Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 231
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Nat Immunol ; 22(3): 347-357, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33432229

RESUMO

Activated Vγ9Vδ2 (γδ2) T lymphocytes that sense parasite-produced phosphoantigens are expanded in Plasmodium falciparum-infected patients. Although previous studies suggested that γδ2 T cells help control erythrocytic malaria, whether γδ2 T cells recognize infected red blood cells (iRBCs) was uncertain. Here we show that iRBCs stained for the phosphoantigen sensor butyrophilin 3A1 (BTN3A1). γδ2 T cells formed immune synapses and lysed iRBCs in a contact, phosphoantigen, BTN3A1 and degranulation-dependent manner, killing intracellular parasites. Granulysin released into the synapse lysed iRBCs and delivered death-inducing granzymes to the parasite. All intra-erythrocytic parasites were susceptible, but schizonts were most sensitive. A second protective γδ2 T cell mechanism was identified. In the presence of patient serum, γδ2 T cells phagocytosed and degraded opsonized iRBCs in a CD16-dependent manner, decreasing parasite multiplication. Thus, γδ2 T cells have two ways to control blood-stage malaria-γδ T cell antigen receptor (TCR)-mediated degranulation and phagocytosis of antibody-coated iRBCs.


Assuntos
Antígenos de Protozoários/imunologia , Citotoxicidade Imunológica , Eritrócitos/imunologia , Linfócitos Intraepiteliais/imunologia , Ativação Linfocitária , Malária Falciparum/imunologia , Fagocitose , Plasmodium falciparum/microbiologia , Antígenos CD/metabolismo , Antígenos de Diferenciação de Linfócitos T/metabolismo , Antígenos de Protozoários/sangue , Boston , Brasil , Butirofilinas/metabolismo , Células Cultivadas , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Feminino , Granzimas/metabolismo , Interações Hospedeiro-Parasita , Humanos , Sinapses Imunológicas/metabolismo , Sinapses Imunológicas/parasitologia , Linfócitos Intraepiteliais/metabolismo , Linfócitos Intraepiteliais/parasitologia , Malária Falciparum/sangue , Malária Falciparum/parasitologia , Masculino , Plasmodium falciparum/crescimento & desenvolvimento
2.
Immunol Rev ; 293(1): 163-189, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31642531

RESUMO

Plasmodium vivax infection, the predominant cause of malaria in Asia and Latin America, affects ~14 million individuals annually, with considerable adverse effects on wellbeing and socioeconomic development. A clinical hallmark of Plasmodium infection, the paroxysm, is driven by pyrogenic cytokines produced during the immune response. Here, we review studies on the role of specific immune cell types, cognate innate immune receptors, and inflammatory cytokines on parasite control and disease symptoms. This review also summarizes studies on recurrent infections in individuals living in endemic regions as well as asymptomatic infections, a serious barrier to eliminating this disease. We propose potential mechanisms behind these repeated and subclinical infections, such as poor induction of immunological memory cells and inefficient T effector cells. We address the role of antibody-mediated resistance to P. vivax infection and discuss current progress in vaccine development. Finally, we review immunoregulatory mechanisms, such as inhibitory receptors, T regulatory cells, and the anti-inflammatory cytokine, IL-10, that antagonizes both innate and acquired immune responses, interfering with the development of protective immunity and parasite clearance. These studies provide new insights for the clinical management of symptomatic as well as asymptomatic individuals and the development of an efficacious vaccine for vivax malaria.


Assuntos
Interações Hospedeiro-Parasita/imunologia , Imunidade , Malária Vivax/imunologia , Malária Vivax/parasitologia , Plasmodium vivax/fisiologia , Imunidade Adaptativa , Anticorpos Antiprotozoários/imunologia , Antígenos de Protozoários/imunologia , Citocinas/metabolismo , Suscetibilidade a Doenças , Interações Hospedeiro-Parasita/genética , Humanos , Imunidade Inata , Mediadores da Inflamação/metabolismo , Vacinas Antimaláricas/imunologia , Malária Vivax/genética , Malária Vivax/metabolismo , Plasmodium vivax/crescimento & desenvolvimento , Receptores Toll-Like/metabolismo
3.
J Immunol ; 206(7): 1642-1652, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33627376

RESUMO

Human primary monocytes are composed of a minor, more mature CD16+(CD14low/neg) population and a major CD16neg(CD14+) subset. The specific functions of CD16+ versus CD16neg monocytes in steady state or inflammation remain poorly understood. In previous work, we found that IL-12 is selectively produced by the CD16+ subset in response to the protozoan pathogen, Toxoplasma gondii In this study, we demonstrated that this differential responsiveness correlates with the presence of an IFN-induced transcriptional signature in CD16+ monocytes already at baseline. Consistent with this observation, we found that in vitro IFN-γ priming overcomes the defect in the IL-12 response of the CD16neg subset. In contrast, pretreatment with IFN-γ had only a minor effect on IL-12p40 secretion by the CD16+ population. Moreover, inhibition of the mTOR pathway also selectively increased the IL-12 response in CD16neg but not in CD16+ monocytes. We further demonstrate that in contrast to IFN-γ, IFN-α fails to promote IL-12 production by the CD16neg subset and blocks the effect of IFN-γ priming. Based on these observations, we propose that the acquisition of IL-12 responsiveness by peripheral blood monocyte subsets depends on extrinsic signals experienced during their developmental progression in vivo. This process can be overridden during inflammation by the opposing regulatory effects of type I and II IFN as well as the mTOR inhibition.


Assuntos
Inflamação/imunologia , Subunidade p40 da Interleucina-12/metabolismo , Monócitos/imunologia , Toxoplasma/fisiologia , Toxoplasmose/imunologia , Diferenciação Celular , Células Cultivadas , Humanos , Interferon gama/metabolismo , Receptores de Lipopolissacarídeos/metabolismo , Cultura Primária de Células , Receptores de IgG/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Transcriptoma
4.
PLoS Pathog ; 16(9): e1008840, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32913355

RESUMO

P. vivax-infected Retics (iRetics) express human leukocyte antigen class I (HLA-I), are recognized by CD8+ T cells and killed by granulysin (GNLY) and granzymes. However, how Plasmodium infection induces MHC-I expression on Retics is unknown. In addition, whether GNLY helps control Plasmodium infection in vivo has not been studied. Here, we examine these questions using rodent infection with the P. yoelii 17XNL strain, which has tropism for Retics. Infection with P. yoelii caused extramedullary erythropoiesis, reticulocytosis and expansion of CD8+CD44+CD62L- IFN-γ-producing T cells that form immune synapses with iRetics. We now provide evidence that MHC-I expression by iRetic is dependent on IFN-γ-induced transcription of IRF-1, MHC-I and ß2-microglobulin (ß2-m) in erythroblasts. Consistently, CTLs from infected wild type (WT) mice formed immune synapses with iRetics in an IFN-γ- and MHC-I-dependent manner. When challenged with P. yoelii 17XNL, WT mice cleared parasitemia and survived, while IFN-γ KO mice remained parasitemic and all died. ß2-m KO mice that do not express MHC-I and have virtually no CD8+ T cells had prolonged parasitemia, and 80% survived. Because mice do not express GNLY, GNLY-transgenic mice can be used to assess the in vivo importance of GNLY. Parasite clearance was accelerated in GNLY-transgenic mice and depletion of CD8+ T cells ablated the GNLY-mediated resistance to P. yoelii. Altogether, our results indicate that in addition to previously described mechanisms, IFN-γ promotes host resistance to the Retic-tropic P. yoelii 17XNL strain by promoting MHC-I expression on iRetics that become targets for CD8+ cytotoxic T lymphocytes and GNLY.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Linfócitos T CD8-Positivos/imunologia , Interferon gama/imunologia , Malária/imunologia , Plasmodium yoelii/imunologia , Animais , Antígenos de Diferenciação de Linfócitos T/genética , Linfócitos T CD8-Positivos/patologia , Interferon gama/genética , Malária/genética , Malária/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout
5.
PLoS Pathog ; 16(8): e1008781, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32810179

RESUMO

Chagas disease is caused by Trypanosoma cruzi, a protozoan parasite that has a heterogeneous population composed of a pool of strains with distinct characteristics, including variable levels of virulence. In previous work, transcriptome analyses of parasite genes after infection of human foreskin fibroblasts (HFF) with virulent (CL Brener) and non-virulent (CL-14) clones derived from the CL strain, revealed a reduced expression of genes encoding parasite surface proteins in CL-14 compared to CL Brener during the final steps of the intracellular differentiation from amastigotes to trypomastigotes. Here we analyzed changes in the expression of host genes during in vitro infection of HFF cells with the CL Brener and CL-14 strains by analyzing total RNA extracted from cells at 60 and 96 hours post-infection (hpi) with each strain, as well as from uninfected cells. Similar transcriptome profiles were observed at 60 hpi with both strains compared to uninfected samples. However, at 96 hpi, significant differences in the number and expression levels of several genes, particularly those involved with immune response and cytoskeleton organization, were observed. Further analyses confirmed the difference in the chemokine/cytokine signaling involved with the recruitment and activation of immune cells such as neutrophils upon T. cruzi infection. These findings suggest that infection with the virulent CL Brener strain induces a more robust inflammatory response when compared with the non-virulent CL-14 strain. Importantly, the RNA-Seq data also exposed an unexplored role of fibroblasts as sentinel cells that may act by recruiting neutrophils to the initial site of infection. This role for fibroblasts in the regulation of the inflammatory response during infection by T. cruzi was corroborated by measurements of levels of different chemokines/cytokines during in vitro infection and in plasma from Chagas disease patients as well as by neutrophil activation and migration assays.


Assuntos
Doença de Chagas/metabolismo , Fibroblastos , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Ativação de Neutrófilo , Neutrófilos , Trypanosoma cruzi/metabolismo , Doença de Chagas/genética , Doença de Chagas/patologia , Fibroblastos/metabolismo , Fibroblastos/parasitologia , Fibroblastos/patologia , Humanos , Neutrófilos/metabolismo , Neutrófilos/parasitologia , Neutrófilos/patologia , Trypanosoma cruzi/genética , Trypanosoma cruzi/patogenicidade , Fatores de Virulência/genética , Fatores de Virulência/metabolismo
6.
Mem Inst Oswaldo Cruz ; 117: e200314, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35544856

RESUMO

This review does not intend to convey detailed experimental or bibliographic data. Instead, it expresses the informal authors' personal views on topics that range from basic research on antigens and experimental models for Trypanosoma cruzi infection to vaccine prospects and vaccine production. The review also includes general aspects of Chagas' disease control and international and national policies on the subject. The authors contributed equally to the paper.


Assuntos
Doença de Chagas , Trypanosoma cruzi , Vacinas , Antígenos , Doença de Chagas/prevenção & controle , Humanos
7.
Emerg Infect Dis ; 26(12)2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33219657

RESUMO

In 2015, an outbreak of presumed waterborne toxoplasmosis occurred in Gouveia, Brazil. We conducted a 3-year prospective study on a cohort of 52 patients from this outbreak, collected clinical and multimodal imaging findings, and determined risk factors for ocular involvement. At baseline examination, 12 (23%) patients had retinochoroiditis; 4 patients had bilateral and 2 had macular lesions. Multimodal imaging revealed 2 distinct retinochoroiditis patterns: necrotizing focal retinochoroiditis and punctate retinochoroiditis. Older age, worse visual acuity, self-reported recent reduction of visual acuity, and presence of floaters were associated with retinochoroiditis. Among patients, persons >40 years of age had 5 times the risk for ocular involvement. Five patients had recurrences during follow-up, a rate of 22% per person-year. Recurrences were associated with binocular involvement. Two patients had late ocular involvement that occurred >34 months after initial diagnosis. Patients with acquired toxoplasmosis should have long-term ophthalmic follow-up, regardless of initial ocular involvement.


Assuntos
Coriorretinite/diagnóstico por imagem , Surtos de Doenças , Imagem Multimodal/métodos , Toxoplasmose Ocular/diagnóstico por imagem , Idoso , Brasil/epidemiologia , Coriorretinite/epidemiologia , Humanos , Estudos Prospectivos , Fatores de Risco , Toxoplasmose Ocular/epidemiologia
8.
Immunity ; 35(2): 194-207, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21820332

RESUMO

Although Toll-like receptor 9 (TLR9) has been implicated in cytokine and type I interferon (IFN) production during malaria in humans and mice, the high AT content of the Plasmodium falciparum genome prompted us to examine the possibility that malarial DNA triggered TLR9-independent pathways. Over 6000 ATTTTTAC ("AT-rich") motifs are present in the genome of P. falciparum, which we show here potently induce type I IFNs. Parasite DNA, parasitized erythrocytes and oligonucleotides containing the AT-rich motif induce type I IFNs via a pathway that did not involve the previously described sensors TLR9, DAI, RNA polymerase-III or IFI16/p204. Rather, AT-rich DNA sensing involved an unknown receptor that coupled to the STING, TBK1 and IRF3-IRF7 signaling pathway. Mice lacking IRF3, IRF7, the kinase TBK1 or the type I IFN receptor were resistant to otherwise lethal cerebral malaria. Collectively, these observations implicate AT-rich DNA sensing via STING, TBK1 and IRF3-IRF7 in P. falciparum malaria.


Assuntos
Sequência Rica em At/genética , DNA de Protozoário/genética , Malária Falciparum/imunologia , Oligonucleotídeos/genética , Plasmodium falciparum/fisiologia , Animais , DNA de Protozoário/metabolismo , Perfilação da Expressão Gênica , Humanos , Imunidade Inata/genética , Fator Regulador 3 de Interferon/metabolismo , Fator Regulador 7 de Interferon/metabolismo , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Malária Falciparum/parasitologia , Malária Falciparum/fisiopatologia , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Oligonucleotídeos/metabolismo , Plasmodium falciparum/patogenicidade , Proteínas Serina-Treonina Quinases/metabolismo , Receptor de Interferon alfa e beta/genética , Transdução de Sinais/genética
9.
J Immunol ; 200(4): 1243-1248, 2018 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-29330325

RESUMO

Malarial infection in naive individuals induces a robust innate immune response. In the recently described model of innate immune memory, an initial stimulus primes the innate immune system to either hyperrespond (termed training) or hyporespond (tolerance) to subsequent immune challenge. Previous work in both mice and humans demonstrated that infection with malaria can both serve as a priming stimulus and promote tolerance to subsequent infection. In this study, we demonstrate that initial stimulation with Plasmodium falciparum-infected RBCs or the malaria crystal hemozoin induced human adherent PBMCs to hyperrespond to subsequent ligation of TLR2. This hyperresponsiveness correlated with increased H3K4me3 at important immunometabolic promoters, and these epigenetic modifications were also seen in Kenyan children naturally infected with malaria. However, the use of epigenetic and metabolic inhibitors indicated that the induction of trained immunity by malaria and its ligands may occur via a previously unrecognized mechanism(s).


Assuntos
Imunidade Inata/imunologia , Malária Falciparum/imunologia , Criança , Pré-Escolar , Epigênese Genética/imunologia , Feminino , Regulação da Expressão Gênica/imunologia , Histonas/biossíntese , Humanos , Lactente , Malária Falciparum/metabolismo , Masculino
10.
PLoS Pathog ; 13(12): e1006767, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-29240831

RESUMO

Trypanosoma cruzi, the protozoan that causes Chagas disease, has a complex life cycle involving several morphologically and biochemically distinct stages that establish intricate interactions with various insect and mammalian hosts. It has also a heterogeneous population structure comprising strains with distinct properties such as virulence, sensitivity to drugs, antigenic profile and tissue tropism. We present a comparative transcriptome analysis of two cloned T. cruzi strains that display contrasting virulence phenotypes in animal models of infection: CL Brener is a virulent clone and CL-14 is a clone that is neither infective nor pathogenic in in vivo models of infection. Gene expression analysis of trypomastigotes and intracellular amastigotes harvested at 60 and 96 hours post-infection (hpi) of human fibroblasts revealed large differences that reflect the parasite's adaptation to distinct environments during the infection of mammalian cells, including changes in energy sources, oxidative stress responses, cell cycle control and cell surface components. While extensive transcriptome remodeling was observed when trypomastigotes of both strains were compared to 60 hpi amastigotes, differences in gene expression were much less pronounced when 96 hpi amastigotes and trypomastigotes of CL Brener were compared. In contrast, the differentiation of the avirulent CL-14 from 96 hpi amastigotes to extracellular trypomastigotes was associated with considerable changes in gene expression, particularly in gene families encoding surface proteins such as trans-sialidases, mucins and the mucin associated surface proteins (MASPs). Thus, our comparative transcriptome analysis indicates that the avirulent phenotype of CL-14 may be due, at least in part, to a reduced or delayed expression of genes encoding surface proteins that are associated with the transition of amastigotes to trypomastigotes, an essential step in the establishment of the infection in the mammalian host. Confirming the role of members of the trans-sialidase family of surface proteins for parasite differentiation, transfected CL-14 constitutively expressing a trans-sialidase gene displayed faster kinetics of trypomastigote release in the supernatant of infected cells compared to wild type CL-14.


Assuntos
Doença de Chagas/parasitologia , Trypanosoma cruzi/genética , Trypanosoma cruzi/patogenicidade , Animais , Perfilação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Ontologia Genética , Genes de Protozoários , Glicoproteínas/genética , Interações Hospedeiro-Parasita , Humanos , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Neuraminidase/genética , Proteínas de Protozoários/genética , Proteínas de Ligação a RNA/genética , Trypanosoma cruzi/crescimento & desenvolvimento , Virulência/genética
11.
PLoS Pathog ; 13(7): e1006484, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28700710

RESUMO

Although the importance of humoral immunity to malaria has been established, factors that control antibody production are poorly understood. Follicular helper T cells (Tfh cells) are pivotal for generating high-affinity, long-lived antibody responses. While it has been proposed that expansion of antigen-specific Tfh cells, interleukin (IL) 21 production and robust germinal center formation are associated with protection against malaria in mice, whether Tfh cells are found during Plasmodium vivax (P. vivax) infection and if they play a role during disease remains unknown. Our goal was to define the role of Tfh cells during P. vivax malaria. We demonstrate that P. vivax infection triggers IL-21 production and an increase in Tfh cells (PD-1+ICOS+CXCR5+CD45RO+CD4+CD3+). As expected, FACS-sorted Tfh cells, the primary source of IL-21, induced immunoglobulin production by purified naïve B cells. Furthermore, we found that P. vivax infection alters the B cell compartment and these alterations were dependent on the number of previous infections. First exposure leads to increased proportions of activated and atypical memory B cells and decreased frequencies of classical memory B cells, whereas patients that experienced multiple episodes displayed lower proportions of atypical B cells and higher frequencies of classical memory B cells. Despite the limited sample size, but consistent with the latter finding, the data suggest that patients who had more than five infections harbored more Tfh cells and produce more specific antibodies. P. vivax infection triggers IL-21 production by Tfh that impact B cell responses in humans.


Assuntos
Anticorpos Antiprotozoários/imunologia , Linfócitos B/imunologia , Malária Vivax/imunologia , Plasmodium vivax/fisiologia , Linfócitos T Auxiliares-Indutores/imunologia , Adolescente , Adulto , Animais , Feminino , Humanos , Ativação Linfocitária , Malária Vivax/parasitologia , Masculino , Camundongos , Pessoa de Meia-Idade , Plasmodium vivax/imunologia , Adulto Jovem
12.
Cytokine ; 113: 285-290, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30037707

RESUMO

This study investigated the efficacy of the vaccine in liver of mice infected with the Trypanosoma cruzi (T. cruzi) and immunized with AdASP-2. For this purpose, histopathological analysis and gene expression of COX-2, TNF-alpha, TNFR, iNOS, cytochrome C, caspase-3, TLR4, IL-6 and IL10 were evaluated. The following groups were used in this study: Group 1 - Control Group (CTRL) animals received AdßGal vehicle; Group 2 - Infected Group (TC) animals were infected with T. cruzi; Group 3 - Immunized Group (AdASP-2): animals were immunized by AdASP-2 vaccine; Group 4 - Immunized and Infected Group (AdASP-2+TC) animals were infected with T. cruzi and immunized by AdSP-2 vaccine. A significant decrease of amastigote nests was noticed in the group of animals that were immunized with AdASP-2 and infected on the same day. COX-2 and TNF-alpha gene expressions increased in TC group, whereas TNF-alpha decreased in the TC+AdASP-2 group. TNFR expression was high in AdASP-2+TC group. iNOS expression was high for all experimental groups whereas cytochrome C decreased for all experimental groups. Caspase 3 increased in TC and TC+AdASP-2 groups. The gene expression of TLR4 and IL-10 showed an increase in AdASP-2+TC group. Finally, hepatic fibrosis was noticed to TC and AdASP-2 + TC groups. Taken together, our results demonstrated that vaccination with AdASP-2 was effective against the acute phase of experimental Chagas disease as a result of a more powerful and rapid immune response closely related to expression of some inflammatory genes, such as iNOS, TNF-alpha, TLR 4, and IL-10.


Assuntos
Cardiomiopatia Chagásica/imunologia , Cirrose Hepática/imunologia , Fígado/imunologia , Neuraminidase/imunologia , Vacinas Protozoárias/imunologia , Trypanosoma cruzi/imunologia , Adenoviridae , Animais , Caspase 3/imunologia , Cardiomiopatia Chagásica/parasitologia , Cardiomiopatia Chagásica/patologia , Cardiomiopatia Chagásica/prevenção & controle , Ciclo-Oxigenase 2/imunologia , Citocromos c/imunologia , Citocinas/imunologia , Feminino , Fígado/parasitologia , Fígado/patologia , Cirrose Hepática/parasitologia , Cirrose Hepática/patologia , Cirrose Hepática/prevenção & controle , Camundongos , Óxido Nítrico Sintase Tipo II/imunologia , Receptor 4 Toll-Like/imunologia
13.
Mem Inst Oswaldo Cruz ; 114: e180405, 2019 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-30726344

RESUMO

BACKGROUND: Visceral Leishmaniasis (VL) is an infectious disease that is a significant cause of death among infants aged under 1 year and the elderly in Brazil. Serodiagnosis is a mainstay of VL elimination programs; however, it has significant limitations due to low accuracy. OBJECTIVE: This study aimed to evaluate three recombinant Leishmania infantum proteins (rFc, rC9, and rA2) selected from previous proteomics and genomics analyses to develop enzyme-linked immunosorbent assay (ELISA) and immunochromatographic tests (ICT) for the serodiagnosis of human VL (HVL) and canine VL (CVL). METHODS: A total of 186 human (70 L. infantum-infected symptomatic, 20 other disease-infected, and 96 healthy) and 185 canine (82 L. infantum-infected symptomatic, 27 L. infantum-infected asymptomatic, and 76 healthy) sera samples were used for antibody detection. FINDINGS: Of the three proteins, rA2 (91.5% sensitivity and 87% specificity) and rC9 (95.7% sensitivity and 87.5% specificity) displayed the best performance in ELISA-HVL and ELISA-CVL, respectively. ICT-rA2 also displayed the best performance for HVL diagnosis (92.3% sensitivity and 88.0% specificity) and had high concordance with immunofluorescence antibody tests (IFAT), ELISA-rK39, IT-LEISH®, and ELISAEXT. ICT-rFc, ICT-rC9, and ICT-rA2 had sensitivities of 88.6%, 86.5%, and 87.0%, respectively, with specificity values of 84.0%, 92.0%, and 100%, respectively for CVL diagnosis. MAIN CONCLUSIONS: The three antigens selected by us are promising candidates for VL diagnosis regardless of the test format, although the antigen combinations and test parameters may warrant further optimisation.


Assuntos
Anticorpos Antiprotozoários/sangue , Antígenos de Protozoários/sangue , Leishmania infantum/imunologia , Leishmaniose Visceral/diagnóstico , Proteínas de Protozoários/sangue , Animais , Antígenos de Protozoários/imunologia , Estudos de Casos e Controles , Cromatografia de Afinidade , Cães , Ensaio de Imunoadsorção Enzimática , Humanos , Leishmaniose Visceral/veterinária , Proteínas de Protozoários/imunologia , Proteínas Recombinantes/sangue , Proteínas Recombinantes/imunologia , Sensibilidade e Especificidade
14.
J Infect Dis ; 218(8): 1314-1323, 2018 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-29800313

RESUMO

The balance between pro- and antiinflammatory mechanisms is essential to limit immune-mediated pathology, and CD4+ forkhead box P3 (Foxp3+) regulatory T cells (Treg) play an important role in this process. The expression of inhibitory receptors regulates cytokine production by Plasmodium vivax-specific T cells. Our goal was to assess the induction of programmed death-1 (PD-1) and cytotoxic T-lymphocyte antigen (CTLA-4) on Treg during malaria and to evaluate their function. We found that P. vivax infection triggered an increase in circulating Treg and their expression of CTLA-4 and PD-1. Functional analysis demonstrated that Treg from malaria patients had impaired suppressive ability and PD-1+Treg displayed lower levels of Foxp3 and Helios, but had higher frequencies of T-box transcription factor+ and interferon-gamma+ cells than PD-1-Treg. Thus malaria infection alters the function of circulating Treg by triggering increased expression of PD-1 on Treg that is associated with decreased regulatory function and increased proinflammatory characteristics.


Assuntos
Malária Vivax/imunologia , Malária Vivax/parasitologia , Linfócitos T Reguladores/fisiologia , Adulto , Proliferação de Células , Citocinas/genética , Citocinas/metabolismo , Feminino , Regulação da Expressão Gênica/imunologia , Humanos , Imunofenotipagem , Masculino , Pessoa de Meia-Idade , Plasmodium vivax , Reticulócitos/parasitologia , Reticulócitos/fisiologia , Adulto Jovem
15.
PLoS Pathog ; 12(6): e1005669, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27280707

RESUMO

Leishmania amastigotes manipulate the activity of macrophages to favor their own success. However, very little is known about the role of innate recognition and signaling triggered by amastigotes in this host-parasite interaction. In this work we developed a new infection model in adult Drosophila to take advantage of its superior genetic resources to identify novel host factors limiting Leishmania amazonensis infection. The model is based on the capacity of macrophage-like cells, plasmatocytes, to phagocytose and control the proliferation of parasites injected into adult flies. Using this model, we screened a collection of RNAi-expressing flies for anti-Leishmania defense factors. Notably, we found three CD36-like scavenger receptors that were important for defending against Leishmania infection. Mechanistic studies in mouse macrophages showed that CD36 accumulates specifically at sites where the parasite contacts the parasitophorous vacuole membrane. Furthermore, CD36-deficient macrophages were defective in the formation of the large parasitophorous vacuole typical of L. amazonensis infection, a phenotype caused by inefficient fusion with late endosomes and/or lysosomes. These data identify an unprecedented role for CD36 in the biogenesis of the parasitophorous vacuole and further highlight the utility of Drosophila as a model system for dissecting innate immune responses to infection.


Assuntos
Antígenos CD36/imunologia , Endocitose/fisiologia , Interações Hospedeiro-Parasita/fisiologia , Leishmaniose/imunologia , Macrófagos/microbiologia , Animais , Antígenos CD36/metabolismo , Modelos Animais de Doenças , Drosophila , Imunidade Inata/imunologia , Leishmania mexicana/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Microscopia Confocal , Reação em Cadeia da Polimerase , Vacúolos
16.
PLoS Pathog ; 12(4): e1005593, 2016 04.
Artigo em Inglês | MEDLINE | ID: mdl-27128676

RESUMO

The ß1i, ß2i and ß5i immunoproteasome subunits have an important role in defining the repertoire of MHC class I-restricted epitopes. However, the impact of combined deficiency of the three immunoproteasome subunits in the development of protective immunity to intracellular pathogens has not been investigated. Here, we demonstrate that immunoproteasomes play a key role in host resistance and genetic vaccination-induced protection against the human pathogen Trypanosoma cruzi (the causative agent of Chagas disease), immunity to which is dependent on CD8+ T cells and IFN-γ (the classical immunoproteasome inducer). We observed that infection with T. cruzi triggers the transcription of immunoproteasome genes, both in mice and humans. Importantly, genetically vaccinated or T. cruzi-infected ß1i, ß2i and ß5i triple knockout (TKO) mice presented significantly lower frequencies and numbers of splenic CD8+ effector T cells (CD8+CD44highCD62Llow) specific for the previously characterized immunodominant (VNHRFTLV) H-2Kb-restricted T. cruzi epitope. Not only the quantity, but also the quality of parasite-specific CD8+ T cell responses was altered in TKO mice. Hence, the frequency of double-positive (IFN-γ+/TNF+) or single-positive (IFN-γ+) cells specific for the H-2Kb-restricted immunodominant as well as subdominant T. cruzi epitopes were higher in WT mice, whereas TNF single-positive cells prevailed among CD8+ T cells from TKO mice. Contrasting with their WT counterparts, TKO animals were also lethally susceptible to T. cruzi challenge, even after an otherwise protective vaccination with DNA and adenoviral vectors. We conclude that the immunoproteasome subunits are key determinants in host resistance to T. cruzi infection by influencing both the magnitude and quality of CD8+ T cell responses.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Doença de Chagas/imunologia , Complexo de Endopeptidases do Proteassoma/imunologia , Vacinas Protozoárias/imunologia , Adolescente , Adulto , Animais , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Trypanosoma cruzi , Adulto Jovem
17.
J Immunol ; 196(12): 5056-63, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27183605

RESUMO

Leishmaniasis is an important parasitic disease found in the tropics and subtropics. Cutaneous and visceral leishmaniasis affect an estimated 1.5 million people worldwide. Despite its human health relevance, relatively little is known about the cell death pathways that control Leishmania replication in the host. Necroptosis is a recently identified form of cell death with potent antiviral effects. Receptor interacting protein kinase 1 (RIPK1) is a critical kinase that mediates necroptosis downstream of death receptors and TLRs. Heme, a product of hemoglobin catabolism during certain intracellular pathogen infections, is also a potent inducer of macrophage necroptosis. We found that human visceral leishmaniasis patients exhibit elevated serum levels of heme. Therefore, we examined the impact of heme and necroptosis on Leishmania replication. Indeed, heme potently inhibited Leishmania replication in bone marrow-derived macrophages. Moreover, we found that inhibition of RIPK1 kinase activity also enhanced parasite replication in the absence of heme. We further found that the mitochondrial phosphatase phosphoglycerate mutase family member 5 (PGAM5), a putative downstream effector of RIPK1, was also required for inhibition of Leishmania replication. In mouse infection, both PGAM5 and RIPK1 kinase activity are required for IL-1ß expression in response to Leishmania However, PGAM5, but not RIPK1 kinase activity, was directly responsible for Leishmania-induced IL-1ß secretion and NO production in bone marrow-derived macrophages. Collectively, these results revealed that RIPK1 and PGAM5 function independently to exert optimal control of Leishmania replication in the host.


Assuntos
Interações Hospedeiro-Parasita , Leishmania/crescimento & desenvolvimento , Leishmania/imunologia , Leishmaniose/parasitologia , Fosfoproteínas Fosfatases/metabolismo , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Animais , Morte Celular , Heme/análise , Heme/farmacologia , Humanos , Interleucina-1beta/genética , Interleucina-1beta/imunologia , Interleucina-1beta/metabolismo , Leishmania/efeitos dos fármacos , Leishmaniose/sangue , Leishmaniose/imunologia , Leishmaniose/microbiologia , Leishmaniose Visceral/sangue , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Macrófagos/microbiologia , Macrófagos/fisiologia , Camundongos , Óxido Nítrico/biossíntese , Fosfoproteínas Fosfatases/genética , Fosfoproteínas Fosfatases/imunologia , Proteína Serina-Treonina Quinases de Interação com Receptores/antagonistas & inibidores
18.
PLoS Pathog ; 11(1): e1004594, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25617628

RESUMO

Chagas disease (CD), caused by the protozoan Trypanosoma cruzi, is a prototypical neglected tropical disease. Specific immunity promotes acute phase survival. Nevertheless, one-third of CD patients develop chronic chagasic cardiomyopathy (CCC) associated with parasite persistence and immunological unbalance. Currently, the therapeutic management of patients only mitigates CCC symptoms. Therefore, a vaccine arises as an alternative to stimulate protective immunity and thereby prevent, delay progression and even reverse CCC. We examined this hypothesis by vaccinating mice with replication-defective human Type 5 recombinant adenoviruses (rAd) carrying sequences of amastigote surface protein-2 (rAdASP2) and trans-sialidase (rAdTS) T. cruzi antigens. For prophylactic vaccination, naïve C57BL/6 mice were immunized with rAdASP2+rAdTS (rAdVax) using a homologous prime/boost protocol before challenge with the Colombian strain. For therapeutic vaccination, rAdVax administration was initiated at 120 days post-infection (dpi), when mice were afflicted by CCC. Mice were analyzed for electrical abnormalities, immune response and cardiac parasitism and tissue damage. Prophylactic immunization with rAdVax induced antibodies and H-2Kb-restricted cytotoxic and interferon (IFN)γ-producing CD8+ T-cells, reduced acute heart parasitism and electrical abnormalities in the chronic phase. Therapeutic vaccination increased survival and reduced electrical abnormalities after the prime (analysis at 160 dpi) and the boost (analysis at 180 and 230 dpi). Post-therapy mice exhibited less heart injury and electrical abnormalities compared with pre-therapy mice. rAdVax therapeutic vaccination preserved specific IFNγ-mediated immunity but reduced the response to polyclonal stimuli (anti-CD3 plus anti-CD28), CD107a+ CD8+ T-cell frequency and plasma nitric oxide (NO) levels. Moreover, therapeutic rAdVax reshaped immunity in the heart tissue as reduced the number of perforin+ cells, preserved the number of IFNγ+ cells, increased the expression of IFNγ mRNA but reduced inducible NO synthase mRNA. Vaccine-based immunostimulation with rAd might offer a rational alternative for re-programming the immune response to preserve and, moreover, recover tissue injury in Chagas' heart disease.


Assuntos
Cardiomiopatia Chagásica/prevenção & controle , Doença de Chagas/imunologia , Doença de Chagas/terapia , Vacinas Protozoárias/uso terapêutico , Trypanosoma cruzi/imunologia , Adenoviridae/genética , Adenoviridae/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Doença Crônica , Feminino , Fenômenos do Sistema Imunitário , Camundongos , Camundongos Endogâmicos C57BL , Vacinação , Vacinas de DNA/genética , Vacinas de DNA/imunologia
19.
Nat Rev Immunol ; 6(12): 895-906, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17110955

RESUMO

Toll-like receptors (TLRs) have emerged as a major receptor family involved in non-self recognition. They have a vital role in triggering innate immunity and orchestrate the acquired immune response during bacterial and viral infection. However, the role of TLRs during infection with protozoan pathogens is less clear. Nevertheless, our understanding of how these parasitic microorganisms engage the host TLR signalling system has now entered a phase of rapid expansion. This Review describes recent insights into how parasitic protozoans are sensed by TLR molecules, and how the TLR system itself can be targeted by these microbial pathogens for their own survival.


Assuntos
Eucariotos/imunologia , Infecções por Protozoários/imunologia , Receptores Toll-Like/fisiologia , Animais , Interações Hospedeiro-Parasita , Camundongos , Transdução de Sinais
20.
PLoS Pathog ; 10(9): e1004393, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25233271

RESUMO

Infection with Plasmodium vivax results in strong activation of monocytes, which are important components of both the systemic inflammatory response and parasite control. The overall goal of this study was to define the role of monocytes during P. vivax malaria. Here, we demonstrate that P. vivax-infected patients display significant increase in circulating monocytes, which were defined as CD14(+)CD16- (classical), CD14(+)CD16(+) (inflammatory), and CD14loCD16(+) (patrolling) cells. While the classical and inflammatory monocytes were found to be the primary source of pro-inflammatory cytokines, the CD16(+) cells, in particular the CD14(+)CD16(+) monocytes, expressed the highest levels of activation markers, which included chemokine receptors and adhesion molecules. Morphologically, CD14(+) were distinguished from CD14lo monocytes by displaying larger and more active mitochondria. CD14(+)CD16(+) monocytes were more efficient in phagocytizing P. vivax-infected reticulocytes, which induced them to produce high levels of intracellular TNF-α and reactive oxygen species. Importantly, antibodies specific for ICAM-1, PECAM-1 or LFA-1 efficiently blocked the phagocytosis of infected reticulocytes by monocytes. Hence, our results provide key information on the mechanism by which CD14(+)CD16(+) cells control parasite burden, supporting the hypothesis that they play a role in resistance to P. vivax infection.


Assuntos
Eritrócitos/imunologia , Inflamação/imunologia , Receptores de Lipopolissacarídeos/imunologia , Malária Vivax/imunologia , Mitocôndrias/imunologia , Monócitos/imunologia , Plasmodium vivax/imunologia , Receptores de IgG/imunologia , Doença Aguda , Adolescente , Adulto , Idoso , Feminino , Citometria de Fluxo , Humanos , Imunofenotipagem , Malária Vivax/metabolismo , Malária Vivax/parasitologia , Masculino , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Monócitos/metabolismo , Monócitos/parasitologia , Fagocitose , Espécies Reativas de Oxigênio/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa