Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Malar J ; 23(1): 106, 2024 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-38632607

RESUMO

BACKGROUND: To gain a deeper understanding of protective immunity against relapsing malaria, this study examined sporozoite-specific T cell responses induced by a chemoprophylaxis with sporozoite (CPS) immunization in a relapsing Plasmodium cynomolgi rhesus macaque model. METHODS: The animals received three CPS immunizations with P. cynomolgi sporozoites, administered by mosquito bite, while under two anti-malarial drug regimens. Group 1 (n = 6) received artesunate/chloroquine (AS/CQ) followed by a radical cure with CQ plus primaquine (PQ). Group 2 (n = 6) received atovaquone-proguanil (AP) followed by PQ. After the final immunization, the animals were challenged with intravenous injection of 104 P. cynomolgi sporozoites, the dose that induced reliable infection and relapse rate. These animals, along with control animals (n = 6), were monitored for primary infection and subsequent relapses. Immunogenicity blood draws were done after each of the three CPS session, before and after the challenge, with liver, spleen and bone marrow sampling and analysis done after the challenge. RESULTS: Group 2 animals demonstrated superior protection, with two achieving protection and two experiencing partial protection, while only one animal in group 1 had partial protection. These animals displayed high sporozoite-specific IFN-γ T cell responses in the liver, spleen, and bone marrow after the challenge with one protected animal having the highest frequency of IFN-γ+ CD8+, IFN-γ+ CD4+, and IFN-γ+ γδ T cells in the liver. Partially protected animals also demonstrated a relatively high frequency of IFN-γ+ CD8+, IFN-γ+ CD4+, and IFN-γ+ γδ T cells in the liver. It is important to highlight that the second animal in group 2, which experienced protection, exhibited deficient sporozoite-specific T cell responses in the liver while displaying average to high T cell responses in the spleen and bone marrow. CONCLUSIONS: This research supports the notion that local liver T cell immunity plays a crucial role in defending against liver-stage infection. Nevertheless, there is an instance where protection occurs independently of T cell responses in the liver, suggesting the involvement of the liver's innate immunity. The relapsing P. cynomolgi rhesus macaque model holds promise for informing the development of vaccines against relapsing P. vivax.


Assuntos
Atovaquona , Vacinas Antimaláricas , Plasmodium cynomolgi , Proguanil , Animais , Primaquina/uso terapêutico , Esporozoítos , Macaca mulatta , Imunização , Quimioprevenção , Linfócitos T CD8-Positivos , Combinação de Medicamentos
2.
PLoS One ; 12(2): e0171826, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28182750

RESUMO

Whole malaria sporozoite vaccine regimens are promising new strategies, and some candidates have demonstrated high rates of durable clinical protection associated with memory T cell responses. Little is known about the anatomical distribution of memory T cells following whole sporozoite vaccines, and immunization of nonhuman primates can be used as a relevant model for humans. We conducted a chemoprophylaxis with sporozoite (CPS) immunization in P. knowlesi rhesus monkeys and challenged via mosquito bites. Half of CPS immunized animals developed complete protection, with a marked delay in parasitemia demonstrated in the other half. Antibody responses to whole sporozoites, CSP, and AMA1, but not CelTOS were detected. Peripheral blood T cell responses to whole sporozoites, but not CSP and AMA1 peptides were observed. Unlike peripheral blood, there was a high frequency of sporozoite-specific memory T cells observed in the liver and bone marrow. Interestingly, sporozoite-specific CD4+ and CD8+ memory T cells in the liver highly expressed chemokine receptors CCR5 and CXCR6, both of which are known for liver sinusoid homing. The majority of liver sporozoite-specific memory T cells expressed CD69, a phenotypic marker of tissue-resident memory (TRM) cells, which are well positioned to rapidly control liver-stage infection. Vaccine strategies that aim to elicit large number of liver TRM cells may efficiently increase the efficacy and durability of response against pre-erythrocytic parasites.


Assuntos
Quimioprevenção/métodos , Imunização/métodos , Memória Imunológica , Fígado/imunologia , Malária/prevenção & controle , Plasmodium/imunologia , Esporozoítos/imunologia , Animais , Anopheles/parasitologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Macaca mulatta , Malária/imunologia , Plasmodium/crescimento & desenvolvimento , Plasmodium/patogenicidade
3.
J Immunol ; 195(9): 4378-86, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26408671

RESUMO

Studies of influenza-specific immune responses in humans have largely assessed systemic responses involving serum Ab and peripheral blood T cell responses. However, recent evidence indicates that tissue-resident memory T (TRM) cells play an important role in local murine intrapulmonary immunity. Rhesus monkeys were pulmonary exposed to 2009 pandemic H1N1 virus at days 0 and 28 and immune responses in different tissue compartments were measured. All animals were asymptomatic postinfection. Although only minimal memory immune responses were detected in peripheral blood, a high frequency of influenza nucleoprotein-specific memory T cells was detected in the lung at the "contraction phase," 49-58 d after second virus inoculation. A substantial proportion of lung nucleoprotein-specific memory CD8(+) T cells expressed CD103 and CD69, phenotypic markers of TRM cells. Lung CD103(+) and CD103(-) memory CD8(+) T cells expressed similar levels of IFN-γ and IL-2. Unlike memory T cells, spontaneous Ab secreting cells and memory B cells specific to influenza hemagglutinin were primarily observed in the mediastinal lymph nodes. Little difference in systemic and local immune responses against influenza was observed between young adult (6-8 y) and old animals (18-28 y). Using a nonhuman primate model, we revealed substantial induction of local T and B cell responses following 2009 pandemic H1N1 infection. Our study identified a subset of influenza-specific lung memory T cells characterized as TRM cells in rhesus monkeys. The rhesus monkey model may be useful to explore the role of TRM cells in local tissue protective immunity after rechallenge and vaccination.


Assuntos
Linfócitos B/imunologia , Memória Imunológica/imunologia , Vírus da Influenza A Subtipo H1N1/imunologia , Macaca mulatta/imunologia , Infecções por Orthomyxoviridae/imunologia , Linfócitos T/imunologia , Fatores Etários , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígenos de Diferenciação de Linfócitos T/metabolismo , Linfócitos B/metabolismo , Linfócitos B/virologia , Medula Óssea/imunologia , Medula Óssea/metabolismo , Medula Óssea/virologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/virologia , Células Cultivadas , Interações Hospedeiro-Patógeno/imunologia , Humanos , Vírus da Influenza A Subtipo H1N1/fisiologia , Cadeias alfa de Integrinas/imunologia , Cadeias alfa de Integrinas/metabolismo , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-2/imunologia , Interleucina-2/metabolismo , Lectinas Tipo C/imunologia , Lectinas Tipo C/metabolismo , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/virologia , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/virologia , Macaca mulatta/metabolismo , Macaca mulatta/virologia , Mediastino/virologia , Infecções por Orthomyxoviridae/metabolismo , Infecções por Orthomyxoviridae/virologia , Baço/imunologia , Baço/metabolismo , Baço/virologia , Linfócitos T/metabolismo , Linfócitos T/virologia , Fatores de Tempo
4.
Am J Trop Med Hyg ; 90(1): 149-52, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24277784

RESUMO

We studied cross-reactive antibodies against avian influenza H5N1 and 2009 pandemic (p) H1N1 in 200 serum samples from US military personnel collected before the H1N1 pandemic. Assays used to measure antibodies against viral proteins involved in protection included a hemagglutination inhibition (HI) assay and a neuraminidase inhibition (NI) assay. Viral neutralization by antibodies against avian influenza H5N1 and 2009 pH1N1 was assessed by influenza (H5) pseudotyped lentiviral particle-based and H1N1 microneutralization assays. Some US military personnel had cross-neutralizing antibodies against H5N1 (14%) and 2009 pH1N1 (16.5%). The odds of having cross-neutralizing antibodies against 2009 pH1N1 were 4.4 times higher in subjects receiving more than five inactivated whole influenza virus vaccinations than those subjects with no record of vaccination. Although unclear if the result of prior vaccination or disease exposure, these pre-existing antibodies may prevent or reduce disease severity.


Assuntos
Anticorpos Antivirais/sangue , Vírus da Influenza A Subtipo H1N1/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Influenza Humana/virologia , Militares , Adulto , Idoso , Anticorpos Neutralizantes , Feminino , Humanos , Influenza Humana/sangue , Influenza Humana/imunologia , Masculino , Pessoa de Meia-Idade , Testes de Neutralização , Razão de Chances , Estados Unidos
5.
PLoS One ; 8(3): e59674, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555741

RESUMO

INTRODUCTION: Recent studies have demonstrated that inactivated seasonal influenza vaccines (IIV) may elicit production of heterosubtypic antibodies, which can neutralize avian H5N1 virus in a small proportion of subjects. We hypothesized that prime boost regimens of live and inactivated trivalent seasonal influenza vaccines (LAIV and IIV) would enhance production of heterosubtypic immunity and provide evidence of cross-protection against other influenza viruses. METHODS: In an open-label study, 26 adult volunteers were randomized to receive one of four vaccine regimens containing two doses of 2009-10 seasonal influenza vaccines administered 8 (±1) weeks apart: 2 doses of LAIV; 2 doses of IIV; LAIV then IIV; IIV then LAIV. Humoral immunity assays for avian H5N1, 2009 pandemic H1N1 (pH1N1), and seasonal vaccine strains were performed on blood collected pre-vaccine and 2 and 4 weeks later. The percentage of cytokine-producing T-cells was compared with baseline 14 days after each dose. RESULTS: Subjects receiving IIV had prompt serological responses to vaccine strains. Two subjects receiving heterologous prime boost regimens had enhanced haemagglutination inhibition (HI) and neutralization (NT) titres against pH1N1, and one subject against avian H5N1; all three had pre-existing cross-reactive antibodies detected at baseline. Significantly elevated titres to H5N1 and pH1N1 by neuraminidase inhibition (NI) assay were observed following LAIV-IIV administration. Both vaccines elicited cross-reactive CD4+ T-cell responses to nucleoprotein of avian H5N1 and pH1N1. All regimens were safe and well tolerated. CONCLUSION: Neither homologous nor heterologous prime boost immunization enhanced serum HI and NT titres to 2009 pH1N1 or avian H5N1 compared to single dose vaccine. However heterologous prime-boost vaccination did lead to in vitro evidence of cross-reactivity by NI; the significance of this finding is unclear. These data support the strategy of administering single dose trivalent seasonal influenza vaccine at the outset of an influenza pandemic while a specific vaccine is being developed. TRIAL REGISTRATION: ClinicalTrials.gov NCT01044095.


Assuntos
Reações Cruzadas , Imunização Secundária/métodos , Influenza Aviária/imunologia , Influenza Humana/prevenção & controle , Orthomyxoviridae/imunologia , Pandemias/prevenção & controle , Vacinação/métodos , Adolescente , Adulto , Animais , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Aves , Estudos de Viabilidade , Feminino , Saúde , Humanos , Imunização Secundária/efeitos adversos , Vírus da Influenza A Subtipo H1N1/imunologia , Vírus da Influenza A Subtipo H1N1/fisiologia , Virus da Influenza A Subtipo H5N1/imunologia , Virus da Influenza A Subtipo H5N1/fisiologia , Influenza Aviária/prevenção & controle , Influenza Humana/epidemiologia , Influenza Humana/imunologia , Masculino , Pessoa de Meia-Idade , Orthomyxoviridae/fisiologia , Projetos Piloto , Segurança , Estações do Ano , Linfócitos T/imunologia , Linfócitos T/virologia , Vacinação/efeitos adversos , Vacinas Virais/efeitos adversos , Vacinas Virais/imunologia , Adulto Jovem
6.
Biochem Biophys Res Commun ; 398(4): 752-8, 2010 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-20627090

RESUMO

Information on the immune response against H5N1 within the lung is lacking. Here we describe the sustained antiviral immune responses, as indicated by the expression of MxA protein and IFN-alpha mRNA, in autopsy lung tissue from an H5N1-infected patient. H5N1 infection of primary bronchial/tracheal epithelial cells and lung microvascular endothelial cells induced IP-10, and also up-regulated the retinoic acid-inducible gene-I (RIG-I). Down-regulation of RIG-I gene expression decreased IP-10 response. Co-culturing of H5N1-infected pulmonary cells with TNF-alpha led to synergistically enhanced production of IP-10. In the absence of viral infection, TNF-alpha and IFN-alpha also synergistically enhanced IP-10 response. Methylprednisolone showed only a partial inhibitory effect on this chemokine response. Our findings strongly suggest that both the H5N1 virus and the locally produced antiviral cytokines; IFN-alpha and TNF-alpha may have an important role in inducing IP-10 hyperresponse, leading to inflammatory damage in infected lung.


Assuntos
Quimiocina CXCL10/biossíntese , Virus da Influenza A Subtipo H5N1 , Influenza Humana/imunologia , Pulmão/imunologia , Pulmão/virologia , Pneumonia Viral/imunologia , Células Cultivadas , Quimiocina CXCL10/antagonistas & inibidores , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Proteínas de Ligação ao GTP/biossíntese , Humanos , Interferon-alfa/biossíntese , Interferon-alfa/farmacologia , Metilprednisolona/farmacologia , Proteínas de Resistência a Myxovirus , Receptores Imunológicos , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
8.
J Immunol ; 179(8): 5220-7, 2007 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-17911607

RESUMO

There is worldwide concern that the avian influenza H5N1 virus, with a mortality rate of >50%, might cause the next influenza pandemic. Unlike most other influenza infections, H5N1 infection causes a systemic disease. The underlying mechanisms for this effect are still unclear. In this study, we investigate the interplay between avian influenza H5N1 and human dendritic cells (DC). We showed that H5N1 virus can infect and replicate in monocyte-derived and blood myeloid DC, leading to cell death. These results suggest that H5N1 escapes viral-specific immunity, and could disseminate via DC. In contrast, blood pDC were resistant to infection and produced high amounts of IFN-alpha. Addition of this cytokine to monocyte-derived DC or pretreatment with TLR ligands protected against infection and the cytopathic effects of H5N1 virus.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/virologia , Suscetibilidade a Doenças/imunologia , Virus da Influenza A Subtipo H5N1/imunologia , Interferon-alfa/metabolismo , Receptores Toll-Like/metabolismo , Animais , Antivirais/metabolismo , Aves , Linhagem da Célula/imunologia , Células Cultivadas , Técnicas de Cocultura , Efeito Citopatogênico Viral/imunologia , Células Dendríticas/metabolismo , Humanos , Imunidade Inata , Virus da Influenza A Subtipo H5N1/patogenicidade , Influenza Aviária/imunologia , Influenza Aviária/metabolismo , Influenza Aviária/prevenção & controle , Interferon-alfa/fisiologia , Ligantes , Lipopolissacarídeos/metabolismo , Lipopolissacarídeos/farmacologia , Dados de Sequência Molecular , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/virologia , Receptor 3 Toll-Like/metabolismo , Receptor 3 Toll-Like/fisiologia , Receptor 4 Toll-Like/metabolismo , Receptor 4 Toll-Like/fisiologia , Receptores Toll-Like/fisiologia , Replicação Viral/imunologia
9.
J Immunol ; 172(8): 4926-33, 2004 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-15067072

RESUMO

A common feature of severe Plasmodium falciparum infection is the increased systemic release of proinflammatory cytokines that contributes to the pathogenesis of malaria. Using human blood, we found that blood stage schizonts or soluble schizont extracts activated plasmacytoid dendritic cells (PDCs) to up-regulate CD86 expression and produce IFN-alpha. IFN-alpha production was also detected in malaria-infected patients, but the levels of circulating PDCs were markedly reduced, possibly because of schizont-stimulated up-regulation of CCR7, which is critical for PDC migration. The schizont-stimulated PDCs elicited a poor T cell response, but promoted gamma delta T cell proliferation and IFN-gamma production. The schizont immune stimulatory effects could be reproduced using murine DCs and required the Toll-like receptor 9 (TLR9)-MyD88 signaling pathway. Although the only known TLR9 ligand is CpG motifs in pathogen DNA, the activity of the soluble schizont extract was far greater than that of schizont DNA, and it was heat labile and precipitable with ammonium sulfate, unlike the activity of bacterial DNA. These results demonstrate that schizont extracts contain a novel and previously unknown ligand for TLR9 and suggest that the stimulatory effects of this ligand on PDCs may play a key role in immunoregulation and immunopathogenesis of human falciparum malaria.


Assuntos
Proteínas de Ligação a DNA/fisiologia , Células Dendríticas/imunologia , Células Dendríticas/parasitologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Plasmodium falciparum/crescimento & desenvolvimento , Plasmodium falciparum/imunologia , Receptores de Superfície Celular/fisiologia , Transdução de Sinais/imunologia , Proteínas Adaptadoras de Transdução de Sinal , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/fisiologia , Fracionamento Celular , Movimento Celular/imunologia , Células Cultivadas , Técnicas de Cocultura , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Humanos , Imunidade Celular , Interferon-alfa/biossíntese , Interferon-alfa/sangue , Malária Falciparum/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 88 de Diferenciação Mieloide , Plasmodium falciparum/química , Receptores de Antígenos de Linfócitos T gama-delta/biossíntese , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/genética , Receptores Imunológicos/deficiência , Receptores Imunológicos/genética , Receptores Imunológicos/fisiologia , Transdução de Sinais/genética , Solubilidade , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/parasitologia , Receptor Toll-Like 9 , Regulação para Cima/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA