub-Saharan Africa are associated with case fatality rates of up to 90%. Currently, neither a vaccine nor an effective antiviral treatment is available for use in humans. Here, we evaluated the efficacy of the pyrazinecarboxamide derivative T-705 (favipiravir) against Zaire Ebola virus (EBOV) in vitro and in vivo. T-705 suppressed replication of Zaire EBOV in cell culture by 4log units with an IC90 of 110µM. Mice lacking the type I interferon receptor (IFNAR(-)(/)(-)) were used as in vivo model for Zaire EBOV-induced disease. Initiation of T-705 administration at day 6 post infection induced rapid virus clearance, reduced biochemical parameters of disease severity, and prevented a lethal outcome in 100% of the animals. The findings suggest that T-705 is a candidate for treatment of Ebola hemorrhagic fever.


Assuntos
Amidas/uso terapêutico , Antivirais/uso terapêutico , Ebolavirus/isolamento & purificação , Doença pelo Vírus Ebola/tratamento farmacológico , Pirazinas/uso terapêutico , Animais , Modelos Animais de Doenças , Ebolavirus/efeitos dos fármacos , Ebolavirus/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Análise de Sobrevida , Carga Viral , Replicação Viral/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA