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.
Life Sci Alliance ; 4(7)2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34078740

RESUMO

Interferon-inducible GTPases, such as immunity-related GTPases (IRGs) and guanylate-binding proteins (GBPs), are essential for cell-autonomous immunity against a wide variety of intracellular pathogens including Toxoplasma IRGs comprise regulatory and effector subfamily proteins. Regulatory IRGs Irgm1 and Irgm3 play important roles in anti-Toxoplasma immunity by globally controlling effector IRGs and GBPs. There is a remaining regulatory IRG, called Irgm2, which highly accumulates on parasitophorous vacuole membranes (PVMs). Very little is known about the mechanism of the unique localization on Toxoplasma PVMs. Here, we show that Irgm2 is important to control parasite killing through recruitment of Gbp1 and Irgb6, which does not require Irgm2 localization at Toxoplasma PVMs. Ubiquitination of Irgm2 in the cytosol, but not at the PVM, is also important for parasite killing through recruitment of Gbp1 to the PVM. Conversely, PVM ubiquitination and p62/Sqstm1 loading at later time points post-Toxoplasma infection require Irgm2 localization at the PVM. Irgm2-deficient mice are highly susceptible to Toxoplasma infection. Taken together, these data indicate that Irgm2 selectively controls accumulation of anti-Toxoplasma effectors to the vacuole in a manner dependent or independent on Irgm2 localization at the Toxoplasma PVM, which mediates parasite killing.


Assuntos
Proteínas de Ligação ao GTP/metabolismo , Imunidade Celular/imunologia , Toxoplasma/metabolismo , Animais , Proteínas de Transporte/metabolismo , Linhagem Celular , Fibroblastos/metabolismo , GTP Fosfo-Hidrolases/metabolismo , Interações Hospedeiro-Parasita/imunologia , Imunidade Inata/imunologia , Interferon gama/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Monoméricas de Ligação ao GTP , Toxoplasma/patogenicidade , Toxoplasmose/metabolismo , Toxoplasmose/parasitologia , Vacúolos
2.
J Exp Med ; 218(7)2021 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-33970189

RESUMO

Because of their common signaling molecules, the main T cell receptor (TCR) signaling cascades in CD4+ and CD8+ T cells are considered qualitatively identical. Herein, we show that TCR signaling in CD8+ T cells is qualitatively different from that in CD4+ T cells, since CD8α ignites another cardinal signaling cascade involving phospholipase C ß4 (PLCß4). TCR-mediated responses were severely impaired in PLCß4-deficient CD8+ T cells, whereas those in CD4+ T cells were intact. PLCß4-deficient CD8+ T cells showed perturbed activation of peripheral TCR signaling pathways downstream of IP3 generation. Binding of PLCß4 to the cytoplasmic tail of CD8α was important for CD8+ T cell activation. Furthermore, GNAQ interacted with PLCß4, mediated double phosphorylation on threonine 886 and serine 890 positions of PLCß4, and activated CD8+ T cells in a PLCß4-dependent fashion. PLCß4-deficient mice exhibited defective antiparasitic host defense and antitumor immune responses. Altogether, PLCß4 differentiates TCR signaling in CD4+ and CD8+ T cells and selectively promotes CD8+ T cell-dependent adaptive immunity.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Fosfolipase C beta/imunologia , Transdução de Sinais/imunologia , Animais , Linhagem Celular , Citoplasma/imunologia , Células HEK293 , Humanos , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação/imunologia , Receptores de Antígenos de Linfócitos T/imunologia
3.
Parasitol Int ; 83: 102335, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33771680

RESUMO

Sporozoites of the etiological agent of malaria, Plasmodium, form parasitophorous vacuoles (PVs) in hepatocytes. The PV membranes (PVM) are coated with a well-known host autophagy marker LC3 and parasite-derived protein called Upregulated in infective sporozoites 3 (UIS3), which has been shown to interact with LC3 and inhibit LC3-mediated autophagic disruption at the PV. Although uis3(-) sporozoites cannot proliferate in wild-type cells, they can replicate efficiently in cells defective in autophagy due to the lack of Atg proteins such as Atg3, Atg5 and Atg7, since these Atg proteins are essential for processing of LC3. However, it remains to be seen whether other Atg proteins participate in the restriction of uis3(-) parasite growth. Here we show that, despite essential roles of Atg9 and Atg14 in autophagy, both proteins are dispensable for the restriction of uis3(-) parasite growth. Moreover, we found that cells lacking LC3 proteins are also able to restrict uis3(-) parasite growth. In sharp contrast, GABARAPs, another subfamily of mammalian Atg8, participated in suppression of uis3(-) parasite growth. Taken together, contrary to a previous model in which UIS3 avoids host LC3- and autophagy-dependent parasite elimination program, our data demonstrate a role of GABARAPs for suppression of uis3(-) parasite growth in a manner independent on autophagy.


Assuntos
Proteínas Reguladoras de Apoptose/genética , Autofagia , Malária/parasitologia , Proteínas Associadas aos Microtúbulos/genética , Plasmodium berghei/fisiologia , Animais , Proteínas Reguladoras de Apoptose/metabolismo , Interações Hospedeiro-Parasita , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo
4.
Front Immunol ; 11: 561948, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33042141

RESUMO

Sepsis is a life-threating multi-organ disease induced by host innate immunity to pathogen-derived endotoxins including lipopolysaccharide (LPS). Direct sensing of LPS by caspase-11 activates inflammasomes and causes lethal sepsis in mice. Inhibition of caspase-11 inflammasomes is important for the prevention of LPS-induced septic shock; however, whether a caspase-11 inflammasome-specific suppressive mechanism exists is unclear. Here we show that deficiency of GABARAP autophagy-related proteins results in over-activation of caspase-11 inflammasomes but not of canonical inflammasomes. Gate-16-/-Gabarap-/- macrophages exhibited elevated guanylate binding protein 2 (GBP2)-dependent caspase-11 activation and inflammatory responses. Deficiency of GABARAPs resulted in formation of GBP2-containing aggregates that promote IL-1ß production. High mortality after low dose LPS challenge in Gate-16-/-Gabarap-/- mice primed with poly(I:C) or polymicrobial sepsis was ameliorated by compound GBP2 deficiency. These results reveal a critical function of Gate-16 and Gabarap to suppress GBP2-dependent caspase-11-induced inflammation and septic shock.


Assuntos
Proteínas Reguladoras de Apoptose/deficiência , Família da Proteína 8 Relacionada à Autofagia/deficiência , Caspases Iniciadoras/metabolismo , Proteínas Associadas aos Microtúbulos/deficiência , Choque Séptico/imunologia , Choque Séptico/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Família da Proteína 8 Relacionada à Autofagia/genética , Proteínas de Ligação ao GTP/deficiência , Imunidade Inata , Inflamassomos/metabolismo , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Lipopolissacarídeos/efeitos adversos , Macrófagos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/genética , Piroptose/genética , Choque Séptico/induzido quimicamente , Transdução de Sinais/genética
5.
Parasitol Int ; 75: 102049, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-31901434

RESUMO

Interferon-γ (IFN-γ) is important for host defense against various intracellular organisms including a protozoan pathogen Toxoplasma gondii. Various immune cells are recently shown to produce IFN-γ in T. gondii infection, however, it remains elusive which cell types are important for anti-T. gondii host defense so far. Here we generate a new IFN-γ reporter "GREVEN" mouse line in which a fusion protein of Venus and NanoLuc to analyze IFN-γ producing cells during T. gondii infection and find that CD4+, CD8+, γδ T cells and natural killer cells express Venus in a time dependent manner. Furthermore, Lck-Cre/Ifngfl/fl mice are highly susceptible to T. gondii infection. Taken together, our results demonstrate that T cell-derived IFN-γ plays an important role in anti-T. gondii host defense.


Assuntos
Resistência à Doença/imunologia , Interferon gama/imunologia , Linfócitos T/imunologia , Toxoplasma/imunologia , Toxoplasmose Animal/imunologia , Animais , Interferon gama/genética , Camundongos , Toxoplasmose/imunologia
6.
Life Sci Alliance ; 3(1)2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31852733

RESUMO

Toxoplasma gondii is an obligate intracellular protozoan parasite capable of infecting warm-blooded animals by ingestion. The organism enters host cells and resides in the cytoplasm in a membrane-bound parasitophorous vacuole (PV). Inducing an interferon response enables IFN-γ-inducible immunity-related GTPase (IRG protein) to accumulate on the PV and to restrict parasite growth. However, little is known about the mechanisms by which IRG proteins recognize and destroy T. gondii PV. We characterized the role of IRG protein Irgb6 in the cell-autonomous response against T. gondii, which involves vacuole ubiquitination and breakdown. We show that Irgb6 is capable of binding a specific phospholipid on the PV membrane. Furthermore, the absence of Irgb6 causes reduced targeting of other effector IRG proteins to the PV. This suggests that Irgb6 has a role as a pioneer in the process by which multiple IRG proteins access the PV. Irgb6-deficient mice are highly susceptible to infection by a strain of T. gondii avirulent in wild-type mice.


Assuntos
Interações Hospedeiro-Parasita/genética , Proteínas Monoméricas de Ligação ao GTP/metabolismo , Fosfolipídeos/metabolismo , Toxoplasma/citologia , Toxoplasmose/metabolismo , Vacúolos/metabolismo , Animais , Células Cultivadas , Feminino , Fibroblastos/metabolismo , Interações Hospedeiro-Parasita/efeitos dos fármacos , Interações Hospedeiro-Parasita/imunologia , Imunidade Inata , Interferon gama/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Monoméricas de Ligação ao GTP/deficiência , Proteínas Monoméricas de Ligação ao GTP/genética , Ligação Proteica , Proteínas Recombinantes/farmacologia , Toxoplasmose/parasitologia , Ubiquitina/metabolismo , Ubiquitinação/efeitos dos fármacos , Ubiquitinação/genética
7.
FASEB J ; 33(10): 10668-10679, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31268747

RESUMO

PLC-ß exerts biologic influences through GPCR. GPCRs are involved in regulating glucose-stimulated insulin secretion (GSIS). Previous studies have suggested that PLC-ßs might play an important role in pancreatic ß cells. However, because of a lack of the specific inhibitors of PLC-ß isozymes and appropriate genetic models, the in vivo function of specific PLC-ß isozymes in pancreatic ß cells and their physiologic relevance in the regulation of insulin secretion have not been studied so far. The present study showed that PLC-ß1 was crucial for ß-cell function by generation of each PLC-ß conditional knockout mouse. Mice lacking PLC-ß1 in ß cells exhibited a marked defect in GSIS, leading to glucose intolerance. In ex vivo studies, the secreted insulin level and Ca2+ response in Plcb1f/f; pancreas/duodenum homeobox protein 1 (Pdx1)-Cre recombinase-estrogen receptor T2 (CreERt2) islets was lower than those in the Plcb1f/f islets under the high-glucose condition. PLC-ß1 led to potentiate insulin secretion via stimulation of particular Gq-protein-coupled receptors. Plcb1f/f; Pdx1-CreERt2 mice fed a high-fat diet developed more severe glucose intolerance because of a defect in insulin secretion. The present study identified PLC-ß1 as an important molecule that regulates ß cell insulin secretion and can be considered a candidate for therapeutic intervention in diabetes mellitus.-Hwang, H.-J., Yang, Y. R., Kim, H. Y., Choi, Y., Park, K.-S., Lee, H., Ma, J. S., Yamamoto, M., Kim, J., Chae, Y. C., Choi, J. H., Cocco, L., Berggren, P.-O., Jang, H.-J., Suh, P.-G. Phospholipase Cß1 potentiates glucose-stimulated insulin secretion.


Assuntos
Glucose/metabolismo , Secreção de Insulina/fisiologia , Fosfolipase C beta/metabolismo , Animais , Linhagem Celular , Dieta Hiperlipídica/efeitos adversos , Intolerância à Glucose/genética , Intolerância à Glucose/metabolismo , Intolerância à Glucose/patologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Técnicas In Vitro , Secreção de Insulina/genética , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Ilhotas Pancreáticas/metabolismo , Ilhotas Pancreáticas/patologia , Isoenzimas/deficiência , Isoenzimas/genética , Isoenzimas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfolipase C beta/deficiência , Fosfolipase C beta/genética , Receptores Acoplados a Proteínas G/metabolismo , Transativadores/genética , Transativadores/metabolismo
8.
J Exp Med ; 216(8): 1733-1748, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31189656

RESUMO

The liver stage of the etiological agent of malaria, Plasmodium, is obligatory for successful infection of its various mammalian hosts. Differentiation of the rod-shaped sporozoites of Plasmodium into spherical exoerythrocytic forms (EEFs) via bulbous expansion is essential for parasite development in the liver. However, little is known about the host factors regulating the morphological transformation of Plasmodium sporozoites in this organ. Here, we show that sporozoite differentiation into EEFs in the liver involves protein kinase C ζ-mediated NF-κB activation, which robustly induces the expression of C-X-C chemokine receptor type 4 (CXCR4) in hepatocytes and subsequently elevates intracellular Ca2+ levels, thereby triggering sporozoite transformation into EEFs. Blocking CXCR4 expression by genetic or pharmacological intervention profoundly inhibited the liver-stage development of the Plasmodium berghei rodent malaria parasite and the human Plasmodium falciparum parasite. Collectively, our experiments show that CXCR4 is a key host factor for Plasmodium development in the liver, and CXCR4 warrants further investigation for malaria prophylaxis.


Assuntos
Hepatócitos/metabolismo , Malária/metabolismo , Plasmodium berghei/crescimento & desenvolvimento , Plasmodium falciparum/crescimento & desenvolvimento , Receptores CXCR4/metabolismo , Animais , Sistemas CRISPR-Cas , Cálcio/metabolismo , Linhagem Celular Tumoral , Humanos , Fígado/metabolismo , Malária/parasitologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , NF-kappa B/metabolismo , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores CXCR4/genética , Esporozoítos/metabolismo , Transfecção
9.
Artigo em Inglês | MEDLINE | ID: mdl-31119110

RESUMO

Toxoplasma gondii is an important human and animal pathogen that causes life-threatening toxoplasmosis. The host immune system produces interferon-γ (IFN-γ) to inhibit T. gondii proliferation. IFN-γ-inducible indole-2,3-dioxygenase 1 (IDO1), which mediates tryptophan degradation, has a major role in anti-T. gondii immune responses in various human cells. In response to the host's immune system, T. gondii secretes many virulence molecules into the host cells to suppress IFN-γ-dependent antiparasitic immune responses. The GRA15-induced proparasitic mechanism for suppressing IDO1-dependent immune responses has previously been tested only in human hepatocyte and monocyte co-cultures. Thus, whether human cells other than hepatocytes contain this virulence mechanism remains unclear. Here, we show that the GRA15-dependent virulence mechanism for suppressing the IDO1-dependent anti-T. gondii response operates in human neuronal cell lines and primary human neurons. Analysis of various human cell lines revealed that IL-1ß-induced iNOS-dependent reduction of IDO1 mRNA expression occurred in brain cell lines (A172; glioblastoma, IMR-32; neuroblastoma, and T98G; glioblastoma) and liver cell lines (Huh7 and HepG2), but not in other cell lines. Moreover, co-culturing type II T. gondii-infected THP-1 human monocytes with the brain cell lines inhibited the IDO1-mediated anti-T. gondii response in a GRA15-dependent manner. These data suggest that a GRA15-dependent virulence mechanism antagonizes the IDO1-dependent host immune response in human brain cells.


Assuntos
Antígenos de Protozoários/metabolismo , Antiparasitários/metabolismo , Interferon gama/metabolismo , Neurônios/metabolismo , Proteínas de Protozoários/metabolismo , Toxoplasma/metabolismo , Toxoplasmose/imunologia , Antígenos de Protozoários/imunologia , Antiparasitários/farmacologia , Linhagem Celular , Hepatócitos/imunologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/farmacologia , Interferon gama/imunologia , Interleucina-1beta/metabolismo , Monócitos/imunologia , Óxido Nítrico Sintase Tipo II/metabolismo , Proteínas de Protozoários/imunologia , RNA Mensageiro/metabolismo , Toxoplasma/efeitos dos fármacos , Toxoplasma/imunologia , Virulência
10.
Front Immunol ; 9: 2073, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30283439

RESUMO

Toxoplasma gondii is an important human and animal pathogen that causes life-threatening toxoplasmosis. Interferon-γ (IFN-γ) is critical for anti-T. gondii cell-autonomous immunity in both humans and mice. To proliferate efficiently within the hosts, virulent strains of T. gondii can suppress IFN-γ-dependent immunity. During parasite infection, it is well-characterized that various virulence effectors are secreted to transcriptionally or post-translationally target IFN-γ-inducible GTPases, which are essential for anti-parasite responses in mice. However, the role of IFN-γ-inducible GTPases in anti-T. gondii responses in human cells is controversial since they are non-functional or absent in humans. Instead, IFN-γ-induced tryptophan degradation by indole-2,3-dioxygenase (IDO) is important for the anti-T. gondii human response. To date, the T. gondii virulent mechanism targeting IDO in human cells remains elusive. Here we show that although humans possess two IDO isozymes, IDO1 and IDO2, human cells of various origins require IDO1 but not IDO2 for IFN-γ-induced cell-autonomous immunity to T. gondii. T. gondii secretes an effector TgIST to inhibit IDO1 mRNA expression. Taken together, the data suggests that T. gondii possesses virulence programs operated by TgIST to antagonize IFN-γ-induced IDO1-mediated anti-parasite cell-autonomous immunity in human cells.


Assuntos
Imunidade Celular/imunologia , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Interferon gama/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Animais , Autofagia/genética , Autofagia/imunologia , Proteínas Relacionadas à Autofagia/genética , Proteínas Relacionadas à Autofagia/imunologia , Proteínas Relacionadas à Autofagia/metabolismo , Linhagem Celular Tumoral , Células Cultivadas , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/imunologia , GTP Fosfo-Hidrolases/metabolismo , Células HeLa , Humanos , Imunidade Celular/genética , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interferon gama/metabolismo , Camundongos Knockout , Toxoplasma/patogenicidade , Toxoplasmose/enzimologia , Toxoplasmose/parasitologia , Virulência/genética , Virulência/imunologia
11.
mBio ; 9(5)2018 10 09.
Artigo em Inglês | MEDLINE | ID: mdl-30301855

RESUMO

Although Toxoplasma virulence mechanisms targeting gamma interferon (IFN-γ)-induced cell-autonomous antiparasitic immunity have been extensively characterized in mice, the virulence mechanisms in humans remain uncertain, partly because cell-autonomous immune responses against Toxoplasma differ markedly between mice and humans. Despite the identification of inducible nitric oxide synthase (iNOS) as an anti-Toxoplasma host factor in mice, here we show that iNOS in humans is a pro-Toxoplasma host factor that promotes the growth of the parasite. The GRA15 Toxoplasma effector-dependent disarmament of IFN-γ-induced parasite growth inhibition was evident when parasite-infected monocytes were cocultured with hepatocytes. Interleukin-1ß (IL-1ß), produced from monocytes in a manner dependent on GRA15 and the host's NLRP3 inflammasome, combined with IFN-γ to strongly stimulate iNOS expression in hepatocytes; this dramatically reduced the levels of indole 2,3-dioxygenase 1 (IDO1), a critically important IFN-γ-inducible anti-Toxoplasma protein in humans, thus allowing parasite growth. Taking the data together, Toxoplasma utilizes human iNOS to antagonize IFN-γ-induced IDO1-mediated cell-autonomous immunity via its GRA15 virulence factor.IMPORTANCEToxoplasma, an important intracellular parasite of humans and animals, causes life-threatening toxoplasmosis in immunocompromised individuals. Gamma interferon (IFN-γ) is produced in the host to inhibit the proliferation of this parasite and eventually cause its death. Unlike mouse disease models, which involve well-characterized virulence strategies that are used by Toxoplasma to suppress IFN-γ-dependent immunity, the strategies used by Toxoplasma in humans remain unclear. Here, we show that GRA15, a Toxoplasma effector protein, suppresses the IFN-γ-induced indole-2,3-dioxygenase 1-dependent antiparasite immune response in human cells. Because NLRP3-dependent production of IL-1ß and nitric oxide (NO) in Toxoplasma-infected human cells is involved in the GRA15-dependent virulence mechanism, blocking NO or IL-1ß production in the host could represent a novel therapeutic approach for treating human toxoplasmosis.


Assuntos
Interferon gama/farmacologia , Óxido Nítrico Sintase Tipo II/imunologia , Proteínas de Protozoários/imunologia , Toxoplasma/imunologia , Fatores de Virulência/imunologia , Animais , Antígenos de Protozoários/imunologia , Sistemas CRISPR-Cas , Linhagem Celular , Técnicas de Cocultura , Hepatócitos/efeitos dos fármacos , Hepatócitos/imunologia , Hepatócitos/parasitologia , Interações Hospedeiro-Parasita/imunologia , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/imunologia , Monócitos/parasitologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Toxoplasma/patogenicidade
12.
Nat Immunol ; 18(8): 899-910, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28604719

RESUMO

Mammalian autophagy-related 8 (Atg8) homologs consist of LC3 proteins and GABARAPs, all of which are known to be involved in canonical autophagy. In contrast, the roles of Atg8 homologs in noncanonical autophagic processes are not fully understood. Here we show a unique role of GABARAPs, in particular gamma-aminobutyric acid (GABA)-A-receptor-associated protein-like 2 (Gabarapl2; also known as Gate-16), in interferon-γ (IFN-γ)-mediated antimicrobial responses. Cells that lacked GABARAPs but not LC3 proteins and mice that lacked Gate-16 alone were defective in the IFN-γ-induced clearance of vacuolar pathogens such as Toxoplasma. Gate-16 but not LC3b specifically associated with the small GTPase ADP-ribosylation factor 1 (Arf1) to mediate uniform distribution of interferon-inducible GTPases. The lack of GABARAPs reduced Arf1 activation, which led to formation of interferon-inducible GTPase-containing aggregates and hampered recruitment of interferon-inducible GTPases to vacuolar pathogens. Thus, GABARAPs are uniquely required for antimicrobial host defense through cytosolic distribution of interferon-inducible GTPases.


Assuntos
Fator 1 de Ribosilação do ADP/imunologia , Autofagia/imunologia , Proteínas de Transporte/imunologia , Interferon gama/imunologia , Proteínas Associadas aos Microtúbulos/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Fator 1 de Ribosilação do ADP/metabolismo , Animais , Proteínas Reguladoras de Apoptose , Família da Proteína 8 Relacionada à Autofagia , Sistemas CRISPR-Cas , Proteínas de Transporte/metabolismo , Simulação por Computador , Proteínas do Citoesqueleto/imunologia , Proteínas do Citoesqueleto/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Imunofluorescência , GTP Fosfo-Hidrolases/imunologia , GTP Fosfo-Hidrolases/metabolismo , Edição de Genes , Immunoblotting , Imunoprecipitação , Interferon gama/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular , Proteínas de Membrana/imunologia , Proteínas de Membrana/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo
13.
Mol Brain ; 9(1): 100, 2016 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-27998287

RESUMO

The transition from wakefulness to a nonrapid eye movement (NREM) sleep state at the onset of sleep involves a transition from low-voltage, high-frequency irregular electroencephalography (EEG) waveforms to large-amplitude, low-frequency EEG waveforms accompanying synchronized oscillatory activity in the thalamocortical circuit. The thalamocortical circuit consists of reciprocal connections between the thalamus and cortex. The cortex sends strong excitatory feedback to the thalamus, however the function of which is unclear. In this study, we investigated the role of the thalamic metabotropic glutamate receptor 1 (mGluR1)-phospholipase C ß4 (PLCß4) pathway in sleep control in PLCß4-deficient (PLCß4-/-) mice. The thalamic mGluR1-PLCß4 pathway contains synapses that receive corticothalamic inputs. In PLCß4-/- mice, the transition from wakefulness to the NREM sleep state was stimulated, and the NREM sleep state was stabilized, which resulted in increased NREM sleep. The power density of delta (δ) waves increased in parallel with the increased NREM sleep. These sleep phenotypes in PLCß4-/- mice were consistent in TC-restricted PLCß4 knockdown mice. Moreover, in vitro intrathalamic oscillations were greatly enhanced in the PLCß4-/- slices. The results of our study showed that thalamic mGluR1-PLCß4 pathway was critical in controlling sleep architecture.


Assuntos
Fosfolipase C beta/metabolismo , Receptores de Glutamato Metabotrópico/metabolismo , Sono/fisiologia , Tálamo/metabolismo , Animais , Córtex Cerebral/fisiologia , Ritmo Delta/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfolipase C beta/deficiência , Tálamo/fisiologia
14.
Cell Rep ; 13(2): 223-33, 2015 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-26440898

RESUMO

Also known as Sqstm1, p62 is a selective autophagy adaptor with a ubiquitin-binding domain. However, the role of p62 in the host defense against Toxoplasma gondii infection is unclear. Here, we show that interferon γ (IFN-γ) stimulates ubiquitin and p62 recruitment to T. gondii parasitophorous vacuoles (PVs). Some essential autophagy-related proteins, but not all, are required for this recruitment. Regardless of normal IFN-γ-induced T. gondii clearance activity and ubiquitination, p62 deficiency in antigen-presenting cells (APCs) and mice diminishes the robust IFN-γ-primed activation of CD8(+) T cells that recognize the T. gondii-derived antigen secreted into PVs. Because the expression of Atg3 and Irgm1/m3 in APCs is essential for PV disruption, ubiquitin and p62 recruitment, and vacuolar-antigen-specific CD8(+) T cell activation, IFN-γ-mediated ubiquitination and the subsequent recruitment of p62 to T. gondii are specifically required for the acquired immune response after PV disruption by IFN-γ-inducible GTPases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Apresentação de Antígeno , Antígenos de Protozoários/imunologia , Proteínas de Choque Térmico/imunologia , Interferon gama/metabolismo , Toxoplasma/imunologia , Vacúolos/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células Apresentadoras de Antígenos/imunologia , Autofagia , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular , Células Cultivadas , Proteínas de Choque Térmico/genética , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Proteína Sequestossoma-1 , Ubiquitina/metabolismo , Ubiquitinação
15.
Proc Natl Acad Sci U S A ; 112(33): E4581-90, 2015 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-26240314

RESUMO

IFN-γ orchestrates cell-autonomous host defense against various intracellular vacuolar pathogens. IFN-γ-inducible GTPases, such as p47 immunity-related GTPases (IRGs) and p65 guanylate-binding proteins (GBPs), are recruited to pathogen-containing vacuoles, which is important for disruption of the vacuoles, culminating in the cell-autonomous clearance. Although the positive regulation for the proper recruitment of IRGs and GBPs to the vacuoles has been elucidated, the suppressive mechanism is unclear. Here, we show that Rab GDP dissociation inhibitor α (RabGDIα), originally identified as a Rab small GTPase inhibitor, is a negative regulator of IFN-γ-inducible GTPases in cell-autonomous immunity to the intracellular pathogen Toxoplasma gondii. Overexpression of RabGDIα, but not of RabGDIß, impaired IFN-γ-dependent reduction of T. gondii numbers. Conversely, RabGDIα deletion in macrophages and fibroblasts enhanced the IFN-γ-induced clearance of T. gondii. Furthermore, upon a high dose of infection by T. gondii, RabGDIα-deficient mice exhibited a decreased parasite burden in the brain and increased resistance in the chronic phase than did control mice. Among members of IRGs and GBPs important for the parasite clearance, Irga6 and Gbp2 alone were more frequently recruited to T. gondii-forming parasitophorous vacuoles in RabGDIα-deficient cells. Notably, Gbp2 positively controlled Irga6 recruitment that was inhibited by direct and specific interactions of RabGDIα with Gbp2 through the lipid-binding pocket. Taken together, our results suggest that RabGDIα inhibits host defense against T. gondii by negatively regulating the Gbp2-Irga6 axis of IFN-γ-dependent cell-autonomous immunity.


Assuntos
GTP Fosfo-Hidrolases/metabolismo , Regulação Enzimológica da Expressão Gênica , Inibidores de Dissociação do Nucleotídeo Guanina/metabolismo , Interferon gama/imunologia , Toxoplasma/patogenicidade , Toxoplasmose/imunologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Chlorocebus aethiops , Primers do DNA/genética , Feminino , Fibroblastos/metabolismo , Inflamação/imunologia , Lipídeos/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Ligação Proteica , Homologia de Sequência de Aminoácidos , Células Vero
16.
J Exp Med ; 211(10): 2013-32, 2014 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-25225460

RESUMO

Toxoplasma gondii infection results in co-option and subversion of host cellular signaling pathways. This process involves discharge of T. gondii effector molecules from parasite secretory organelles such as rhoptries and dense granules. We report that the T. gondii polymorphic dense granule protein GRA6 regulates activation of the host transcription factor nuclear factor of activated T cells 4 (NFAT4). GRA6 overexpression robustly and selectively activated NFAT4 via calcium modulating ligand (CAMLG). Infection with wild-type (WT) but not GRA6-deficient parasites induced NFAT4 activation. Moreover, GRA6-deficient parasites failed to exhibit full virulence in local infection, and the treatment of WT mice with an NFAT inhibitor mitigated virulence of WT parasites. Notably, NFAT4-deficient mice displayed prolonged survival, decreased recruitment of CD11b(+) Ly6G(+) cells to the site of infection, and impaired expression of chemokines such as Cxcl2 and Ccl2. In addition, infection with type I parasites culminated in significantly higher NFAT4 activation than type II parasites due to a polymorphism in the C terminus of GRA6. Collectively, our data suggest that GRA6-dependent NFAT4 activation is required for T. gondii manipulation of host immune responses to maximize the parasite virulence in a strain-dependent manner.


Assuntos
Antígenos de Protozoários/metabolismo , Regulação da Expressão Gênica/fisiologia , Fatores de Transcrição NFATC/metabolismo , Proteínas de Protozoários/metabolismo , Transdução de Sinais/fisiologia , Toxoplasma/patogenicidade , Toxoplasmose/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antígenos de Protozoários/genética , Proteínas de Bactérias , Western Blotting , Feminino , Citometria de Fluxo , Imunoprecipitação , Luciferases , Proteínas Luminescentes , Camundongos , Camundongos Endogâmicos BALB C , Microscopia de Fluorescência , Fatores de Transcrição NFATC/antagonistas & inibidores , Proteínas de Protozoários/genética , Reação em Cadeia da Polimerase em Tempo Real , Toxoplasma/metabolismo
17.
PLoS One ; 8(2): e55800, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23409047

RESUMO

Although the pyrazolone derivative sulpyrine is widely used as an antipyretic analgesic drug, side effects, including fatal shock, have been reported. However, the molecular mechanism underlying such a severe side effect is largely unclear. Here, we report that the transcription factor CREBH that is highly expressed in the liver plays an important role in fatal shock induced by sulpyrine in mice. CREBH-deficient mice were resistant to experimental fatal sulpyrine shock. We found that sulpyrine-induced expression of cytochrome P450 2B (CYP2B) family genes, which are involved in sulpyrine metabolism, in the liver was severely impaired in CREBH-deficient mice. Moreover, introduction of CYP2B in CREBH-deficient liver restored susceptibility to sulpyrine. Furthermore, ectopic expression of CREBH up-regulated CYP2B10 promoter activity, and in vivo knockdown of CREBH in wild-type mice conferred a significant resistance to fatal sulpyrine shock. These data demonstrate that CREBH is a positive regulator of CYP2B in response to sulpyrine administration, which possibly results in fatal shock.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Dipirona/efeitos adversos , Choque/induzido quimicamente , Choque/genética , Ampirona/sangue , Animais , Hidrocarboneto de Aril Hidroxilases/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/deficiência , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Citocromo P-450 CYP2B1/genética , Citocromo P-450 CYP2B1/metabolismo , Família 2 do Citocromo P450 , Dipirona/farmacocinética , Modelos Animais de Doenças , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Knockout , Regiões Promotoras Genéticas , Interferência de RNA , Índice de Gravidade de Doença , Choque/mortalidade , Esteroide Hidroxilases/genética , Ativação Transcricional
18.
Immunity ; 37(2): 302-13, 2012 Aug 24.
Artigo em Inglês | MEDLINE | ID: mdl-22795875

RESUMO

Interferon-γ (IFN-γ) is essential for host defense against intracellular pathogens. Stimulation of innate immune cells by IFN-γ upregulates ∼2,000 effector genes such as immunity-related GTPases including p65 guanylate-binding protein (Gbp) family genes. We show that a cluster of Gbp genes was required for host cellular immunity against the intracellular parasite Toxoplasma gondii. We generated mice deficient for all six Gbp genes located on chromosome 3 (Gbp(chr3)) by targeted chromosome engineering. Mice lacking Gbp(chr3) were highly susceptible to T. gondii infection, resulting in increased parasite burden in immune organs. Furthermore, Gbp(chr3)-deleted macrophages were defective in IFN-γ-mediated suppression of T. gondii intracellular growth and recruitment of IFN-γ-inducible p47 GTPase Irgb6 to the parasitophorous vacuole. In addition, some members of Gbp(chr3) restored the protective response against T. gondii in Gbp(chr3)-deleted cells. Our results suggest that Gbp(chr3) play a pivotal role in anti-T. gondii host defense by controlling IFN-γ-mediated Irgb6-dependent cellular innate immunity.


Assuntos
Proteínas de Ligação ao GTP/imunologia , Imunidade Inata/imunologia , Interferon gama/imunologia , Macrófagos/imunologia , Toxoplasma/imunologia , Toxoplasmose/imunologia , Animais , Cromossomos de Mamíferos/genética , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Humanos , Imunidade Celular/imunologia , Interferon gama/antagonistas & inibidores , Interferon gama/metabolismo , Listeria monocytogenes/imunologia , Medições Luminescentes , Macrófagos/metabolismo , Macrófagos/ultraestrutura , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Eletrônica de Transmissão , Óxido Nítrico/metabolismo , Toxoplasma/metabolismo
19.
PLoS Pathog ; 8(5): e1002714, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22693446

RESUMO

Specific intestinal microbiota has been shown to induce Foxp3(+) regulatory T cell development. However, it remains unclear how development of another regulatory T cell subset, Tr1 cells, is regulated in the intestine. Here, we analyzed the role of two probiotic strains of intestinal bacteria, Lactobacillus casei and Bifidobacterium breve in T cell development in the intestine. B. breve, but not L. casei, induced development of IL-10-producing Tr1 cells that express cMaf, IL-21, and Ahr in the large intestine. Intestinal CD103(+) dendritic cells (DCs) mediated B. breve-induced development of IL-10-producing T cells. CD103(+) DCs from Il10(-/-), Tlr2(-/-), and Myd88(-/-) mice showed defective B. breve-induced Tr1 cell development. B. breve-treated CD103(+) DCs failed to induce IL-10 production from co-cultured Il27ra(-/-) T cells. B. breve treatment of Tlr2(-/-) mice did not increase IL-10-producing T cells in the colonic lamina propria. Thus, B. breve activates intestinal CD103(+) DCs to produce IL-10 and IL-27 via the TLR2/MyD88 pathway thereby inducing IL-10-producing Tr1 cells in the large intestine. Oral B. breve administration ameliorated colitis in immunocompromised mice given naïve CD4(+) T cells from wild-type mice, but not Il10(-/-) mice. These findings demonstrate that B. breve prevents intestinal inflammation through the induction of intestinal IL-10-producing Tr1 cells.


Assuntos
Bifidobacterium/imunologia , Colo/microbiologia , Interleucina-10/metabolismo , Lacticaseibacillus casei/imunologia , Probióticos/administração & dosagem , Linfócitos T Reguladores/citologia , Transferência Adotiva , Animais , Infecções por Bifidobacteriales/imunologia , Infecções por Bifidobacteriales/microbiologia , Infecções por Bifidobacteriales/terapia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/transplante , Diferenciação Celular , Técnicas de Cocultura , Colite/imunologia , Colite/microbiologia , Colite/terapia , Colo/imunologia , Células Dendríticas/citologia , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Interações Hospedeiro-Patógeno , Hospedeiro Imunocomprometido , Interleucina-10/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
20.
Proc Natl Acad Sci U S A ; 109(13): 5010-5, 2012 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-22403066

RESUMO

Adequate activation of CD4(+) T lymphocytes is essential for host defense against invading pathogens; however, exaggerated activity of effector CD4(+) T cells induces tissue damage, leading to inflammatory disorders such as inflammatory bowel diseases. Several unique subsets of intestinal innate immune cells have been identified. However, the direct involvement of innate immune cell subsets in the suppression of T-cell-dependent intestinal inflammation is poorly understood. Here, we report that intestinal CX(3)C chemokine receptor 1(high) (CX(3)CR1(high)) CD11b(+) CD11c(+) cells are responsible for prevention of intestinal inflammation through inhibition of T-cell responses. These cells inhibit CD4(+) T-cell proliferation in a cell contact-dependent manner and prevent T-cell-dependent colitis. The suppressive activity is abrogated in the absence of the IL-10/Stat3 pathway. These cells inhibit T-cell proliferation by two steps. Initially, CX(3)CR1(high) CD11b(+) CD11c(+) cells preferentially interact with T cells through highly expressed intercellular adhesion molecule-1/vascular cell adhesion molecule-1; then, they fail to activate T cells because of defective expression of CD80/CD86. The IL-10/Stat3 pathway mediates the reduction of CD80/CD86 expression. Transfer of wild-type CX(3)CR1(high) CD11b(+) CD11c(+) cells prevents development of colitis in myeloid-specific Stat3-deficient mice. Thus, these cells are regulatory myeloid cells that are responsible for maintaining intestinal homeostasis.


Assuntos
Colite/imunologia , Colite/patologia , Intestinos/imunologia , Intestinos/patologia , Células Mieloides/imunologia , Receptores de Quimiocinas/metabolismo , Linfócitos T/imunologia , Animais , Antígenos CD11/metabolismo , Receptor 1 de Quimiocina CX3C , Proliferação de Células , Colite/complicações , Colite/prevenção & controle , Inflamação/complicações , Inflamação/imunologia , Inflamação/patologia , Inflamação/prevenção & controle , Interleucina-10/metabolismo , Camundongos , Mucosa/imunologia , Mucosa/patologia , Fator de Transcrição STAT3/deficiência , Fator de Transcrição STAT3/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...