Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
mBio ; : e0052724, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39302131

RESUMO

Glycosylphosphatidylinositols (GPIs) are highly conserved anchors for eukaryotic cell surface proteins. The apicomplexan parasite, Toxoplasma gondii, is a widespread intracellular parasite of warm-blooded animals whose plasma membrane is covered with GPI-anchored proteins, and free GPIs called GIPLs. While the glycan portion is conserved, species differ in sidechains added to the triple mannose core. The functional significance of the Glcα1,4GalNAcß1- sidechain reported in Toxoplasma gondii has remained largely unknown without understanding its biosynthesis. Here we identify and disrupt two glycosyltransferase genes and confirm their respective roles by serology and mass spectrometry. Parasites lacking the sidechain on account of deletion of the first glycosyltransferase, PIGJ, exhibit increased virulence during primary and secondary infections, suggesting it is an important pathogenesis factor. Cytokine responses, antibody recognition of GPI-anchored SAGs, and complement binding to PIGJ mutants are intact. By contrast, the scavenger receptor CD36 shows enhanced binding to PIGJ mutants, potentially explaining a subtle tropism for macrophages detected early in infection. Galectin-3, which binds GIPLs, exhibits an enhancement of binding to PIGJ mutants, and the protection of galectin-3 knockout mice from lethality suggests that Δpigj parasite virulence in this context is sidechain dependent. Parasite numbers are not affected by Δpigj early in the infection in wild-type mice, suggesting a breakdown of tolerance. However, increased tissue cysts in the brains of mice infected with Δpigj parasites indicate an advantage over wild-type strains. Thus, the GPI sidechain of T. gondii plays a crucial and diverse role in regulating disease outcomes in the infected host.IMPORTANCEThe functional significance of sidechain modifications to the glycosylphosphatidylinositol (GPI) anchor in parasites has yet to be determined because the glycosyltransferases responsible for these modifications have not been identified. Here we present identification and characterization of both Toxoplasmsa gondii GPI sidechain-modifying glycosyltransferases. Removal of the glycosyltransferase that adds the first GalNAc to the sidechain results in parasites without a sidechain on the GPI, and increased host susceptibility to infection. Loss of the second glycosyltransferase results in a sidechain with GalNAc alone, and no glucose added, and has negligible effect on disease outcomes. This indicates GPI sidechains are fundamental to host-parasite interactions.

2.
Trends Parasitol ; 40(6): 449-451, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38762372

RESUMO

Polymeric guanylate-binding proteins (GBPs) physically dismember the vacuole membrane formed by Toxoplasma gondii while nitric oxide (NO) poisons and inhibits parasite replication within interferon (IFN)-γ activated macrophages. Zhao et al. report a novel mechanism for synergy between these classical microbicidal and microbistatic effectors in cell-autonomous immunity to the intracellular parasites.


Assuntos
Toxoplasma , Toxoplasma/imunologia , Óxido Nítrico/metabolismo , Animais , Humanos , Proteínas de Ligação ao GTP/imunologia , Proteínas de Ligação ao GTP/metabolismo , Macrófagos/imunologia , Macrófagos/parasitologia
3.
Trends Parasitol ; 39(1): 10-11, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36470783

RESUMO

Toxoplasma gondii exploits the migratory properties of monocytes and dendritic cells to promote tissue dissemination. Recently, ten Hoeve et al. reported that the parasite effector protein GRA28 conspires with host chromatin modifiers to confer dendritic cell-like features that convert sessile macrophages into migratory cells that transport infection to distal organs.


Assuntos
Toxoplasma , Macrófagos/parasitologia , Células Dendríticas/parasitologia
4.
Infect Immun ; 90(8): e0020522, 2022 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-35913173

RESUMO

The role of specific host cell surface receptors during Toxoplasma gondii invasion of host cells is poorly defined. Here, we interrogated the role of the well-known malarial invasion receptor, basigin, in T. gondii infection of astrocytes. We found that primary astrocytes express two members of the BASIGIN (BSG) immunoglobulin family, basigin and embigin, but did not express neuroplastin. Antibody blockade of either basigin or embigin caused a significant reduction of parasite infectivity in astrocytes. The specific role of basigin during T. gondii invasion was further examined using a mouse astrocytic cell line (C8-D30), which exclusively expresses basigin. CRISPR-mediated deletion of basigin in C8-D30 cells resulted in decreased T. gondii infectivity. T. gondii replication and invasion efficiency were not altered by basigin deficiency, but parasite attachment to astrocytes was markedly reduced. We also conducted a proteomic screen to identify T. gondii proteins that interact with basigin. Toxoplasma-encoded cyclophilins, the protein 14-3-3, and protein disulfide isomerase (TgPDI) were among the putative basigin-ligands identified. Recombinant TgPDI produced in E. coli bound to basigin and pretreatment of tachyzoites with a PDI inhibitor decreased parasite attachment to host cells. Finally, mutagenesis of the active site cysteines of TgPDI abolished enzyme binding to basigin. Thus, basigin and its related immunoglobulin family members may represent host receptors that mediate attachment of T. gondii to diverse cell types.


Assuntos
Toxoplasma , Toxoplasmose , Basigina , Escherichia coli , Humanos , Proteômica
5.
Cell Rep ; 40(5): 111150, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35926464

RESUMO

Intestinal nematode parasites can cross the epithelial barrier, causing tissue damage and release of danger-associated molecular patterns (DAMPs) that may promote host protective type 2 immunity. We investigate whether adenosine binding to the A2B adenosine receptor (A2BAR) on intestinal epithelial cells (IECs) plays an important role. Specific blockade of IEC A2BAR inhibits the host protective memory response to the enteric helminth, Heligmosomoides polygyrus bakeri (Hpb), including disruption of granuloma development at the host-parasite interface. Memory T cell development is blocked during the primary response, and transcriptional analyses reveal profound impairment of IEC activation. Extracellular ATP is visualized 24 h after inoculation and is shown in CD39-deficient mice to be critical for the adenosine production mediating the initiation of type 2 immunity. Our studies indicate a potent adenosine-mediated IEC pathway that, along with the tuft cell circuit, is critical for the activation of type 2 immunity.


Assuntos
Adenosina , Receptor A2B de Adenosina , Adenosina/metabolismo , Trifosfato de Adenosina , Animais , Células Epiteliais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptor A2B de Adenosina/metabolismo
6.
Immunity ; 53(2): 398-416.e8, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32814028

RESUMO

Paneth cells are the primary source of C-type lysozyme, a ß-1,4-N-acetylmuramoylhydrolase that enzymatically processes bacterial cell walls. Paneth cells are normally present in human cecum and ascending colon, but are rarely found in descending colon and rectum; Paneth cell metaplasia in this region and aberrant lysozyme production are hallmarks of inflammatory bowel disease (IBD) pathology. Here, we examined the impact of aberrant lysozyme production in colonic inflammation. Targeted disruption of Paneth cell lysozyme (Lyz1) protected mice from experimental colitis. Lyz1-deficiency diminished intestinal immune responses to bacterial molecular patterns and resulted in the expansion of lysozyme-sensitive mucolytic bacteria, including Ruminococcus gnavus, a Crohn's disease-associated pathobiont. Ectopic lysozyme production in colonic epithelium suppressed lysozyme-sensitive bacteria and exacerbated colitis. Transfer of R. gnavus into Lyz1-/- hosts elicited a type 2 immune response, causing epithelial reprograming and enhanced anti-colitogenic capacity. In contrast, in lysozyme-intact hosts, processed R. gnavus drove pro-inflammatory responses. Thus, Paneth cell lysozyme balances intestinal anti- and pro-inflammatory responses, with implications for IBD.


Assuntos
Clostridiales/imunologia , Colite Ulcerativa/patologia , Muramidase/genética , Muramidase/metabolismo , Celulas de Paneth/metabolismo , Animais , Clostridiales/genética , Colite Ulcerativa/microbiologia , Doença de Crohn/patologia , Feminino , Microbioma Gastrointestinal/genética , Células Caliciformes/citologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição STAT6/genética
7.
PLoS Pathog ; 16(5): e1008579, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32421753

RESUMO

Anti-helminth responses require robust type 2 cytokine production that simultaneously promotes worm expulsion and initiates the resolution of helminth-induced wounds and hemorrhaging. However, how infection-induced changes in hematopoiesis contribute to these seemingly distinct processes remains unknown. Recent studies have suggested the existence of a hematopoietic progenitor with dual mast cell-erythrocyte potential. Nonetheless, whether and how these progenitors contribute to host protection during an active infection remains to be defined. Here, we employed single cell RNA-sequencing and identified that the metabolic enzyme, carbonic anhydrase (Car) 1 marks a predefined bone marrow-resident hematopoietic progenitor cell (HPC) population. Next, we generated a Car1-reporter mouse model and found that Car1-GFP positive progenitors represent bipotent mast cell/erythrocyte precursors. Finally, we show that Car1-expressing HPCs simultaneously support mast cell and erythrocyte responses during Trichinella spiralis infection. Collectively, these data suggest that mast cell/erythrocyte precursors are mobilized to promote type 2 cytokine responses and alleviate helminth-induced blood loss, developmentally linking these processes. Collectively, these studies reveal unappreciated hematopoietic events initiated by the host to combat helminth parasites and provide insight into the evolutionary pressure that may have shaped the developmental relationship between mast cells and erythrocytes.


Assuntos
Células Precursoras Eritroides/imunologia , Eritropoese/imunologia , Mastócitos/imunologia , Mastocitose/imunologia , Trichinella spiralis/imunologia , Triquinelose/imunologia , Animais , Anidrase Carbônica I/genética , Anidrase Carbônica I/imunologia , Células Precursoras Eritroides/parasitologia , Células Precursoras Eritroides/patologia , Feminino , Mastócitos/parasitologia , Mastócitos/patologia , Mastocitose/genética , Mastocitose/patologia , Camundongos , Camundongos Transgênicos , Triquinelose/genética , Triquinelose/patologia
8.
J Exp Med ; 215(9): 2265-2278, 2018 09 03.
Artigo em Inglês | MEDLINE | ID: mdl-30087163

RESUMO

An IRF8-dependent subset of conventional dendritic cells (cDCs), termed cDC1, effectively cross-primes CD8+ T cells and facilitates tumor-specific T cell responses. Etv6 is an ETS family transcription factor that controls hematopoietic stem and progenitor cell (HSPC) function and thrombopoiesis. We report that like HSPCs, cDCs express Etv6, but not its antagonist, ETS1, whereas interferon-producing plasmacytoid dendritic cells (pDCs) express both factors. Deletion of Etv6 in the bone marrow impaired the generation of cDC1-like cells in vitro and abolished the expression of signature marker CD8α on cDC1 in vivo. Moreover, Etv6-deficient primary cDC1 showed a partial reduction of cDC-specific and cDC1-specific gene expression and chromatin signatures and an aberrant up-regulation of pDC-specific signatures. Accordingly, DC-specific Etv6 deletion impaired CD8+ T cell cross-priming and the generation of tumor antigen-specific CD8+ T cells. Thus, Etv6 optimizes the resolution of cDC1 and pDC expression programs and the functional fitness of cDC1, thereby facilitating T cell cross-priming and tumor-specific responses.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade Celular , Neoplasias/imunologia , Proteínas Proto-Oncogênicas c-ets/imunologia , Proteínas Repressoras/imunologia , Animais , Antígenos de Neoplasias/genética , Antígenos CD8/genética , Antígenos CD8/imunologia , Linfócitos T CD8-Positivos/patologia , Células Dendríticas/patologia , Deleção de Genes , Células-Tronco Hematopoéticas/imunologia , Células-Tronco Hematopoéticas/patologia , Camundongos , Camundongos Knockout , Neoplasias/genética , Neoplasias/patologia , Proteína Proto-Oncogênica c-ets-1/genética , Proteína Proto-Oncogênica c-ets-1/imunologia , Proteínas Proto-Oncogênicas c-ets/genética , Proteínas Repressoras/genética , Trombopoese/genética , Trombopoese/imunologia , Variante 6 da Proteína do Fator de Translocação ETS
9.
Cell Stem Cell ; 23(1): 46-59.e5, 2018 Jul 05.
Artigo em Inglês | MEDLINE | ID: mdl-29887318

RESUMO

Paneth cells are post-mitotic intestinal epithelial cells supporting the stem cell niche and mucosal immunity. Paneth cell pathologies are observed in various gastrointestinal diseases, but their plasticity and response to genomic and environmental challenges remain unclear. Using a knockin allele engineered at the mouse Lyz1 locus, we performed detailed Paneth cell-lineage tracing. Irradiation induced a subset of Paneth cells to proliferate and differentiate into villus epithelial cells. RNA sequencing (RNA-seq) revealed that Paneth cells sorted from irradiated mice acquired a stem cell-like transcriptome; when cultured in vitro, these individual Paneth cells formed organoids. Irradiation activated Notch signaling, and forced expression of Notch intracellular domain (NICD) in Paneth cells, but not Wnt/ß-catenin pathway activation, induced their dedifferentiation. This study documents Paneth cell plasticity, particularly their ability to participate in epithelial replenishment following stem cell loss, adding to a growing body of knowledge detailing the molecular pathways controlling injury-induced regeneration.


Assuntos
Celulas de Paneth/patologia , Receptores Notch/metabolismo , Adenoma/tratamento farmacológico , Adenoma/patologia , Animais , Células Cultivadas , Modelos Animais de Doenças , Injeções Intraperitoneais , Injeções Subcutâneas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Celulas de Paneth/efeitos dos fármacos , Receptores Notch/antagonistas & inibidores , Tamoxifeno/administração & dosagem , Tamoxifeno/farmacologia
10.
J Immunol ; 200(1): 186-195, 2018 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-29180487

RESUMO

Plasmacytoid dendritic cells (pDCs) are the major producers of IFN-α, an antiviral cytokine involved in immunomodulation and control of HIV type 1 replication, whereas Toxoplasma gondii is a life-threatening opportunistic infection in AIDS patients. During infection with HIV type 1, human pDCs decrease in circulation and remaining pDC produce lower amounts of IFN-α in response to viral stimulation. In this study, we investigated the impact of coinfection with T. gondii on the innate virus-directed responses of human pDCs. Using intracellular flow cytometry and fluorescence microscopy, we determined that T. gondii invaded but did not induce IFN-α or TNF-α in human pDC. However, T. gondii inhibited IFN-α and TNF-α produced in response to HSV and HIV, thus functionally inactivating pDC. IFN-α production was inhibited only in cells infected by T. gondii, which inhibited neither uptake of GFP-HSV nor localization of TLR9 in CD71+ endosomes, directing us to investigate downstream events. Using imaging flow cytometry, we found that both T. gondii and IL-10 inhibited virus-induced nuclear translocation, but not phosphorylation, of IFN response factor 7. Blockade of IFN response factor 7 nuclear translocation and inhibition of the IFN-α response was partially reversed by a deficiency in the T. gondii-derived ROP16 kinase, known to directly phosphorylate STAT3, a critical mediator of IL-10's anti-inflammatory effects. Taken together, our results indicate that T. gondii suppresses pDC activation by mimicking IL-10's regulatory effects through an ROP16 kinase-dependent mechanism. Our findings further imply a convergent mechanism of inhibition of TLR signaling by T. gondii and IL-10 and suggest potential negative consequences of HIV/T. gondii coinfection.


Assuntos
Células Dendríticas/imunologia , Infecções por HIV/imunologia , HIV-1/imunologia , Interleucina-10/metabolismo , Infecções Oportunistas/imunologia , Proteínas Tirosina Quinases/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/imunologia , Toxoplasmose/imunologia , Diferenciação Celular , Células Cultivadas , Coinfecção , Células Dendríticas/parasitologia , Humanos , Imunidade Inata , Imunomodulação , Fator Regulador 7 de Interferon/metabolismo , Interferon-alfa/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Receptor Toll-Like 9/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
11.
Nat Immunol ; 17(4): 356-63, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-27002843

RESUMO

Innate cells are responsible for the rapid recognition of infection and mediate essential mechanisms of pathogen elimination, and also facilitate adaptive immune responses. We review here the numerous intricate interactions among innate cells that initiate protective immunity. The efficient eradication of pathogens depends on the coordinated actions of multiple cells, including innate cells and epithelial cells. Rather than acting as isolated effector cells, innate cells are in constant communication with other responding cells of the immune system, locally and distally. These interactions are critically important for the efficient control of primary infections as well for the development of 'trained' innate cells that facilitate the rapid elimination of homologous or heterologous infections.


Assuntos
Imunidade Adaptativa/imunologia , Citocinas/imunologia , Imunidade Inata/imunologia , Infecções/imunologia , Células Matadoras Naturais/imunologia , Células Mieloides/imunologia , Animais , Basófilos/imunologia , Eosinófilos/imunologia , Humanos , Macrófagos/imunologia , Mastócitos/imunologia , Monócitos/imunologia , Neutrófilos/imunologia
12.
Proc Natl Acad Sci U S A ; 111(17): 6437-42, 2014 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-24733931

RESUMO

Unlike most intracellular pathogens that gain access into host cells through endocytic pathways, Toxoplasma gondii initiates infection at the cell surface by active penetration through a moving junction and subsequent formation of a parasitophorous vacuole. Here, we describe a noncanonical pathway for T. gondii infection of macrophages, in which parasites are initially internalized through phagocytosis, and then actively invade from within a phagosomal compartment to form a parasitophorous vacuole. This phagosome to vacuole invasion (PTVI) pathway may represent an intermediary link between the endocytic and the penetrative routes for host cell entry by intracellular pathogens. The PTVI pathway is preferentially used by avirulent strains of T. gondii and confers an infectious advantage over virulent strains for macrophage tropism.


Assuntos
Macrófagos/parasitologia , Fagossomos/parasitologia , Toxoplasma/patogenicidade , Animais , Linhagem Celular , Macrófagos/patologia , Macrófagos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Fagocitose , Fagossomos/patologia , Fagossomos/ultraestrutura , Toxoplasma/ultraestrutura , Toxoplasmose/parasitologia , Toxoplasmose/patologia , Tropismo , Vacúolos/parasitologia , Vacúolos/patologia , Vacúolos/ultraestrutura
13.
Nat Immunol ; 14(11): 1118-1126, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24145791

RESUMO

Helminth infections are ubiquitous worldwide and can trigger potent immune responses that differ from and potentially antagonize host protective responses to microbial pathogens. In this Review we focus on the three main killers in infectious disease-AIDS, tuberculosis and malaria-and critically assesses whether helminths adversely influence host control of these diseases. We also discuss emerging concepts for how M2 macrophages and helminth-modulated dendritic cells can potentially influence the protective immune response to concurrent infections. Finally, we present evidence advocating for more efforts to determine how and to what extent helminths interfere with the successful control of specific concurrent coinfections.


Assuntos
Síndrome da Imunodeficiência Adquirida/imunologia , Células Dendríticas/imunologia , Helmintíase/imunologia , Macrófagos/imunologia , Malária/imunologia , Tuberculose/imunologia , Síndrome da Imunodeficiência Adquirida/epidemiologia , Síndrome da Imunodeficiência Adquirida/virologia , África/epidemiologia , Animais , Ásia/epidemiologia , Coinfecção , Células Dendríticas/microbiologia , Células Dendríticas/parasitologia , Células Dendríticas/virologia , Helmintíase/epidemiologia , Helmintíase/parasitologia , Helmintos/imunologia , Interações Hospedeiro-Parasita , Humanos , América Latina/epidemiologia , Macrófagos/microbiologia , Macrófagos/parasitologia , Macrófagos/virologia , Malária/epidemiologia , Malária/parasitologia , Tuberculose/epidemiologia , Tuberculose/microbiologia
14.
PLoS One ; 7(8): e42850, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22880122

RESUMO

Various cell types in both lymphoid and non-lymphoid tissues produce the anti-inflammatory cytokine interleukin (IL)-10 during murine cytomegalovirus (MCMV) infection. The functions of IL-10 in the liver during acute infection and the cells that generate this cytokine at this site have not been extensively investigated. In this study, we demonstrate that the production of IL-10 in the liver is elevated in C57BL/6 mice during late acute MCMV infection. Using IL-10 green fluorescence protein (GFP) reporter knock-in mice, designated IL-10-internal ribosomal entry site (IRES)-GFP-enhanced reporter (tiger), NK cells are identified as major IL-10 expressing cells in the liver after infection, along with T cells and other leukocytes. In the absence of IL-10, mice exhibit marked elevations in proinflammatory cytokines and in the numbers of mononuclear cells and lymphocytes infiltrating the liver during this infection. IL-10-deficiency also enhances liver injury without improving viral clearance from this site. Collectively, the results indicate that IL-10-producing cells in the liver provide protection from collateral injury by modulating the inflammatory response associated with MCMV infection.


Assuntos
Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/patologia , Inflamação/patologia , Interleucina-10/metabolismo , Fígado/imunologia , Fígado/patologia , Muromegalovirus/fisiologia , Doença Aguda , Alanina Transaminase/sangue , Animais , Linfócitos T CD8-Positivos , Quimiocinas/biossíntese , Infecções por Citomegalovirus/sangue , Infecções por Citomegalovirus/virologia , Inflamação/sangue , Inflamação/complicações , Inflamação/microbiologia , Mediadores da Inflamação/metabolismo , Interferon gama/metabolismo , Interleucina-10/biossíntese , Interleucina-10/deficiência , Leucócitos/metabolismo , Fígado/virologia , Camundongos , Camundongos Endogâmicos C57BL , Fator de Necrose Tumoral alfa/metabolismo , Carga Viral
15.
J Exp Med ; 206(6): 1227-35, 2009 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-19487420

RESUMO

Prior studies have demonstrated that cholera toxin (CT) and other cAMP-inducing factors inhibit interleukin (IL)-12 production from monocytes and dendritic cells (DCs). We show that CT inhibits Th1 responses in vivo in mice infected with Toxoplasma gondii. This correlated with low serum IL-12 levels and a selective reduction in the numbers of CD8alpha(+) conventional DCs (cDCs) in lymphoid organs. CT inhibited the function of interferon (IFN) regulatory factor (IRF) 8, a transcription factor known to positively regulate IL-12p35 and p40 gene expression, and the differentiation of CD8alpha(+) and plasmacytoid DCs (pDCs). Fluorescence recovery after photobleaching analysis showed that exposure to CT, forskolin, or dibutyryl (db) cAMP blocked LPS and IFN-gamma-induced IRF8 binding to chromatin. Moreover, CT and dbcAMP inhibited the binding of IRF8 to the IFN-stimulated response element (ISRE)-like element in the mouse IL-12p40 promoter, likely by blocking the formation of ISRE-binding IRF1-IRF8 heterocomplexes. Furthermore, CT inhibited the differentiation of pDCs from fms-like tyrosine kinase 3 ligand-treated bone marrow cells in vitro. Therefore, because IRF8 is essential for IL-12 production and the differentiation of CD8alpha(+) cDCs and pDCs, these data suggest that CT and other Gs-protein agonists can affect IL-12 production and DC differentiation via a common mechanism involving IRF8.


Assuntos
Antígenos CD8/imunologia , Diferenciação Celular/fisiologia , Toxina da Cólera/metabolismo , AMP Cíclico/metabolismo , Células Dendríticas/fisiologia , Fatores Reguladores de Interferon/imunologia , Interleucina-12/biossíntese , Animais , Linfócitos T CD8-Positivos , Células Cultivadas , Células Dendríticas/citologia , Células Dendríticas/imunologia , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Humanos , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/imunologia , Fatores Reguladores de Interferon/genética , Interferon gama/imunologia , Interleucina-12/genética , Subunidade p40 da Interleucina-12/genética , Subunidade p40 da Interleucina-12/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Baço/citologia , Toxoplasmose/imunologia
16.
J Immunol ; 180(9): 5935-45, 2008 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-18424713

RESUMO

IFN-gamma-producing CD8(+) T lymphocytes are essential effector cells that mediate protective immunity during murine toxoplasmosis, and yet their effector development remains poorly characterized. Vaccination with the carbamoyl phosphate synthase (CPS) mutant strain of Toxoplasma gondii was used to examine the CD8(+) T cell response in the peritoneal effector site. Four CTL subpopulations with varying effector potentials were defined based on the expression of effector molecules and the cell surface activation markers CD62L and killer cell lectin-like receptor G1 (KLRG1). Further phenotypic analysis revealed that the acquisition of KLRG1 among effector subpopulations correlated with the down-regulation of both IL-7R and CD27, suggesting that KLRG1 marks dominant, end-stage effector cells. Using gene-targeted mice, we tested the in vivo requirements of key IL-12 signaling components for effector CTL differentiation. Contrary to established models of viral and bacterial infection, CD8(+) T cell-intrinsic IL-12 signaling was required for the generation of IFN-gamma-producing CTLs in response to T. gondii. Importantly, the development of the KLRG1(+) effector subpopulations, but not the memory precursor-containing KLRG1(-) effector subset, was critically reliant on IL-12. Furthermore, IL-12 signaling-dependent T-bet expression was also found to be important for differentiation of KLRG1(+) effectors. Our results underscore a vital role for IL-12 in not only the induction of IFN-gamma expression but also in the development of heterogeneous subpopulations of effector CD8(+) T cells generated in response to the intracellular parasite T. gondii.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Interferon gama/imunologia , Interleucina-12/imunologia , Receptores Imunológicos/imunologia , Toxoplasma/imunologia , Toxoplasmose Animal/imunologia , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/parasitologia , Carbono-Nitrogênio Ligases/genética , Carbono-Nitrogênio Ligases/imunologia , Carbono-Nitrogênio Ligases/metabolismo , Diferenciação Celular/genética , Regulação para Baixo/genética , Regulação para Baixo/imunologia , Interferon gama/genética , Interferon gama/metabolismo , Interleucina-12/genética , Interleucina-12/metabolismo , Selectina L/genética , Selectina L/imunologia , Selectina L/metabolismo , Lectinas Tipo C , Camundongos , Camundongos Knockout , Mutação , Proteínas de Protozoários/genética , Proteínas de Protozoários/imunologia , Proteínas de Protozoários/metabolismo , Vacinas Protozoárias/genética , Vacinas Protozoárias/imunologia , Vacinas Protozoárias/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Receptores de Interleucina-7/genética , Receptores de Interleucina-7/imunologia , Receptores de Interleucina-7/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Toxoplasma/enzimologia , Toxoplasmose Animal/enzimologia , Toxoplasmose Animal/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
17.
Infect Immun ; 75(10): 4799-803, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17682046

RESUMO

Autophagy has been implicated in the intracellular destruction of Toxoplasma gondii by primed macrophages following gamma interferon (IFN-gamma) activation of p47 GTPases. CD40 ligation has also been shown to trigger autophagic elimination of T. gondii independent of IFN-gamma and p47 GTPases. Here we demonstrate that IFN-gamma/p47 GTPase-dependent elimination of T. gondii by strain CPS vaccine-primed macrophages is independent of CD40/tumor necrosis factor signaling. Similar to wild-type controls, both CD40-deficient and tumor necrosis factor receptor 1/2 (TNFR1/2)-deficient macrophages can efficiently eliminate invaded strain GFP-PTG and restrain its replication following priming. In contrast, macrophages from mice lacking the IFN-gamma receptor gene neither clear the parasites nor repress their proliferation. Thus, CD40 and IFN-gamma-induced pathogen elimination might represent two independent resistance pathways, the latter of which plays a primary role in anti-Toxoplasma immunity in mice.


Assuntos
Antígenos CD40/imunologia , Interferon gama/imunologia , Macrófagos/imunologia , Macrófagos/parasitologia , Transdução de Sinais/imunologia , Toxoplasma/imunologia , Animais , Antígenos CD40/deficiência , Células Cultivadas , Feminino , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Tipo I de Fatores de Necrose Tumoral/deficiência , Receptores Tipo I de Fatores de Necrose Tumoral/imunologia , Receptores Tipo II do Fator de Necrose Tumoral/deficiência , Receptores Tipo II do Fator de Necrose Tumoral/imunologia , Toxoplasma/crescimento & desenvolvimento
18.
Blood ; 110(6): 1970-81, 2007 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-17548579

RESUMO

CpG-DNA or its synthetic analog CpG-ODN activates innate immunity through Toll-like receptor 9 (TLR9). However, the mechanism of TLR9 activation by CpG-DNA remains elusive. Here we have identified HMGB1 as a CpG-ODN-binding protein. HMGB1 interacts and preassociates with TLR9 in the endoplasmic reticulum-Golgi intermediate compartment (ERGIC), and hastens TLR9's redistribution to early endosomes in response to CpG-ODN. CpG-ODN stimulates macrophages and dendritic cells to secrete HMGB1; in turn, extracellular HMGB1 accelerates the delivery of CpG-ODNs to its receptor, leading to a TLR9-dependent augmentation of IL-6, IL-12, and TNFalpha secretion. Loss of HMGB1 leads to a defect in the IL-6, IL-12, TNFalpha, and iNOS response to CpG-ODN. However, lack of intracellular TLR9-associated HMGB1 can be compensated by extracellular HMGB1. Thus, the DNA-binding protein HMGB1 shuttles in and out of immune cells and regulates inflammatory responses to CpG-DNA.


Assuntos
Proteína HMGB1/fisiologia , Interleucina-12/metabolismo , Interleucina-6/metabolismo , Oligodesoxirribonucleotídeos/farmacologia , Receptor Toll-Like 9/fisiologia , Fator de Necrose Tumoral alfa/metabolismo , Animais , Calnexina/metabolismo , Núcleo Celular/metabolismo , Ilhas de CpG , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/metabolismo , Ensaio de Imunoadsorção Enzimática , Matriz Extracelular/metabolismo , Citometria de Fluxo , Complexo de Golgi/efeitos dos fármacos , Complexo de Golgi/metabolismo , Proteína HMGB1/genética , Immunoblotting , Imunoprecipitação , Lisossomos/metabolismo , Macrófagos/citologia , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Lectinas de Ligação a Manose/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Óxido Nítrico Sintase Tipo II/metabolismo , Vesículas Secretórias/efeitos dos fármacos , Vesículas Secretórias/metabolismo , Receptor Toll-Like 9/genética
19.
Autophagy ; 3(2): 163-5, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17204853

RESUMO

Autophagy has recently been implicated in the immune elimination of the intracellular protozoan parasite, Toxoplasma gondii. Toxoplasma and other apicomplexan parasites actively invade host cells and form nonfusogenic parasitophorous vacuoles. Nevertheless, following entry into IFN-gamma-activated effector macrophages, vesiculation of the parasite vacuole or PV membrane ensues, in a process dependent upon the activity of p47 GTPases induced by IFN-gamma signaling. Subsequent disruption of the plasma membrane of the stripped parasites precedes autophagolysosomal elimination of T. gondii. In contrast, ligation of the CD40 receptor and autocrine signaling by TNF activate a seemingly distinct, p47 GTPase-independent mechanism leading to autophagic elimination of intracellular T. gondii, without prior disruption of the pathogen vacuole. Thus, two key pathways of the cell-mediated immune response, namely IFN-gamma and CD40/CD40L, trigger a common autophagolysosomal endpoint of parasite elimination, via distinct intermediary mechanisms.


Assuntos
Autofagia , Antígenos CD40/metabolismo , Interferon gama/metabolismo , Toxoplasma/imunologia , Animais , Antígenos CD40/imunologia , Indução Enzimática , GTP Fosfo-Hidrolases/metabolismo , Humanos , Interferon gama/imunologia , Camundongos , Vacúolos/metabolismo
20.
J Exp Med ; 203(9): 2063-71, 2006 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-16940170

RESUMO

Apicomplexan protozoan pathogens avoid destruction and establish a replicative niche within host cells by forming a nonfusogenic parasitophorous vacuole (PV). Here we present evidence for lysosome-mediated degradation of Toxoplasma gondii after invasion of macrophages activated in vivo. Pathogen elimination was dependent on the interferon gamma inducible-p47 GTPase, IGTP, required PI3K activity, and was preceded by PV membrane indentation, vesiculation, disruption, and, surprisingly, stripping of the parasite plasma membrane. Denuded parasites were enveloped in autophagosome-like vacuoles, which ultimately fused with lysosomes. These observations outline a series of mechanisms used by effector cells to redirect the fate of a classically nonfusogenic intracellular pathogen toward a path of immune elimination.


Assuntos
Autofagia/fisiologia , Membrana Celular/metabolismo , Macrófagos , Toxoplasma/metabolismo , Vacúolos/metabolismo , Animais , Membrana Celular/parasitologia , Membrana Celular/ultraestrutura , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Humanos , Imuno-Histoquímica , Interferon gama/imunologia , Lisossomos/metabolismo , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/parasitologia , Fusão de Membrana/fisiologia , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Toxoplasma/imunologia , Vacúolos/parasitologia , Vacúolos/ultraestrutura
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA