Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
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
2.
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
3.
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
4.
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
5.
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
6.
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
7.
PLoS One ; 12(5): e0177818, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28542307

RESUMO

Pyruvate kinase (PKLR) deficiency protects mice and humans against blood-stage malaria. Although mouse strain AcB62 carries a malaria-protective PklrI90N genetic mutation, it is phenotypically susceptible to blood stage malaria induced by infection with Plasmodium chabaudi AS, suggesting a genetic modifier of the PklrI90N protective effect. Linkage analysis in a F2 cross between AcB62 (PklrI90N) and another PK deficient strain CBA/Pk (PklrG338D) maps this modifier (designated Char10) to chromosome 9 (LOD = 10.8, 95% Bayesian CI = 50.7-75Mb). To study the mechanistic basis of the Char10 effect, we generated an incipient congenic line (Char10C) that harbors the Char10 chromosome 9 segment from AcB62 fixed on the genetic background of CBA/Pk. The Char10 effect is shown to be highly penetrant as the Char10C line recapitulates the AcB62 phenotype, displaying high parasitemia following P. chabaudi infection, compared to CBA/Pk. Char10C mice also display a reduction in anemia phenotypes associated with the PklrG338D mutation including decreased splenomegaly, decreased circulating reticulocytes, increased density of mature erythrocytes, increased hematocrit, as well as decreased iron overload in kidney and liver and decreased serum iron. Erythroid lineage analyses indicate that the number of total TER119+ cells as well as the numbers of the different CD71+/CD44+ erythroblast sub-populations were all found to be lower in Char10C spleen compared to CBA/Pk. Char10C mice also displayed lower number of CFU-E per spleen compared to CBA/Pk. Taken together, these results indicate that the Char10 locus modulates the severity of pyruvate kinase deficiency by regulating erythroid responses in the presence of PK-deficiency associated haemolytic anemia.


Assuntos
Anemia Hemolítica Congênita não Esferocítica/genética , Cromossomos de Mamíferos/genética , Loci Gênicos/genética , Predisposição Genética para Doença/genética , Malária/genética , Piruvato Quinase/deficiência , Erros Inatos do Metabolismo dos Piruvatos/genética , Anemia Hemolítica Congênita não Esferocítica/metabolismo , Anemia Hemolítica Congênita não Esferocítica/fisiopatologia , Animais , Eritrócitos/metabolismo , Eritrócitos/patologia , Eritropoese/genética , Humanos , Ferro/metabolismo , Camundongos , Piruvato Quinase/genética , Piruvato Quinase/metabolismo , Erros Inatos do Metabolismo dos Piruvatos/metabolismo , Erros Inatos do Metabolismo dos Piruvatos/fisiopatologia
8.
Acta Biomater ; 48: 247-257, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27769943

RESUMO

Mucoadhesive drug delivery systems stick to mucosal tissues and prolong the local retention time of drugs. Since the colon is covered by a mucosal layer, mucoadhesive rectal formulations may improve treatment of such diseases as hypertension or colon cancer. Ulcerative colitis (UC) is an inflammatory bowel disease characterized by chronic inflammation of the colonic mucosa. It is commonly treated with sulfasalazine (SSZ), which is metabolized by the intestinal flora into the therapeutic 5-aminosalicylic acid (5-ASA) and a toxic by-product sulfapyridine (SP). SSZ can be administered orally or rectally. The latter route avoids unintended absorption of the drug or its degradation products in the upper gastrointestinal tract, but often fails due to limited retention time. Here, we propose a mucoadhesive hydrogel to improve the efficacy of rectal SSZ administration. The gel is made of catechol modified-chitosan (Cat-CS) crosslinked by genipin. After loading the gel with SSZ, we evaluated its efficacy in a mouse model of UC. Compared to oral SSZ treatment, rectal SSZ/Cat-CS delivery was more therapeutic, showed equivalent histological scores, and induced a lower plasma concentration of the potentially toxic SP by-product. These results show SSZ/Cat-CS rectal hydrogels are more effective and safer formulations for UC treatment than oral SSZ. STATEMENT OF SIGNIFICANCE: Ulcerative colitis affects the colon by causing chronic inflammation on the mucosa. One of the most common drugs to treat mild to moderate UC is sulfasalazine, which can be administrated both orally and rectally. Rectal formulations are preferable, since their therapeutic effect happens topically, and they prevent side effects related to absorption of the drug in the small intestine. However, the efficacy of rectal sulfasalazine formulations is decreased by their limited colon residence time. Here we propose a chitosan-catechol mucoadhesive gel that allows delivering sulfasalazine more effectively and safely than oral administration. Our results bring new insights into the field of mussel-inspired catechol hydrogels, showing their potential as drug delivery systems to treat a widespread disease such as ulcerative colitis.


Assuntos
Adesivos/química , Quitosana/química , Colite Ulcerativa/tratamento farmacológico , Sistemas de Liberação de Medicamentos , Hidrogéis/química , Mucinas/química , Reto/efeitos dos fármacos , Animais , Peso Corporal/efeitos dos fármacos , Catecóis/química , Colite Ulcerativa/sangue , Colite Ulcerativa/patologia , Colo/efeitos dos fármacos , Colo/patologia , Relação Dose-Resposta a Droga , Camundongos , Sangue Oculto , Sulfassalazina/sangue , Sulfassalazina/metabolismo , Sulfassalazina/farmacologia , Sulfassalazina/uso terapêutico , Fator de Necrose Tumoral alfa/metabolismo
9.
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
10.
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
11.
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
12.
PLoS Negl Trop Dis ; 10(4): e0004624, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27058578

RESUMO

Helminth infections affect more than a third of the world's population. Despite very broad phylogenetic differences among helminth parasite species, a systemic Th2 host immune response is typically associated with long-term helminth infections, also known as the "helminth effect". Many investigations have been carried out to study host gene expression profiles during helminth infections. The objective of this study is to determine if there is a common transcriptomic signature characteristic of the helminth effect across multiple helminth species and tissue types. To this end, we performed a comprehensive meta-analysis of publicly available gene expression datasets. After data processing and adjusting for study-specific effects, we identified ~700 differentially expressed genes that are changed consistently during helminth infections. Functional enrichment analyses indicate that upregulated genes are predominantly involved in various immune functions, including immunomodulation, immune signaling, inflammation, pathogen recognition and antigen presentation. Down-regulated genes are mainly involved in metabolic process, with only a few of them are involved in immune regulation. This common immune gene signature confirms previous observations and indicates that the helminth effect is robust across different parasite species as well as host tissue types. To the best of our knowledge, this study is the first comprehensive meta-analysis of host transcriptome profiles during helminth infections.


Assuntos
Perfilação da Expressão Gênica , Helmintíase Animal/imunologia , Helmintos/imunologia , Animais , Camundongos
13.
Nat Protoc ; 9(12): 2740-54, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25375989

RESUMO

Heligmosomoides polygyrus bakeri (Hpb) infection in mice is a convenient model for studying the pathophysiology and immunology of gastrointestinal (GI) helminth infection. Hpb infection suppresses immune responses to bystander antigens and unrelated pathogens, and it slows the progression and modifies the outcome of immune-mediated diseases. Hpb-derived excretory-secretory (ES) products potently modulate CD4(+) helper T cell (TH) responses by inducing regulatory T cells, tolerogenic dendritic cells (DCs) and immunoregulatory cytokines. This observation has spiked interest in identifying the immunomodulatory molecules, especially proteins, in ES products from Hpb and other GI nematodes for development as novel therapies to treat individuals with immune-mediated diseases, such as inflammatory bowel diseases (IBDs). In this protocol, we describe how to (i) maintain Hpb in the laboratory for experimental infections, (ii) collect adult worms from infected mice to generate ES products and (iii) evaluate the modulatory effects of ES products on toll-like receptor (TLR) ligand-induced maturation of CD11c(+) DCs. The three major sections of the PROCEDURE can be used independently, and they require ∼6, 10 and 27 h, respectively. Although other methods use a modified Baermann apparatus to collect Hpb adult worms, we describe a method that involves dissection of adult worms from intestinal tissue. The protocol will be useful to investigators studying the host-parasite interface and identifying and analyzing helminth-derived molecules with therapeutic potential.


Assuntos
Fatores Imunológicos/farmacologia , Nematospiroides dubius/imunologia , Nematospiroides dubius/isolamento & purificação , Infecções por Strongylida/imunologia , Animais , Antígeno CD11c/metabolismo , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Fezes/parasitologia , Feminino , Interações Hospedeiro-Parasita/imunologia , Fatores Imunológicos/isolamento & purificação , Intestinos/parasitologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nematospiroides dubius/fisiologia , Infecções por Strongylida/parasitologia
14.
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
15.
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
16.
PLoS Negl Trop Dis ; 5(10): e1370, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22039562

RESUMO

The murine parasite Heligmosomoides polygyrus is a convenient experimental model to study immune responses and pathology associated with gastrointestinal nematode infections. The excretory-secretory products (ESP) produced by this parasite have potent immunomodulatory activity, but the protein(s) responsible has not been defined. Identification of the protein composition of ESP derived from H. polygyrus and other relevant nematode species has been hampered by the lack of genomic sequence information required for proteomic analysis based on database searches. To overcome this, a transcriptome next generation sequencing (RNA-seq) de novo assembly containing 33,641 transcripts was generated, annotated, and used to interrogate mass spectrometry (MS) data derived from 1D-SDS PAGE and LC-MS/MS analysis of ESP. Using the database generated from the 6 open reading frames deduced from the RNA-seq assembly and conventional identification programs, 209 proteins were identified in ESP including homologues of vitellogenins, retinol- and fatty acid-binding proteins, globins, and the allergen V5/Tpx-1-related family of proteins. Several potential immunomodulators, such as macrophage migration inhibitory factor, cysteine protease inhibitors, galectins, C-type lectins, peroxiredoxin, and glutathione S-transferase, were also identified. Comparative analysis of protein annotations based on the RNA-seq assembly and proteomics revealed processes and proteins that may contribute to the functional specialization of ESP, including proteins involved in signalling pathways and in nutrient transport and/or uptake. Together, these findings provide important information that will help to illuminate molecular, biochemical, and in particular immunomodulatory aspects of host-H. polygyrus biology. In addition, the methods and analyses presented here are applicable to study biochemical and molecular aspects of the host-parasite relationship in species for which sequence information is not available.


Assuntos
Perfilação da Expressão Gênica , Proteínas de Helminto/análise , Nematospiroides dubius/química , Nematospiroides dubius/genética , Proteoma/análise , Animais , Cromatografia Líquida , Eletroforese em Gel de Poliacrilamida , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência de DNA , Espectrometria de Massas em Tandem
17.
Int J Biol Sci ; 7(9): 1311-22, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22110383

RESUMO

Although a clearer understanding of the underlying mechanisms involved in protection and immunopathology during blood-stage malaria has emerged, the mechanisms involved in regulating the adaptive immune response especially those required to maintain a balance between beneficial and deleterious responses remain unclear. Recent evidence suggests the importance of CD11c⁺ dendritic cells (DC) and CD4⁺Foxp3⁺ regulatory T cells in regulating immune responses during infection and autoimmune disease, but information concerning the contribution of these cells to regulating immunity to malaria is limited. Here, we review recent findings from our laboratory and others in experimental models of malaria in mice and in Plasmodium-infected humans on the roles of DC and natural regulatory T cells in regulating adaptive immunity to blood-stage malaria.


Assuntos
Células Dendríticas/imunologia , Malária/imunologia , Linfócitos T Reguladores/imunologia , Imunidade Adaptativa/imunologia , Animais , Células Dendríticas/metabolismo , Humanos , Malária/metabolismo , Plasmodium/imunologia , Plasmodium/patogenicidade , Linfócitos T Reguladores/metabolismo
18.
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
19.
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
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...