Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
Front Immunol ; 13: 906338, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35958580

RESUMO

Schistosomiasis is a disease of global significance, with severity and pathology directly related to how the host responds to infection. The immunological narrative of schistosomiasis has been constructed through decades of study, with researchers often focussing on isolated time points, cell types and tissue sites of interest. However, the field currently lacks a comprehensive and up-to-date understanding of the immune trajectory of schistosomiasis over infection and across multiple tissue sites. We have defined schistosome-elicited immune responses at several distinct stages of the parasite lifecycle, in three tissue sites affected by infection: the liver, spleen, and mesenteric lymph nodes. Additionally, by performing RNA-seq on the livers of schistosome infected mice, we have generated novel transcriptomic insight into the development of schistosome-associated liver pathology and fibrosis across the breadth of infection. Through depletion of CD11c+ cells during peak stages of schistosome-driven inflammation, we have revealed a critical role for CD11c+ cells in the co-ordination and regulation of Th2 inflammation during infection. Our data provide an updated and high-resolution account of how host immune responses evolve over the course of murine schistosomiasis, underscoring the significance of CD11c+ cells in dictating host immunopathology against this important helminth infection.


Assuntos
Esquistossomose mansoni , Esquistossomose , Animais , Antígeno CD11c , Imunidade , Inflamação , Camundongos , Schistosoma mansoni
3.
Immunohorizons ; 5(8): 721-732, 2021 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-34462311

RESUMO

Plasmacytoid dendritic cells (pDCs) are potent producers of type I IFN (IFN-I) during viral infection and respond to IFN-I in a positive feedback loop that promotes their function. IFN-I shapes dendritic cell responses during helminth infection, impacting their ability to support Th2 responses. However, the role of pDCs in type 2 inflammation is unclear. Previous studies have shown that pDCs are dispensable for hepatic or splenic Th2 responses during the early stages of murine infection with the trematode Schistosoma mansoni at the onset of parasite egg laying. However, during S. mansoni infection, an ongoing Th2 response against mature parasite eggs is required to protect the liver and intestine from acute damage and how pDCs participate in immune responses to eggs and adult worms in various tissues beyond acute infection remains unclear. We now show that pDCs are required for optimal Th2 cytokine production in response to S. mansoni eggs in the intestinal-draining mesenteric lymph nodes throughout infection and for egg-specific IFN-γ at later time points of infection. Further, pDC depletion at chronic stages of infection led to increased hepatic and splenic pathology as well as abrogated Th2 cell cytokine production and activation in the liver. In vitro, mesenteric lymph node pDCs supported Th2 cell responses from infection-experienced CD4+ T cells, a process dependent on pDC IFN-I responsiveness, yet independent of Ag. Together, these data highlight a previously unappreciated role for pDCs and IFN-I in maintaining and reinforcing type 2 immunity in the lymph nodes and inflamed tissue during helminth infection.


Assuntos
Citocinas/imunologia , Células Dendríticas/imunologia , Ativação Linfocitária/imunologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/parasitologia , Citocinas/metabolismo , Células Dendríticas/parasitologia , Feminino , Citometria de Fluxo/métodos , Interações Hospedeiro-Parasita/imunologia , Contagem de Linfócitos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Schistosoma mansoni/fisiologia , Esquistossomose mansoni/metabolismo , Esquistossomose mansoni/parasitologia , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/parasitologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/parasitologia
4.
Front Microbiol ; 11: 702, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32373101

RESUMO

Severe malaria anemia is one of the most common causes of morbidity and mortality arising from infection with Plasmodium falciparum. The pathogenesis of malarial anemia is complex, involving both parasite and host factors. As mouse models of malaria also develop anemia, they can provide a useful resource to study the impact of Plasmodium infections and the resulting host innate immune response on erythropoiesis. In this study, we have characterized the bone marrow and splenic responses of the erythroid as well as other hematopoietic lineages after an acute infection of Balb/c mice with Plasmodium berghei. Such characterization of the hematopoietic changes is critical to underpin future studies, using knockout mice and transgenic parasites, to tease out the interplay between host genes and parasite modulators implicated in susceptibility to malaria anemia. P. berghei infection led to a clear perturbation of steady-state erythropoiesis, with the most profound defects in polychromatic and orthochromatic erythroblasts as well as erythroid colony- and burst-forming units (CFU-E and BFU-E), resulting in an inability to compensate for anemia. The perturbation in erythropoiesis was not attributable to parasites infecting erythroblasts and affecting differentiation, nor to insufficient erythropoietin (EPO) production or impaired activation of the Signal transducer and activator of transcription 5 (STAT5) downstream of the EPO receptor, indicating EPO-signaling remained functional in anemia. Instead, the results point to acute anemia in P. berghei-infected mice arising from increased myeloid cell production in order to clear the infection, and the concomitant release of pro-inflammatory cytokines and chemokines from myeloid cells that inhibit erythroid development, in a manner that resembles the pathophysiology of anemia of chronic disease.

5.
Immunol Cell Biol ; 98(1): 42-53, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31559654

RESUMO

Methicillin-resistant Staphylococcus aureus (MRSA) presents an increasing threat to public health, with antimicrobial resistance on the rise and infections endemic in the hospital setting. Despite a global research effort to understand and combat antimicrobial resistance, less work has focused on understanding the nuances in the immunopathogenesis of clinical strains. In particular, there is a surprising gap of knowledge in the literature pertaining to how clinical strains are recognized by dendritic cells (DCs). Here, we show that the activation of DCs is compromised in response to MRSA strains resistant to the last-line antibiotic daptomycin. We found a significant reduction in the secretion of proinflammatory cytokines including tumor necrosis factor-α, interleukin-6, regulated upon activation, normal T cell expressed, and secreted and macrophage inflammatory protein-1ß, as well as decreased expression of CD80 by DCs responding to daptomycin-resistant MRSA. We further demonstrate that this phenotype is coincident with the acquisition of specific point mutations in the cardiolipin synthase gene cls2, and, partly, in the bifunctional lysylphosphatidylglycerol flippase/synthetase gene mprF, which are genes that are often mutated in clinical daptomycin-resistant strains. Therefore, throughout infection and antibiotic therapy, MRSA has the capacity to not only develop further antibiotic resistance, but also develop resistance to immunological recognition by DCs, because of single amino acid point mutations occurring under the selective pressures of both host immunity and antibiotic therapy. Understanding the diversity of clinical MRSA isolates and the nuances in their immune recognition will have important implications for future therapeutics and the treatment of these infections.


Assuntos
Daptomicina , Células Dendríticas/imunologia , Farmacorresistência Bacteriana/imunologia , Staphylococcus aureus Resistente à Meticilina/imunologia , Animais , Antígeno B7-1/imunologia , Citocinas/imunologia , Regulação da Expressão Gênica , Humanos , Staphylococcus aureus Resistente à Meticilina/isolamento & purificação , Camundongos
6.
Clin Transl Immunology ; 8(11): e01083, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31700626

RESUMO

This article summarises recent advances reported at the 9th Lorne Infection and Immunity Conference. This exciting conference hosted speakers in the fields of innate and adaptive responses to infection including host-pathogen interactions as well as novel strategies for the detection, control and treatment of infectious diseases such as fluorescent antibiotics and vaccine development. Host-pathogen studies focused on a broad range of pathogens including malaria, CMV, influenza, dengue and Zika viruses, listeria and tuberculosis.

7.
Front Immunol ; 10: 357, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30886619

RESUMO

Malaria remains a serious threat to global health. Sustained malaria control and, eventually, eradication will only be achieved with a broadly effective malaria vaccine. Yet a fundamental lack of knowledge about how antimalarial immunity is acquired has hindered vaccine development efforts to date. Understanding how malaria-causing parasites modulate the host immune system, specifically dendritic cells (DCs), key initiators of adaptive and vaccine antigen-based immune responses, is vital for effective vaccine design. This review comprehensively summarizes how exposure to Plasmodium spp. impacts human DC function in vivo and in vitro. We have highlighted the heterogeneity of the data observed in these studies, compared and critiqued the models used to generate our current understanding of DC function in malaria, and examined the mechanisms by which Plasmodium spp. mediate these effects. This review highlights potential research directions which could lead to improved efficacy of existing vaccines, and outlines novel targets for next-generation vaccine strategies to target malaria.


Assuntos
Antígenos de Protozoários/imunologia , Células Dendríticas/imunologia , Vacinas Antimaláricas/imunologia , Malária/imunologia , Plasmodium/imunologia , Humanos , Malária/prevenção & controle , Vacinas Antimaláricas/uso terapêutico
8.
Front Immunol ; 10: 32, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30766530

RESUMO

Dendritic cells are key linkers of innate and adaptive immunity. Efficient dendritic cell activation is central to the acquisition of immunity and the efficacy of vaccines. Understanding how dendritic cells are affected by Plasmodium falciparum blood-stage parasites will help to understand how immunity is acquired and maintained, and how vaccine responses may be impacted by malaria infection or exposure. This study investigates the response of dendritic cells to two different life stages of the malaria parasite, parasitized red blood cells and merozoites, using a murine model. We demonstrate that the dendritic cell responses to merozoites are robust whereas dendritic cell activation, particularly CD40 and pro-inflammatory cytokine expression, is compromised in the presence of freshly isolated parasitized red blood cells. The mechanism of dendritic cell suppression by parasitized red blood cells is host red cell membrane-independent. Furthermore, we show that cryopreserved parasitized red blood cells have a substantially reduced capacity for dendritic cell activation.


Assuntos
Células Dendríticas/imunologia , Interações Hospedeiro-Parasita/imunologia , Estágios do Ciclo de Vida/imunologia , Malária Falciparum/imunologia , Malária Falciparum/parasitologia , Plasmodium falciparum/imunologia , Biomarcadores , Citocinas/metabolismo , Células Dendríticas/metabolismo , Eritrócitos/imunologia , Eritrócitos/parasitologia , Humanos , Ligantes , Plasmodium falciparum/crescimento & desenvolvimento , Receptor Toll-Like 9/metabolismo
9.
Immunity ; 47(3): 498-509.e6, 2017 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-28916264

RESUMO

Double-stranded RNA (dsRNA) is a common by-product of viral infections and acts as a potent trigger of antiviral immunity. In the nematode C. elegans, sid-1 encodes a dsRNA transporter that is highly conserved throughout animal evolution, but the physiological role of SID-1 and its orthologs remains unclear. Here, we show that the mammalian SID-1 ortholog, SIDT2, is required to transport internalized extracellular dsRNA from endocytic compartments into the cytoplasm for immune activation. Sidt2-deficient mice exposed to extracellular dsRNA, encephalomyocarditis virus (EMCV), and herpes simplex virus 1 (HSV-1) show impaired production of antiviral cytokines and-in the case of EMCV and HSV-1-reduced survival. Thus, SIDT2 has retained the dsRNA transport activity of its C. elegans ortholog, and this transport is important for antiviral immunity.


Assuntos
Imunidade Inata , Proteínas de Membrana/metabolismo , Transporte de RNA , RNA de Cadeia Dupla/imunologia , RNA de Cadeia Dupla/metabolismo , Animais , Infecções por Cardiovirus/genética , Infecções por Cardiovirus/imunologia , Linhagem Celular , Citoplasma , Proteína DEAD-box 58/metabolismo , Modelos Animais de Doenças , Vírus da Encefalomiocardite/genética , Vírus da Encefalomiocardite/imunologia , Endossomos/metabolismo , Feminino , Expressão Gênica , Técnicas de Inativação de Genes , Herpes Simples/genética , Herpes Simples/imunologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/imunologia , Interações Hospedeiro-Patógeno/genética , Interações Hospedeiro-Patógeno/imunologia , Lisossomos/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Knockout , Proteínas de Transporte de Nucleotídeos , Ligação Proteica , Transporte Proteico , RNA Viral/genética , RNA Viral/metabolismo , Transdução de Sinais , Receptor 3 Toll-Like/metabolismo
10.
EMBO J ; 36(16): 2404-2418, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28716804

RESUMO

Type 2 inflammation is a defining feature of infection with parasitic worms (helminths), as well as being responsible for widespread suffering in allergies. However, the precise mechanisms involved in T helper (Th) 2 polarization by dendritic cells (DCs) are currently unclear. We have identified a previously unrecognized role for type I IFN (IFN-I) in enabling this process. An IFN-I signature was evident in DCs responding to the helminth Schistosoma mansoni or the allergen house dust mite (HDM). Further, IFN-I signaling was required for optimal DC phenotypic activation in response to helminth antigen (Ag), and efficient migration to, and localization with, T cells in the draining lymph node (dLN). Importantly, DCs generated from Ifnar1-/- mice were incapable of initiating Th2 responses in vivo These data demonstrate for the first time that the influence of IFN-I is not limited to antiviral or bacterial settings but also has a central role to play in DC initiation of Th2 responses.


Assuntos
Células Dendríticas/imunologia , Interferon Tipo I/metabolismo , Células Th2/imunologia , Alérgenos/imunologia , Animais , Camundongos , Camundongos Knockout , Pyroglyphidae/imunologia , Receptor de Interferon alfa e beta/deficiência , Schistosoma mansoni/imunologia
11.
Immunol Cell Biol ; 94(4): 400-10, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26657145

RESUMO

Dendritic cells (DCs) are the key initiators of T-helper (Th) 2 immune responses against the parasitic helminth Schistosoma mansoni. Although the liver is one of the main sites of antigen deposition during infection with this parasite, it is not yet clear how distinct DC subtypes in this tissue respond to S. mansoni antigens in vivo, or how the liver microenvironment might influence DC function during establishment of the Th2 response. In this study, we show that hepatic DC subsets undergo distinct activation processes in vivo following murine infection with S. mansoni. Conventional DCs (cDCs) from schistosome-infected mice upregulated expression of the costimulatory molecule CD40 and were capable of priming naive CD4(+) T cells, whereas plasmacytoid DCs (pDCs) upregulated expression of MHC class II, CD86 and CD40 but were unable to support the expansion of either naive or effector/memory CD4(+) T cells. Importantly, in vivo depletion of pDCs revealed that this subset was dispensable for either maintenance or regulation of the hepatic Th2 effector response during acute S. mansoni infection. Our data provides strong evidence that S. mansoni infection favors the establishment of an immunogenic, rather than tolerogenic, liver microenvironment that conditions cDCs to initiate and maintain Th2 immunity in the context of ongoing antigen exposure.


Assuntos
Células Dendríticas/imunologia , Fígado/imunologia , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia , Células Th2/imunologia , Animais , Antígenos de Helmintos/imunologia , Diferenciação Celular , Células Cultivadas , Células Dendríticas/parasitologia , Fígado/parasitologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL
12.
Nat Commun ; 6: 6920, 2015 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-25908537

RESUMO

Dendritic cells (DCs) direct CD4(+) T-cell differentiation into diverse helper (Th) subsets that are required for protection against varied infections. However, the mechanisms used by DCs to promote Th2 responses, which are important both for immunity to helminth infection and in allergic disease, are currently poorly understood. We demonstrate a key role for the protein methyl-CpG-binding domain-2 (Mbd2), which links DNA methylation to repressive chromatin structure, in regulating expression of a range of genes that are associated with optimal DC activation and function. In the absence of Mbd2, DCs display reduced phenotypic activation and a markedly impaired capacity to initiate Th2 immunity against helminths or allergens. These data identify an epigenetic mechanism that is central to the activation of CD4(+) T-cell responses by DCs, particularly in Th2 settings, and reveal methyl-CpG-binding proteins and the genes under their control as possible therapeutic targets for type-2 inflammation.


Assuntos
Proteínas de Ligação a DNA/imunologia , Células Dendríticas/imunologia , Regulação da Expressão Gênica/genética , RNA Mensageiro/metabolismo , Células Th2/imunologia , Alérgenos , Animais , Linfócitos T CD4-Positivos/imunologia , Polaridade Celular , Imunoprecipitação da Cromatina , Metilação de DNA , Proteínas de Ligação a DNA/genética , Ensaio de Imunoadsorção Enzimática , Epigênese Genética , Citometria de Fluxo , Hipersensibilidade/imunologia , Ativação Linfocitária/imunologia , Camundongos , Camundongos Knockout , Pyroglyphidae/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Schistosoma mansoni/imunologia , Esquistossomose mansoni/imunologia
13.
PLoS Negl Trop Dis ; 8(2): e2701, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24587461

RESUMO

BACKGROUND: MicroRNAs (miRNAs) are a class of short non-coding RNA that play important roles in disease processes in animals and are present in a highly stable cell-free form in body fluids. Here, we examine the capacity of host and parasite miRNAs to serve as tissue or serum biomarkers of Schistosoma mansoni infection. METHODS/PRINCIPAL FINDINGS: We used Exiqon miRNA microarrays to profile miRNA expression in the livers of mice infected with S. mansoni at 7 weeks post-infection. Thirty-three mouse miRNAs were differentially expressed in infected compared to naïve mice (>2 fold change, p<0.05) including miR-199a-3p, miR-199a-5p, miR-214 and miR-21, which have previously been associated with liver fibrosis in other settings. Five of the mouse miRNAs were also significantly elevated in serum by twelve weeks post-infection. Sequencing of small RNAs from serum confirmed the presence of these miRNAs and further revealed eleven parasite-derived miRNAs that were detectable by eight weeks post infection. Analysis of host and parasite miRNA abundance by qRT-PCR was extended to serum of patients from low and high infection sites in Zimbabwe and Uganda. The host-derived miRNAs failed to distinguish uninfected from infected individuals. However, analysis of three of the parasite-derived miRNAs (miR-277, miR-3479-3p and bantam) could detect infected individuals from low and high infection intensity sites with specificity/sensitivity values of 89%/80% and 80%/90%, respectively. CONCLUSIONS: This work identifies parasite-derived miRNAs as novel markers of S. mansoni infection in both mice and humans, with the potential to be used with existing techniques to improve S. mansoni diagnosis. In contrast, although host miRNAs are differentially expressed in the liver during infection their abundance levels in serum are variable in human patients and may be useful in cases of extreme pathology but likely hold limited value for detecting prevalence of infection.


Assuntos
Biomarcadores/sangue , MicroRNAs/sangue , RNA de Helmintos/sangue , Schistosoma mansoni/isolamento & purificação , Esquistossomose mansoni/sangue , Adolescente , Adulto , Animais , Criança , Pré-Escolar , Feminino , Humanos , Fígado/parasitologia , Fígado/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/química , MicroRNAs/metabolismo , Pessoa de Meia-Idade , Schistosoma mansoni/genética , Schistosoma mansoni/metabolismo , Esquistossomose mansoni/epidemiologia , Esquistossomose mansoni/parasitologia , Adulto Jovem
14.
EMBO J ; 33(6): 542-58, 2014 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-24514026

RESUMO

The sensing of nucleic acids by receptors of the innate immune system is a key component of antimicrobial immunity. RNA:DNA hybrids, as essential intracellular replication intermediates generated during infection, could therefore represent a class of previously uncharacterised pathogen-associated molecular patterns sensed by pattern recognition receptors. Here we establish that RNA:DNA hybrids containing viral-derived sequences efficiently induce pro-inflammatory cytokine and antiviral type I interferon production in dendritic cells. We demonstrate that MyD88-dependent signalling is essential for this cytokine response and identify TLR9 as a specific sensor of RNA:DNA hybrids. Hybrids therefore represent a novel molecular pattern sensed by the innate immune system and so could play an important role in host response to viruses and the pathogenesis of autoimmune disease.


Assuntos
Células Dendríticas/metabolismo , Imunidade Inata/imunologia , Modelos Imunológicos , Ácidos Nucleicos Heteroduplexes/metabolismo , Transdução de Sinais/imunologia , Receptor Toll-Like 9/metabolismo , Animais , Western Blotting , Células Dendríticas/imunologia , Endossomos , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Polarização de Fluorescência , Imunofluorescência , Humanos , Immunoblotting , Camundongos , Camundongos Endogâmicos C57BL , Fator 88 de Diferenciação Mieloide/imunologia , Ácidos Nucleicos Heteroduplexes/imunologia , Reação em Cadeia da Polimerase em Tempo Real , Receptor Toll-Like 9/imunologia
15.
Immunobiology ; 218(2): 263-71, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22704523

RESUMO

Cerebral malaria (CM) is the most severe syndrome associated with Plasmodium falciparum infections. Experimental evidence suggests that disease results from the sequestration of parasitized-red blood cells (pRBCs) together with inflammatory leukocytes within brain capillaries. We have previously shown that NK cells stimulate migration of CXCR3(+) T cells to the brain of Plasmodium berghei ANKA-infected mice. Here we investigated whether interactions between NK cells and dendritic cells (DCs) are required for the induction of T cell responses involved in disease. For that, NK cell-depleted and control mice were infected with transgenic parasites expressing model T cell epitopes. T cells from TCR transgenic mice specific for those epitopes were adoptively transferred and proliferation was determined. NK cell depletion significantly reduced CD8(+) but not CD4(+) DC-mediated T cell priming. Lack of NK cells did not compromise CD8(+) T cell responses in IL-12(-/-) mice, suggesting that NK cells stimulate IL-12 output by DCs required for optimal T cell priming. The contribution of DCs to NK cell function was also investigated. DC depletion and genetic deletion of IL-12 dramatically reduced NK cell-mediated IFN-γ responses to malaria. Thus NK cells and DCs engage in reciprocal activation for the induction of inflammatory responses involved in severe malaria.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Células Matadoras Naturais/imunologia , Malária Cerebral/imunologia , Plasmodium berghei/imunologia , Transferência Adotiva , Animais , Animais Geneticamente Modificados , Linfócitos T CD4-Positivos/transplante , Linfócitos T CD8-Positivos/transplante , Comunicação Celular , Células Cultivadas , Modelos Animais de Doenças , Epitopos de Linfócito T/genética , Humanos , Interferon gama/metabolismo , Interleucina-12/genética , Ativação Linfocitária/genética , Depleção Linfocítica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Plasmodium berghei/genética , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores CXCR3/metabolismo
16.
J Immunol ; 188(5): 2350-8, 2012 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-22287718

RESUMO

Infection with schistosome helminths is associated with granulomatous inflammation that forms around parasite eggs trapped in host tissues. In severe cases, the resulting fibrosis can lead to organ failure, portal hypertension, and fatal bleeding. Murine studies identified IL-17 as a critical mediator of this immunopathology, and mouse strains that produce high levels of IL-17 in response to schistosome infection show increased mortality. In this article, we demonstrate that schistosome-specific IL-17 induction by dendritic cells from low-pathology C57BL/6 mice is normally regulated by their concomitant induction of IL-10. Simultaneous stimulation of schistosome-exposed C57BL/6 dendritic cells with a heat-killed bacterium enabled these cells to overcome IL-10 regulation and induce IL-17, even in wild-type C57BL/6 recipients. This schistosome-specific IL-17 was dependent on IL-6 production by the copulsed dendritic cells. Coimmunization of C57BL/6 animals with bacterial and schistosome Ags also resulted in schistosome-specific IL-17, and this response was enhanced in the absence of IL-10-mediated immune regulation. Together, our data suggest that the balance of pro- and anti-inflammatory cytokines that determines the severity of pathology during schistosome infection can be influenced not only by host and parasite, but also by concurrent bacterial stimulation.


Assuntos
Antígenos de Helmintos/fisiologia , Células Dendríticas/imunologia , Interleucina-17/biossíntese , Propionibacterium acnes/imunologia , Transferência Adotiva , Animais , Antígenos de Helmintos/metabolismo , Células Cultivadas , Células Dendríticas/metabolismo , Células Dendríticas/transplante , Interleucina-10/metabolismo , Interleucina-10/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Schistosoma mansoni/imunologia , Esquistossomose/imunologia , Esquistossomose/metabolismo , Esquistossomose/patologia
17.
Curr Opin Immunol ; 23(1): 119-23, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20951016

RESUMO

Understanding the initiation of cellular immune responses during blood-stage malaria infection is essential for the development of an effective vaccine that improves upon the naturally acquired immune response and induces rapid and long-lasting protection against disease. Recent studies have identified the dendritic cell (DC) subtypes responsible for priming Plasmodium-specific T cells that mediate protection and/or pathology during blood-stage infection. Significant progress has also been made towards understanding DC recognition of Plasmodium parasites through engagement of TLR signalling pathways, as well as the potential for non-TLR ligands to mediate Plasmodium-induced suppression of DC antigen presentation.


Assuntos
Apresentação de Antígeno , Malária/imunologia , Plasmodium/imunologia , Animais , Apresentação Cruzada , Células Dendríticas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Humanos , Malária/parasitologia
18.
J Exp Med ; 207(10): 2089-96, 2010 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-20819926

RESUMO

Although dendritic cells (DCs) are adept initiators of CD4(+) T cell responses, their fundamental importance in this regard in Th2 settings remains to be demonstrated. We have used CD11c-diphtheria toxin (DTx) receptor mice to deplete CD11c(+) cells during the priming stage of the CD4(+) Th2 response against the parasitic helminth Schistosoma mansoni. DTx treatment significantly depleted CD11c(+) DCs from all tissues tested, with 70-80% efficacy. Even this incomplete depletion resulted in dramatically impaired CD4(+) T cell production of Th2 cytokines, altering the balance of the immune response and causing a shift toward IFN-γ production. In contrast, basophil depletion using Mar-1 antibody had no measurable effect on Th2 induction in this system. These data underline the vital role that CD11c(+) antigen-presenting cells can play in orchestrating Th2 development against helminth infection in vivo, a response that is ordinarily balanced so as to prevent the potentially damaging production of inflammatory cytokines.


Assuntos
Apresentação de Antígeno , Antígeno CD11c/imunologia , Células Dendríticas/imunologia , Esquistossomose mansoni/imunologia , Células Th2/imunologia , Animais , Basófilos/imunologia , Antígeno CD11c/genética , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/imunologia , Interferon gama/biossíntese , Interferon gama/imunologia , Procedimentos de Redução de Leucócitos , Ativação Linfocitária , Camundongos , Schistosoma mansoni/imunologia
19.
J Immunol ; 185(6): 3632-42, 2010 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-20720206

RESUMO

Cerebral malaria is a severe complication of malaria. Sequestration of parasitized RBCs in brain microvasculature is associated with disease pathogenesis, but our understanding of this process is incomplete. In this study, we examined parasite tissue sequestration in an experimental model of cerebral malaria (ECM). We show that a rapid increase in parasite biomass is strongly associated with the induction of ECM, mediated by IFN-gamma and lymphotoxin alpha, whereas TNF and IL-10 limit this process. Crucially, we discovered that host CD4(+) and CD8(+) T cells promote parasite accumulation in vital organs, including the brain. Modulation of CD4(+) T cell responses by helminth coinfection amplified CD4(+) T cell-mediated parasite sequestration, whereas vaccination could generate CD4(+) T cells that reduced parasite biomass and prevented ECM. These findings provide novel insights into immune-mediated mechanisms of ECM pathogenesis and highlight the potential of T cells to both prevent and promote infectious diseases.


Assuntos
Malária Cerebral/imunologia , Malária Cerebral/parasitologia , Plasmodium berghei/imunologia , Animais , Encéfalo/irrigação sanguínea , Encéfalo/imunologia , Encéfalo/parasitologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/parasitologia , Linfócitos T CD4-Positivos/patologia , Modelos Animais de Doenças , Eritrócitos/imunologia , Eritrócitos/parasitologia , Eritrócitos/patologia , Feminino , Trato Gastrointestinal/irrigação sanguínea , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/parasitologia , Rim/irrigação sanguínea , Rim/imunologia , Rim/parasitologia , Fígado/irrigação sanguínea , Fígado/imunologia , Fígado/parasitologia , Pulmão/irrigação sanguínea , Pulmão/imunologia , Pulmão/parasitologia , Malária Cerebral/sangue , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Camundongos Transgênicos , Especificidade de Órgãos/imunologia , Plasmodium berghei/crescimento & desenvolvimento , Índice de Gravidade de Doença , Baço/irrigação sanguínea , Baço/imunologia , Baço/parasitologia
20.
Eur J Immunol ; 40(6): 1674-81, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20391433

RESUMO

Despite extensive evidence that Plasmodium species are capable of stimulating the immune system, the association of malaria with a higher incidence of other infectious diseases and reduced responses to vaccination against unrelated pathogens suggests the existence of immune suppression. Recently, we provided evidence that blood-stage Plasmodium berghei infection leads to suppression of MHC class I-restricted immunity to third party (non-malarial) antigens as a consequence of systemic DC activation. This earlier study did not, however, determine whether reactivity was also impaired to MHC class II-restricted third party antigens or to Plasmodium antigens themselves. Here, we show that while P. berghei-expressed antigens were presented early in infection, there was a rapid decline in presentation within 4 days, paralleling impairment in MHC class I- and II-restricted presentation of third party antigens. This provides important evidence that P. berghei not only causes immunosuppression to subsequently encountered third party antigens, but also rapidly limits the capacity to generate effective parasite-specific immunity.


Assuntos
Apresentação de Antígeno/imunologia , Células Dendríticas/imunologia , Tolerância Imunológica/imunologia , Malária/imunologia , Animais , Antígenos de Protozoários/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Parasitos/imunologia , Plasmodium berghei/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...