Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Immunity ; 45(4): 831-846, 2016 10 18.
Artigo em Inglês | MEDLINE | ID: mdl-27760339

RESUMO

T follicular helper (Tfh) cells are a CD4+ T cell subset critical for long-lived humoral immunity. We hypothesized that integrins play a decisive role in Tfh cell biology. Here we show that Tfh cells expressed a highly active form of leukocyte function-associated antigen-1 (LFA-1) that was required for their survival within the germinal center niche. In addition, LFA-1 promoted expression of Bcl-6, a transcriptional repressor critical for Tfh cell differentiation, and inhibition of LFA-1 abolished Tfh cell generation and prevented protective humoral immunity to intestinal helminth infection. Furthermore, we demonstrated that expression of Talin-1, an adaptor protein that regulates LFA-1 affinity, dictated Tfh versus Th2 effector cell differentiation. Collectively, our results define unique functions for LFA-1 in the Tfh cell effector program and suggest that integrin activity is important in lineage decision-making events in the adaptive immune system.


Assuntos
Diferenciação Celular/imunologia , Ativação Linfocitária/imunologia , Antígeno-1 Associado à Função Linfocitária/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Células Cultivadas , Centro Germinativo/imunologia , Humanos , Imunidade Humoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Proto-Oncogênicas c-bcl-6/imunologia
2.
PLoS Pathog ; 13(10): e1006647, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28968468

RESUMO

Interferon regulatory factor-8 (IRF-8) is critical for Th1 cell differentiation and negatively regulates myeloid cell development including myeloid-derived suppressor cells (MDSC). MDSC expand during infection with various pathogens including the gastrointestinal (GI) nematode Heligmosomoides polygyrus bakeri (Hpb). We investigated if IRF-8 contributes to Th2 immunity to Hpb infection. Irf8 expression was down-regulated in MDSC from Hpb-infected C57BL/6 (B6) mice. IRF-8 deficient Irf8-/- and BXH-2 mice had significantly higher adult worm burdens than B6 mice after primary or challenge Hpb infection. During primary infection, MDSC expanded to a significantly greater extent in mesenteric lymph nodes (MLN) and spleens of Irf8-/- and BXH-2 than B6 mice. CD4+GATA3+ T cells numbers were comparable in MLN of infected B6 and IRF-8 deficient mice, but MLN cells from infected IRF-8 deficient mice secreted significantly less parasite-specific IL-4 ex vivo. The numbers of alternatively activated macrophages in MLN and serum levels of Hpb-specific IgG1 and IgE were also significantly less in infected Irf8-/- than B6 mice. The frequencies of antigen-experienced CD4+CD11ahiCD49dhi cells that were CD44hiCD62L- were similar in MLN of infected Irf8-/- and B6 mice, but the proportions of CD4+GATA3+ and CD4+IL-4+ T cells were lower in infected Irf8-/- mice. CD11b+Gr1+ cells from naïve or infected Irf8-/- mice suppressed CD4+ T cell proliferation and parasite-specific IL-4 secretion in vitro albeit less efficiently than B6 mice. Surprisingly, there were significantly more CD4+ T cells in infected Irf8-/- mice, with a higher frequency of CD4+CD25+Foxp3+ T (Tregs) cells and significantly higher numbers of Tregs than B6 mice. In vivo depletion of MDSC and/or Tregs in Irf8-/- mice did not affect adult worm burdens, but Treg depletion resulted in higher egg production and enhanced parasite-specific IL-5, IL-13, and IL-6 secretion ex vivo. Our data thus provide a previously unrecognized role for IRF-8 in Th2 immunity to a GI nematode.


Assuntos
Gastroenteropatias/imunologia , Fatores Reguladores de Interferon/imunologia , Células Supressoras Mieloides/imunologia , Infecções por Nematoides/imunologia , Nematospiroides dubius/imunologia , Células Th2/imunologia , Animais , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Fatores Reguladores de Interferon/efeitos dos fármacos , Fatores Reguladores de Interferon/genética , Interleucina-4/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Reguladores/imunologia
3.
J Immunol ; 197(7): 2948-57, 2016 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-27559049

RESUMO

Helminthic infections modulate host immunity and may protect people in less-developed countries from developing immunological diseases. In a murine colitis model, the helminth Heligmosomoides polygyrus bakeri prevents colitis via induction of regulatory dendritic cells (DCs). The mechanism driving the development of these regulatory DCs is unexplored. There is decreased expression of the intracellular signaling pathway spleen tyrosine kinase (Syk) in intestinal DCs from H. polygyrus bakeri-infected mice. To explore the importance of this observation, it was shown that intestinal DCs from DC-specific Syk(-/-) mice were powerful inhibitors of murine colitis, suggesting that loss of Syk was sufficient to convert these cells into their regulatory phenotype. DCs sense gut flora and damaged epithelium via expression of C-type lectin receptors, many of which signal through the Syk signaling pathway. It was observed that gut DCs express mRNA encoding for C-type lectin (CLEC) 7A, CLEC9A, CLEC12A, and CLEC4N. H. polygyrus bakeri infection downmodulated CLEC mRNA expression in these cells. Focusing on CLEC7A, which encodes for the dectin-1 receptor, flow analysis showed that H. polygyrus bakeri decreases dectin-1 expression on the intestinal DC subsets that drive Th1/Th17 development. DCs become unresponsive to the dectin-1 agonist curdlan and fail to phosphorylate Syk after agonist stimulation. Soluble worm products can block CLEC7A and Syk mRNA expression in gut DCs from uninfected mice after a brief in vitro exposure. Thus, downmodulation of Syk expression and phosphorylation in intestinal DCs could be important mechanisms through which helminths induce regulatory DCs that limit colitis.


Assuntos
Colite/prevenção & controle , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Intestinos/citologia , Transdução de Sinais , Quinase Syk/metabolismo , Animais , Colite/imunologia , Colite/parasitologia , Modelos Animais de Doenças , Intestinos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Nematospiroides dubius/imunologia , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Infecções por Strongylida/imunologia , Infecções por Strongylida/parasitologia , Quinase Syk/deficiência , Quinase Syk/genética
4.
Exp Parasitol ; 178: 30-36, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28533110

RESUMO

To better control gastrointestinal nematode infections in humans and animals, it is important to understand the strategies used by these parasites to modulate the host immune system. In this regard, molecules released by parasites have been attributed crucially important roles in host-parasite negotiations. We characterized the excretory/secretory (E/S) microRNA (miRNA) and protein profiles from the mouse gastrointestinal nematode parasite Trichuris muris. Released miRNAs were subjected to miRNA sequencing and E/S proteins were analysed by mass spectrometry. Fourteen miRNAs were identified in T. muris exosome-like vesicles, as well as 73 proteins of nematode origin, 11 of which were unique to this study. Comparison with published nematode protein secretomes revealed high conservation at the functional level.


Assuntos
Exossomos/química , Proteínas de Helminto/análise , MicroRNAs/isolamento & purificação , Trichuris/metabolismo , Animais , Meios de Cultura/química , Proteínas de Helminto/genética , Proteínas de Helminto/imunologia , Proteínas de Helminto/fisiologia , Proteômica , Reprodutibilidade dos Testes , Trichuris/genética , Trichuris/imunologia
5.
Malar J ; 15(1): 260, 2016 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-27150250

RESUMO

BACKGROUND: The potential emergence and spread of resistance to artemisinins in the Plasmodium falciparum malaria parasite constitutes a major global health threat. Hence, improving the efficacy of artemisinins and of artemisinin-based combination therapy (ACT) represents a major short-term goal in the global fight against malaria. Mice defective in the enzyme pantetheinase (Vnn3) show increased susceptibility to blood-stage malaria (increased parasitaemia, reduced survival), and supplementation of Vnn3 mutants with the reaction product of pantetheinase, cysteamine, corrects in part the malaria-susceptibility phenotype of the mutants. Cysteamine (Cys) is a small, naturally occurring amino-thiol that has very low toxicity in vivo and is approved for clinical use in the life-long treatment of the kidney disorder nephropathic cystinosis. METHODS: The ability of Cys to improve the anti-plasmodial activity of different clinically used artemisinins was tested. The effect of different CYS/ART combinations on malarial phenotypes (parasite blood-stage replication, overall and survival from lethal infection) was assessed in a series of in vivo experiments using Plasmodium strains that induce either blood-stage (Plasmodium chabaudi AS) or cerebral disease (Plasmodium berghei ANKA). This was also evaluated in an ex vivo experimental protocol that directly assesses the effect of such drug combinations on the viability of Plasmodium parasites, as measured by the ability of tested parasites to induce a productive infection in vivo in otherwise naïve animals. RESULTS: Cys is found to potentiate the anti-plasmodial activity of artesunate, artemether, and arteether, towards the blood-stage malaria parasite P. chabaudi AS. Ex vivo experiments, indicate that potentiation of the anti-plasmodial activity of artemisinins by Cys is direct and does not require the presence of host factors. In addition, potentiation occurs at sub-optimal concentrations of artemisinins and Cys that on their own have little or no effect on parasite growth. Cys also dramatically enhances the efficacy and protective effect of artemisinins against cerebral malaria induced by infection with the P. berghei ANKA parasite. CONCLUSION: These findings indicate that inclusion of Cys in current formulations of ACT, or its use as adjunct therapy could improve the anti-plasmodial activity of artemisinin, decrease mortality in cerebral malaria patients, and prevent or delay the development and spread of artemisinin resistance.


Assuntos
Antimaláricos/administração & dosagem , Artemisininas/administração & dosagem , Cisteamina/administração & dosagem , Sinergismo Farmacológico , Malária/tratamento farmacológico , Animais , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Quimioterapia Combinada , Feminino , Malária Cerebral/tratamento farmacológico , Camundongos , Plasmodium berghei/efeitos dos fármacos , Plasmodium berghei/fisiologia , Plasmodium chabaudi/efeitos dos fármacos , Plasmodium chabaudi/fisiologia , Análise de Sobrevida , Resultado do Tratamento
6.
PLoS Pathog ; 9(7): e1003491, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23853600

RESUMO

Interferon Regulatory Factor 8 (IRF8) is required for development, maturation and expression of anti-microbial defenses of myeloid cells. BXH2 mice harbor a severely hypomorphic allele at Irf8 (Irf8(R294C)) that causes susceptibility to infection with intracellular pathogens including Mycobacterium tuberculosis. We report that BXH2 are completely resistant to the development of cerebral malaria (ECM) following Plasmodium berghei ANKA infection. Comparative transcriptional profiling of brain RNA as well as chromatin immunoprecipitation and high-throughput sequencing (ChIP-seq) was used to identify IRF8-regulated genes whose expression is associated with pathological acute neuroinflammation. Genes increased by infection were strongly enriched for IRF8 binding sites, suggesting that IRF8 acts as a transcriptional activator in inflammatory programs. These lists were enriched for myeloid-specific pathways, including interferon responses, antigen presentation and Th1 polarizing cytokines. We show that inactivation of several of these downstream target genes (including the Irf8 transcription partner Irf1) confers protection against ECM. ECM-resistance in Irf8 and Irf1 mutants is associated with impaired myeloid and lymphoid cells function, including production of IL12p40 and IFNγ. We note strong overlap between genes bound and regulated by IRF8 during ECM and genes regulated in the lungs of M. tuberculosis infected mice. This IRF8-dependent network contains several genes recently identified as risk factors in acute and chronic human inflammatory conditions. We report a common core of IRF8-bound genes forming a critical inflammatory host-response network.


Assuntos
Encéfalo/imunologia , Regulação da Expressão Gênica , Imunidade Inata , Fatores Reguladores de Interferon/metabolismo , Malária Cerebral/imunologia , Proteínas do Tecido Nervoso/metabolismo , Plasmodium berghei/imunologia , Substituição de Aminoácidos , Animais , Sítios de Ligação , Encéfalo/metabolismo , Encéfalo/parasitologia , Células Cultivadas , Citocinas/biossíntese , Citocinas/sangue , Perfilação da Expressão Gênica , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Fatores Reguladores de Interferon/química , Fatores Reguladores de Interferon/genética , Malária Cerebral/sangue , Malária Cerebral/metabolismo , Malária Cerebral/parasitologia , Camundongos , Camundongos Knockout , Camundongos Mutantes , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Proteínas do Tecido Nervoso/química , Proteínas do Tecido Nervoso/genética , Neurônios/imunologia , Neurônios/metabolismo , Neurônios/parasitologia , Organismos Livres de Patógenos Específicos , Baço/imunologia , Baço/metabolismo , Baço/patologia , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/parasitologia
7.
J Infect Dis ; 209(1): 140-9, 2014 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-23922378

RESUMO

Low reticulocytosis, indicating reduced red blood cell (RBC) output, is an important feature of severe malarial anemia. Evidence supports a role for Plasmodium products, especially hemozoin (Hz), in suppressed erythropoiesis during malaria, but the mechanism(s) involved remains unclear. Here, we demonstrated that low reticulocytosis and suppressed erythropoietin (Epo)-induced erythropoiesis are features of malarial anemia in Plasmodium yoelii- and Plasmodium berghei ANKA-infected mice, similar to our previous observations in Plasmodium chabaudi AS-infected mice. The magnitude of decreases in RBC was a reflection of parasitemia level, but low reticulocytosis was evident despite differences in parasitemia, clinical manifestation, and infection outcome. Schizont extracts and Hz from P. falciparum and P. yoelii and synthetic Hz suppressed Epo-induced proliferation of erythroid precursors in vitro but did not inhibit RBC maturation. To determine whether Hz contributes to malarial anemia, P. yoelii-derived or synthetic Hz was administered to naive mice, and the development of anemia, reticulocytosis, and RBC turnover was determined. Parasite-derived Hz induced significant decreases in RBC and increased RBC turnover with compensatory reticulocytosis, but anemia was not as severe as that in infected mice. Our findings suggest that parasite factors, including Hz, contribute to severe malarial anemia by suppressing Epo-induced proliferation of erythroid precursors.


Assuntos
Anemia/parasitologia , Eritropoese/fisiologia , Hemeproteínas/farmacologia , Malária/sangue , Reticulocitose/fisiologia , Análise de Variância , Anemia/sangue , Anemia/metabolismo , Animais , Contagem de Eritrócitos , Eritropoese/efeitos dos fármacos , Feminino , Macrófagos/química , Macrófagos/parasitologia , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/química , Monócitos/parasitologia , Plasmodium , Reticulocitose/efeitos dos fármacos , Esquizontes/fisiologia
8.
J Exp Med ; 204(3): 511-24, 2007 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-17312006

RESUMO

Mouse strains AcB55 and AcB61 are resistant to malaria by virtue of a mutation in erythrocyte pyruvate kinase (Pklr(I90N)). Linkage analysis in [AcB55 x A/J] F2 mice detected a second locus (Char9; logarithm of odds = 4.74) that regulates the blood-stage replication of Plasmodium chabaudi AS independently of Pklr. We characterized the 77 genes of the Char9 locus for tissue-specific expression, strain-specific alterations in gene expression, and polymorphic variants that are possibly associated with differential susceptibility. We identified Vnn1/Vnn3 as the likely candidates responsible for Char9. Vnn3/Vnn1 map within a conserved haplotype block and show expression levels that are strictly cis-regulated by this haplotype. The absence of Vnn messenger RNA expression and lack of pantetheinase protein activity in tissues are associated with susceptibility to malaria and are linked to a complex rearrangement in the Vnn3 promoter region. The A/J strain also carries a unique nonsense mutation that leads to a truncated protein. Vanin genes code for a pantetheinase involved in the production of cysteamine, a key regulator of host responses to inflammatory stimuli. Administration of cystamine in vivo partially corrects susceptibility to malaria in A/J mice, as measured by reduced blood parasitemia and decreased mortality. These studies suggest that pantetheinase is critical for the host response to malaria.


Assuntos
Moléculas de Adesão Celular/genética , Clonagem Molecular , Predisposição Genética para Doença , Hidrolases/genética , Malária/enzimologia , Malária/genética , Amidoidrolases/deficiência , Amidoidrolases/genética , Amidoidrolases/fisiologia , Animais , Mapeamento Cromossômico , Proteínas Ligadas por GPI , Marcadores Genéticos , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos , Plasmodium chabaudi/imunologia , Locos de Características Quantitativas
9.
J Immunol ; 186(8): 4862-71, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21389253

RESUMO

To investigate the role of CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells in blood-stage malaria, we compared Plasmodium chabaudi AS infection in wild-type (WT) C57BL/6 and transgenic mice overexpressing the transcription factor Foxp3 (Foxp3Tg) and observed that Foxp3Tg mice experienced lethal infection and deficient malaria-specific immune responses. Adoptive transfer of total CD4(+) T cells from Foxp3Tg mice or CD4(+)CD25(+) T cells from WT mice to naive WT recipients confirmed that high numbers of Treg cells compromised immune control of malaria. Transfer of GFP(+)CD4(+)CD25(+) T cells to naive WT recipients together with immunohistochemical staining of spleens from infected WT mice demonstrated that Foxp3(+) Treg cells localized in the T cell area of the spleen. Determination of CD4(+)Foxp3(+) Treg cell responses in the spleen of infected WT mice revealed a significant but transient increase in CD4(+)Foxp3(+) Treg cells early in infection. This was followed by a significant and sustained decrease due to reduced proliferation and apoptosis of CD4(+)Foxp3(+) Treg cells. Importantly, the kinetics of IL-2 secretion by effector CD4(+)Foxp3(-) T cells coincided with changes in CD4(+)Foxp3(+) cells and the differentiation of CD4(+)T-bet(+)IFN-γ(+) cells required for immune control of infection. Administration of the IL-2/anti-IL-2 mAb (clone JES6-1) complex to infected WT mice increased the severity of P. chabaudi AS infection and promoted expansion of Foxp3(+) Treg cells. Collectively, these data demonstrate that the ability to control and eliminate P. chabaudi AS infection is due to a tight balance between natural Treg cells and effector CD4(+) Th1 cells, a balance regulated in part by IL-2.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Fatores de Transcrição Forkhead/imunologia , Interleucina-2/imunologia , Malária/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva , Animais , Anticorpos Monoclonais/imunologia , Anticorpos Monoclonais/farmacologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD4-Positivos/transplante , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Interações Hospedeiro-Parasita/imunologia , Imuno-Histoquímica , Interferon gama/sangue , Interleucina-10/sangue , Interleucina-2/metabolismo , Subunidade alfa de Receptor de Interleucina-2/imunologia , Subunidade alfa de Receptor de Interleucina-2/metabolismo , Contagem de Linfócitos , Malária/sangue , Malária/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Plasmodium chabaudi/efeitos dos fármacos , Plasmodium chabaudi/imunologia , Plasmodium chabaudi/fisiologia , Baço/imunologia , Baço/metabolismo , Baço/parasitologia , Linfócitos T Reguladores/metabolismo , Linfócitos T Reguladores/transplante
10.
Nat Genet ; 35(4): 357-62, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14595440

RESUMO

The global health impact of malaria is enormous, with an estimated 300-500 million clinical cases and 1 million annual deaths. In humans, initial susceptibility to infection with Plasmodium species, disease severity and ultimate outcome of malaria (self-healing or lethal) are under complex genetic control. Alleles associated with sickle cell anemia, beta-thalassemia and deficiency in glucose-6-phosphate dehydrogenase have a protective effect against malaria and may have been retained by positive selection in areas of endemic malaria. Likewise, genetic variations in erythrocyte antigens and levels of host cytokines affect type and severity of disease. A mouse model of infection with Plasmodium chabaudi was used to study the genetic component of malaria susceptibility. Segregation analyses in informative F2 crosses derived from resistant C57BL/6J and susceptible A/J, C3H and SJL strains using extent of blood stage replication of the parasite and survival as traits mapped three P. chabaudi resistance (Char) loci on chromosomes 9 (Char1), 8 (Char2) and 17 (Char3, MHC-linked). Recombinant congenic strains AcB55 and AcB61 are unusually resistant to malaria despite carrying susceptibility alleles at Char1 and Char2. Malaria resistance in AcB55 and AcB61 is associated with splenomegaly and constitutive reticulocytosis, is inherited in an autosomal recessive fashion and is controlled by a locus on chromosome 3 (Char4). Sequencing of candidate genes from the Char4 region identified a loss-of-function mutation (269T-->A, resulting in the amino acid substitution I90N) in the pyruvate kinase gene (Pklr) that underlies the malaria resistance in AcB55 and AcB61. These results suggest that pyruvate kinase deficiency may similarly protect humans against malaria.


Assuntos
Predisposição Genética para Doença/genética , Malária/genética , Plasmodium chabaudi/fisiologia , Piruvato Quinase/deficiência , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sequência de Bases , Mapeamento Cromossômico , Eritrócitos/parasitologia , Feminino , Perfilação da Expressão Gênica , Ligação Genética , Imunidade Inata/genética , Malária/enzimologia , Malária/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Dados de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , Parasitemia , Plasmodium chabaudi/crescimento & desenvolvimento , Piruvato Quinase/genética , Homologia de Sequência do Ácido Nucleico , Esplenomegalia
11.
J Immunol ; 185(9): 5495-502, 2010 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-20876354

RESUMO

Pathogen sensing by the inflammasome activates inflammatory caspases that mediate inflammation and cell death. Caspase-12 antagonizes the inflammasome and NF-κB and is associated with susceptibility to bacterial sepsis. A single-nucleotide polymorphism (T(125)C) in human Casp12 restricts its expression to Africa, Southeast Asia, and South America. Here, we investigated the role of caspase-12 in the control of parasite replication and pathogenesis in malaria and report that caspase-12 dampened parasite clearance in blood-stage malaria and modulated susceptibility to cerebral malaria. This response was independent of the caspase-1 inflammasome, as casp1(-/-) mice were indistinguishable from wild-type animals in response to malaria, but dependent on enhanced NF-κB activation. Mechanistically, caspase-12 competed with NEMO for association with IκB kinase-α/ß, effectively preventing the formation of the IκB kinase complex and inhibiting downstream transcriptional activation by NF-κB. Systemic inhibition of NF-κB or Ab neutralization of IFN-γ reversed the increased resistance of casp12(-/-) mice to blood-stage malaria infection.


Assuntos
Caspase 12/imunologia , Inflamação/imunologia , Malária/imunologia , NF-kappa B/imunologia , Transdução de Sinais/imunologia , Animais , Caspase 12/genética , Citocinas/biossíntese , Citocinas/imunologia , Ativação Enzimática/imunologia , Ensaio de Imunoadsorção Enzimática , Feminino , Predisposição Genética para Doença , Humanos , Inflamação/genética , Malária/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética
12.
Front Immunol ; 13: 874308, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35757733

RESUMO

Infection with helminths or parasitic worms are highly prevalent worldwide especially in developing regions. Helminths cause chronic infections that are associated with suppression of immune responses to unrelated pathogens, vaccines, and by-stander antigens responsible for dysregulated immune responses as occurs in diseases such as allergies. Helminths use multiple mechanisms to modulate the immune system to evade the highly polarized type 2 immune response required to expel adult worms and for immunity to reinfection. Anthelmintic drugs are efficient in reducing adult worm burdens in helminth-infected individuals, but resistance to these drugs is rapidly increasing and vaccines against these pathogens are not available. Emerging evidence indicate that helminths induce myeloid-derived suppressor cells (MDSC), originally described in tumor-bearing mice and cancer patients. MDSC are a heterogenous population of immature cells that consist of two distinct sub-populations, polymorphonuclear (PMN)-MDSC and monocytic (M)-MDSC based on morphology and phenotype. MDSC suppress the function of T cells and other innate and adaptive immune cells including NK cells and B cells. During cancer or infection with bacteria or viruses, there is marked expansion of MDSC. Furthermore, the frequencies of MDSC correlate inversely with the prognosis and survival of tumor-bearing hosts as well as bacterial and viral burdens, persistence, and outcome in infected hosts. Currently, there is a paucity of data on MDSC and helminth infections. Here, we provide a survey of the evidence accumulated so far that overall support a role for MDSC in modulating immune responses during helminth infections. We review data from studies in various helminths, including those that infect humans. Finally, we summarize the progress to date in understanding the role of MDSC in helminth infections and briefly discuss potential host-directed strategies to target MDSC-mediated suppression of immune responses to helminths in favor of development of immunity to eliminate adult worms and possibly induce protection against reinfection.


Assuntos
Helmintos , Células Supressoras Mieloides , Neoplasias , Vacinas , Animais , Humanos , Camundongos , Reinfecção , Linfócitos T
13.
Exp Parasitol ; 125(4): 315-24, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20219464

RESUMO

In mice, loss of pantetheinase activity causes susceptibility to infection with Plasmodium chabaudi AS. Treatment of mice with the pantetheinase metabolite cysteamine reduces blood-stage replication of P. chabaudi and significantly increases survival. Similarly, a short exposure of Plasmodium to cysteamine ex vivo is sufficient to suppress parasite infectivity in vivo. This effect of cysteamine is specific and not observed with a related thiol (dimercaptosuccinic acid) or with the pantethine precursor of cysteamine. Also, cysteamine does not protect against infection with the parasite Trypanosoma cruzi or the fungal pathogen Candida albicans, suggesting cysteamine acts directly against the parasite and does not modulate host inflammatory response. Cysteamine exposure also blocks replication of P. falciparum in vitro; moreover, these treated parasites show higher levels of intact hemoglobin. This study highlights the in vivo action of cysteamine against Plasmodium and provides further evidence for the involvement of pantetheinase in host response to this infection.


Assuntos
Antimaláricos/farmacologia , Cisteamina/farmacologia , Malária/tratamento farmacológico , Plasmodium chabaudi/efeitos dos fármacos , Plasmodium falciparum/efeitos dos fármacos , Amidoidrolases/metabolismo , Animais , Antimaláricos/uso terapêutico , Candidíase/tratamento farmacológico , Doença de Chagas/tratamento farmacológico , Cloroquina/farmacologia , Cisteamina/uso terapêutico , Citocinas/sangue , Citocinas/efeitos dos fármacos , Relação Dose-Resposta a Droga , Eritrócitos/efeitos dos fármacos , Eritrócitos/parasitologia , Feminino , Proteínas Ligadas por GPI , Hemoglobinas/metabolismo , Humanos , Malária/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Parasitemia/tratamento farmacológico , Parasitemia/parasitologia , Plasmodium falciparum/metabolismo , Trypanosoma cruzi/efeitos dos fármacos
15.
Infect Immun ; 77(2): 770-82, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19015248

RESUMO

Dendritic cells (DCs) are important accessory cells for promoting NK cell gamma interferon (IFN-gamma) production in vitro in response to Plasmodium falciparum-infected red blood cells (iRBC). We investigated the requirements for reciprocal activation of DCs and NK cells leading to Th1-type innate and adaptive immunity to P. chabaudi AS infection. During the first week of infection, the uptake of iRBC by splenic CD11c(+) DCs in resistant wild-type (WT) C57BL/6 mice was similar to that in interleukin 15(-/-) (IL-15(-/-)) and IL-12p40(-/-) mice, which differ in the severity of P. chabaudi AS infection. DCs from infected IL-15(-/-) mice expressed costimulatory molecules, produced IL-12, and promoted IFN-gamma secretion by WT NK cells in vitro as efficiently as WT DCs. In contrast, DCs from infected IL-12p40(-/-) mice exhibited alterations in maturation and cytokine production and were unable to induce NK cell IFN-gamma production. Coculture of DCs and NK cells demonstrated that DC-mediated NK cell activation required IL-12 and, to a lesser extent, IL-2, as well as cell-cell contact. In turn, NK cells from infected WT mice enhanced DC maturation, IL-12 production, and priming of CD4(+) T-cell proliferation and IFN-gamma secretion. Infected WT mice depleted of NK cells, which exhibit increased parasitemia, had impaired DC maturation and DC-induced CD4(+) Th1 cell priming. These findings indicate that DC-NK cell reciprocal cross talk is critical for control and rapid resolution of P. chabaudi AS infection and provide in vivo evidence for the importance of this interaction in IFN-gamma-dependent immunity to malaria.


Assuntos
Comunicação Celular/fisiologia , Células Dendríticas/fisiologia , Interferon gama/metabolismo , Células Matadoras Naturais/fisiologia , Malária/imunologia , Plasmodium chabaudi/imunologia , Animais , Citocinas/genética , Citocinas/metabolismo , Regulação da Expressão Gênica , Malária/parasitologia , Camundongos , Camundongos Knockout
16.
Haematologica ; 94(2): 195-204, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19109218

RESUMO

BACKGROUND: The contribution of pro-inflammatory cytokines to the pathogenesis of malarial anemia has been studied extensively but the roles of Th2 cytokines remain unknown. Here, we investigated the role of signal transducer and activator of transcription (STAT)6-mediated responses in erythropoietic suppression during acute malaria infection in mice. DESIGN AND METHODS: Naïve and/or erythropoietin-treated wild-type and STAT6(-/-) mice were infected with Plasmodium chabaudi AS (P. chabaudi), and the effects parasitemia, hematologic parameters, erythropoietin receptor, TER119, and CD71 expression, in vitro erythropoietin-stimulated proliferation of splenic erythroid precursors, and serum cytokine levels were analyzed. To explore the role of interleukin-4 in STAT6-dependent erythropoietic suppression, mice were treated in vivo with a monoclonal antibody to interleukin-4 and the effects on parasitemia, hematologic parameters, and cytokine levels were analyzed. RESULTS: Infected STAT6(-/-) mice developed enhanced reticulocytosis compared to wild-type mice despite higher parasitemia and a similar course of anemia. Enhanced reticulocytosis in infected STAT6(-/-) mice was associated with an increased frequency of late-stage erythroblasts, fewer leukocytes expressing CD71, and increased erythropoietin-stimulated proliferation of splenocytes compared to infected wild-type mice. Interleukin-4-depleted wild-type mice had increased levels of parasitemia and a course of reticulocytosis similar to responses observed in infected STAT6(-/-) mice. Determination of serum cytokine levels in STAT6(-/-) and wild-type mice depleted of interleukin-4 by treatment with mAb revealed significantly lower levels of interferon-gamma compared to control wild-type mice during infection. CONCLUSIONS: Together, these findings provide evidence for a STAT6-dependent mechanism in mediating erythropoietic suppression during acute blood-stage malaria and indicate a role for interleukin-4 and possibly interferon-gammain STAT6-induced erythropoietic suppression.


Assuntos
Anemia/sangue , Eritropoese , Malária/sangue , Fator de Transcrição STAT6/fisiologia , Animais , Modelos Animais de Doenças , Interferon gama , Interleucina-4 , Camundongos , Células Th2/imunologia
17.
Front Immunol ; 10: 425, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30915078

RESUMO

Control and elimination of blood-stage Plasmodium chabaudi AS infection requires CD4+ Th1 cells that secrete IFN-γ and T follicular help (Tfh) cells together with B cell production of antibody. Foxp3+ regulatory T cells (Tregs) are also crucial to protect the host from immunopathology and severe disease, but these cells can suppress protective immune responses to malaria. The chemokine receptor CXCR3 expressed by activated T cells is important for trafficking of CD4+ Th1 cells to sites of inflammation and infection. Previous studies demonstrated CXCR3 is expressed on CD4+ T cells in the spleen during malaria, but the phenotype was not defined. We identified the phenotype of CD4+ T cells that expressed CXCR3 in C57BL/6 (B6) mice during acute P. chabaudi AS infection by analyzing expression of the transcription factors T-bet and Foxp3. We also investigated if CXCR3 contributes to control of parasite replication and survival. The frequency and number of CD4+CXCR3+ T cells increased dramatically in the spleen of infected B6 mice coincident with increased CD4+IFN-γ+ T cells. CXCR3 was up-regulated on effector CD4+Foxp3- T cells as well as Foxp3+ Tregs. Consistent with our previous observations, CD4+T-bet+Foxp3- T cells increased in B6 mice during acute infection. T-bet+Foxp3+ Tregs also increased significantly and a high frequency of these cells expressed CXCR3 supporting the notion that these cells may be Th1-like Tregs. Despite this, the percentage of CD4+Foxp3+ Tregs from infected B6 mice that migrated in vitro to the CXCR3 ligands CXCL9 and CXCL10 was significantly less than naïve mice. To investigate the in vivo contribution of CXCR3 to control of acute blood-stage malaria, we compared the course and outcome of P. chabaudi AS infection in wild-type (WT) B6 and CXCR3-deficient mice. Parasitemia levels were significantly higher around the time of peak parasitemia in CXCR3-/- compared to WT mice but survival was similar suggesting a role for CXCR3 in controlling parasite replication during acute P. chabaudi AS infection. Together, our findings indicate Th1-like CD4+T-bet+Foxp3+ Tregs that express CXCR3 are induced during acute blood-stage malaria and suggest CXCR3 expression on CD4+ Th1 cells may contribute to their migration to the spleen.


Assuntos
Malária/imunologia , Plasmodium chabaudi , Receptores CXCR3/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia , Animais , Movimento Celular , Feminino , Fatores de Transcrição Forkhead/imunologia , Ligantes , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores CXCR3/genética , Baço/imunologia , Proteínas com Domínio T/imunologia
18.
J Inorg Biochem ; 200: 110808, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31487576

RESUMO

In malaria, Plasmodium parasites produce hemozoin (Hz) as a route to detoxify free heme released from the catabolism of hemoglobin. Hz isolated from the parasites is encapsulated in an organic layer constituted by parasite and host components. This organic coating may play a role in Hz formation and in the immunomodulatory properties attributed to Hz, and they may influence the mode of action of antimalarials that block Hz formation. In this work, we analyze the organic layer adhered to Hz, and find Na, Cl, Si, Ca and P present, in addition to organic material. Our results suggest that Na, Cl, and P adsorb during Hz release from the red blood cells, while Si and Ca derive from components present during Hz biomineralization within the digestive vacuole of the parasite. Overall, we show that inorganic elements associated with Hz surface provide insights into the biological functions of Plasmodium parasites.


Assuntos
Eritrócitos , Hemeproteínas/metabolismo , Plasmodium chabaudi/metabolismo , Plasmodium falciparum/metabolismo , Animais , Eritrócitos/metabolismo , Eritrócitos/parasitologia , Humanos , Íons/farmacologia , Camundongos
19.
Sci Rep ; 8(1): 15921, 2018 10 29.
Artigo em Inglês | MEDLINE | ID: mdl-30374177

RESUMO

Parasitic worms have a remarkable ability to modulate host immune responses through several mechanisms including excreted/secreted proteins (ESP), yet the exact nature of these proteins and their targets often remains elusive. Here, we performed mass spectrometry analyses of ESP (TsESP) from larval and adult stages of the pig whipworm Trichuris suis (Ts) and identified ~350 proteins. Transcriptomic analyses revealed large subsets of differentially expressed genes in the various life cycle stages of the parasite. Exposure of bone marrow-derived macrophages and dendritic cells to TsESP markedly diminished secretion of the pro-inflammatory cytokines TNFα and IL-12p70. Conversely, TsESP exposure strongly induced release of the anti-inflammatory cytokine IL-10, and also induced high levels of nitric oxide (NO) and upregulated arginase activity in macrophages. Interestingly, TsESP failed to directly induce CD4+ CD25+ FoxP3+ regulatory T cells (Treg cells), while OVA-pulsed TsESP-treated dendritic cells suppressed antigen-specific OT-II CD4+ T cell proliferation. Fractionation of TsESP identified a subset of proteins that promoted anti-inflammatory functions, an activity that was recapitulated using recombinant T. suis triosephosphate isomerase (TPI) and nucleoside diphosphate kinase (NDK). Our study helps illuminate the intricate balance that is characteristic of parasite-host interactions at the immunological interface, and further establishes the principle that specific parasite-derived proteins can modulate immune cell functions.


Assuntos
Proteínas de Helminto/metabolismo , Trichuris/metabolismo , Animais , Arginase/metabolismo , Citocinas/metabolismo , Células Dendríticas/citologia , Células Dendríticas/metabolismo , Estágios do Ciclo de Vida , Macrófagos/citologia , Macrófagos/metabolismo , Óxido Nítrico/metabolismo , Suínos/parasitologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Trichuris/crescimento & desenvolvimento
20.
Novartis Found Symp ; 281: 156-65; discussion 165-8, 208-9, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17534072

RESUMO

The onset, progression and outcome of infections are determined by performance of host defence mechanisms and expression of pathogen virulence determinants. Genetic analysis in mouse can identify host genes that play critical roles at the interface of host-pathogen interactions. Genetic effects detected as variations in susceptibility in inbred, recombinant and mutant strains of mice can be mapped as simple traits or quantitative trait loci followed by identification by positional cloning. We have used mouse models of infection with bacterial (Mycobacterium, Legionella) and parasitic pathogens (Plasmodium) to discover genes and proteins that are important for macrophage function against such infectious agents. These studies have identified Nrampl-mediated exclusion of divalent metals from the phagosomal space as a key regulator of intracellular replication of Mycobacteria. Also, intracellular sensing of Legionella by functional Birc1e/Naip5 protein is essential to prevent replication of this bacterium in macrophages. Finally, we have identified two new loci that affect blood-stage replication of Plasmodium chabaudi AS in mice, and have cloned the corresponding genes.


Assuntos
Infecções Bacterianas/genética , Predisposição Genética para Doença/genética , Macrófagos/imunologia , Doenças Parasitárias/genética , Animais , Infecções Bacterianas/imunologia , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/imunologia , Clonagem Molecular , Legionella/imunologia , Camundongos , Mycobacteriaceae/imunologia , Proteína Inibidora de Apoptose Neuronal/genética , Proteína Inibidora de Apoptose Neuronal/imunologia , Doenças Parasitárias/imunologia , Plasmodium/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA