Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Gastroenterology ; 167(1): 34-50, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38286392

RESUMO

Celiac disease (CeD) is a chronic autoimmune condition driven by gluten ingestion in genetically predisposed individuals, resulting in inflammatory lesions in the proximal small intestine. Although the presence of specific HLA-linked haplotypes and gluten consumption are necessary for disease development, they alone do not account for the variable onset of CeD in susceptible individuals. This review explores the multifaceted role of non-host factors in CeD development, including dietary and microbial influences. We discuss clinical associations and observations highlighting the impact of these factors on disease onset and severity. Furthermore, we discuss studies in CeD-relevant animal models that offer mechanistic insights into how diet, the microbiome, and enteric infections modulate CeD pathogenesis. Finally, we address the clinical implications and therapeutic potential of understanding these cofactors offering a promising avenue for preventive and therapeutic interventions in CeD management.


Assuntos
Doença Celíaca , Microbioma Gastrointestinal , Índice de Gravidade de Doença , Doença Celíaca/imunologia , Doença Celíaca/microbiologia , Doença Celíaca/genética , Humanos , Animais , Fatores de Risco , Predisposição Genética para Doença , Glutens/imunologia , Glutens/efeitos adversos , Dieta/efeitos adversos , Modelos Animais de Doenças
2.
Immunity ; 56(8): 1862-1875.e9, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37478853

RESUMO

Loss of oral tolerance (LOT) to gluten, driven by dendritic cell (DC) priming of gluten-specific T helper 1 (Th1) cell immune responses, is a hallmark of celiac disease (CeD) and can be triggered by enteric viral infections. Whether certain commensals can moderate virus-mediated LOT remains elusive. Here, using a mouse model of virus-mediated LOT, we discovered that the gut-colonizing protist Tritrichomonas (T.) arnold promotes oral tolerance and protects against reovirus- and murine norovirus-mediated LOT, independent of the microbiota. Protection was not attributable to antiviral host responses or T. arnold-mediated innate type 2 immunity. Mechanistically, T. arnold directly restrained the proinflammatory program in dietary antigen-presenting DCs, subsequently limiting Th1 and promoting regulatory T cell responses. Finally, analysis of fecal microbiomes showed that T. arnold-related Parabasalid strains are underrepresented in human CeD patients. Altogether, these findings will motivate further exploration of oral-tolerance-promoting protists in CeD and other immune-mediated food sensitivities.


Assuntos
Antígenos , Imunidade Inata , Animais , Camundongos , Humanos , Dieta , Glutens , Células Dendríticas , Tolerância Imunológica
3.
Cell ; 186(9): 1846-1862.e26, 2023 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-37028428

RESUMO

The use of probiotics by cancer patients is increasing, including among those undergoing immune checkpoint inhibitor (ICI) treatment. Here, we elucidate a critical microbial-host crosstalk between probiotic-released aryl hydrocarbon receptor (AhR) agonist indole-3-aldehyde (I3A) and CD8 T cells within the tumor microenvironment that potently enhances antitumor immunity and facilitates ICI in preclinical melanoma. Our study reveals that probiotic Lactobacillus reuteri (Lr) translocates to, colonizes, and persists within melanoma, where via its released dietary tryptophan catabolite I3A, it locally promotes interferon-γ-producing CD8 T cells, thereby bolstering ICI. Moreover, Lr-secreted I3A was both necessary and sufficient to drive antitumor immunity, and loss of AhR signaling within CD8 T cells abrogated Lr's antitumor effects. Further, a tryptophan-enriched diet potentiated both Lr- and ICI-induced antitumor immunity, dependent on CD8 T cell AhR signaling. Finally, we provide evidence for a potential role of I3A in promoting ICI efficacy and survival in advanced melanoma patients.


Assuntos
Limosilactobacillus reuteri , Melanoma , Microambiente Tumoral , Humanos , Dieta , Inibidores de Checkpoint Imunológico , Limosilactobacillus reuteri/metabolismo , Melanoma/terapia , Triptofano/metabolismo , Linfócitos T CD8-Positivos/imunologia , Receptores de Hidrocarboneto Arílico/agonistas
4.
JCI Insight ; 7(16)2022 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-35993365

RESUMO

Celiac disease is an immune-mediated intestinal disorder that results from loss of oral tolerance (LOT) to dietary gluten. Reovirus elicits inflammatory Th1 cells and suppresses Treg responses to dietary antigen in a strain-dependent manner. Strain type 1 Lang (T1L) breaks oral tolerance, while strain type 3 Dearing reassortant virus (T3D-RV) does not. We discovered that intestinal infection by T1L in mice leads to the recruitment and activation of NK cells in mesenteric lymph nodes (MLNs) in a type I IFN-dependent manner. Once activated following infection, NK cells produce type II IFN and contribute to IFN-stimulated gene expression in the MLNs, which in turn induces inflammatory DC and T cell responses. Immune depletion of NK cells impairs T1L-induced LOT to newly introduced food antigen. These studies indicate that NK cells modulate the response to dietary antigen in the presence of a viral infection.


Assuntos
Tolerância Imunológica , Células Matadoras Naturais , Animais , Anticorpos Antivirais , Camundongos
5.
Cell Host Microbe ; 30(7): 1003-1019.e10, 2022 07 13.
Artigo em Inglês | MEDLINE | ID: mdl-35658976

RESUMO

The triggers that drive interferon-γ (IFNγ)-producing CD8 T cell (Tc1 cell)-mediated autoimmune hepatitis (AIH) remain obscure. Here, we show that lack of hematopoietic Tet methylcytosine dioxygenase 2 (Tet2), an epigenetic regulator associated with autoimmunity, results in the development of microbiota-dependent AIH-like pathology, accompanied by hepatic enrichment of aryl hydrocarbon receptor (AhR) ligand-producing pathobionts and rampant Tc1 cell immunity. We report that AIH-like disease development is dependent on both IFNγ and AhR signaling, as blocking either reverts ongoing AIH-like pathology. Illustrating the critical role of AhR-ligand-producing pathobionts in this condition, hepatic translocation of the AhR ligand indole-3-aldehyde (I3A)-releasing Lactobacillus reuteri is sufficient to trigger AIH-like pathology. Finally, we demonstrate that I3A is required for L. reuteri-induced Tc1 cell differentiation in vitro and AIH-like pathology in vivo, both of which are restrained by Tet2 within CD8 T cells. This AIH-disease model may contribute to the development of therapeutics to alleviate AIH.


Assuntos
Proteínas de Ligação a DNA , Dioxigenases , Hepatite Autoimune , Limosilactobacillus reuteri , Fígado , Microbiota , Animais , Proteínas de Ligação a DNA/genética , Dioxigenases/genética , Disbiose/complicações , Hepatite Autoimune/etiologia , Hepatite Autoimune/patologia , Interferon gama , Ligantes , Fígado/imunologia , Fígado/microbiologia , Camundongos , Microbiota/genética , Microbiota/imunologia , Linfócitos T Citotóxicos
6.
Trends Immunol ; 41(11): 967-971, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-33036909

RESUMO

Food sensitivities are on the rise worldwide. Peripheral induced regulatory T cells (pTreg cells) play a central role in oral tolerance to dietary antigens and can contribute to preventing the onset of immune-mediated food sensitivities. Here, we discuss the potential of microbial-derived products in promoting pTreg cell proliferation for re-establishing oral tolerance in immune-mediated food sensitivities.


Assuntos
Dieta , Hipersensibilidade Alimentar/imunologia , Hipersensibilidade Alimentar/microbiologia , Linfócitos T Reguladores , Proliferação de Células , Hipersensibilidade Alimentar/prevenção & controle , Humanos , Tolerância Imunológica/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/imunologia
7.
Blood Adv ; 4(11): 2430-2438, 2020 06 09.
Artigo em Inglês | MEDLINE | ID: mdl-32492156

RESUMO

Clonal hematopoiesis of indeterminate potential (CHIP) is predictive of hematological cancers and cardiovascular diseases, but the etiology of CHIP initiation and clonal expansion is unknown. Several lines of evidence suggest that proinflammatory cytokines may favor mutated hematopoietic stem cell expansion. To investigate the potential link between inflammation and CHIP, we performed targeted deep sequencing of 11 genes previously implicated in CHIP in 1887 subjects aged >70 years from the Montreal Heart Institute Biobank, of which 1359 had prior coronary artery disease (CAD), and 528 controls did not. We assessed association of CHIP with log transformed high-sensitivity C-reactive protein (hs-CRP), a validated biomarker of inflammation. CHIP was identified in 427 of the 1887 subjects (22.6%). CHIP mutations were more frequently identified in DNMT3A (11.6%) and TET2 (6.1%), with a higher proportion of TET2 mutations occurring in controls than in patients with CAD (9.0% vs 4.9%, P < .001). CHIP carriers had 21% higher hs-CRP levels compared with their noncarrier counterparts (eß = 1.21, 95% confidence interval [CI]: 1.08 to 1.36; P = .001). A similar effect was observed in the subgroup of patients with known CAD (eß = 1.22, 95% CI: 1.06 to 1.41; P = .005). These findings confirm the association between inflammation and CHIP. This association may open investigational avenues aimed at documenting mechanisms linking inflammation to clonal progression and ultimately supports prevention interventions to attenuate CHIP's impact on cardiovascular disease and cancer.


Assuntos
Proteína C-Reativa , Hematopoiese Clonal , Hematopoese , Intervenção Coronária Percutânea , Idoso , Antagonistas de Receptores de Angiotensina , Inibidores da Enzima Conversora de Angiotensina , Hematopoese/genética , Células-Tronco Hematopoéticas , Humanos , Masculino
8.
Cell Host Microbe ; 24(5): 677-688.e5, 2018 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-30392830

RESUMO

Intestinal reovirus infection can trigger T helper 1 (TH1) immunity to dietary antigen, raising the question of whether other viruses can have a similar impact. Here we show that the acute CW3 strain of murine norovirus, but not the persistent CR6 strain, induces TH1 immunity to dietary antigen. This property of CW3 is dependent on its major capsid protein, a virulence determinant. Transcriptional profiling of mesenteric lymph nodes following infection reveals an immunopathological signature that does not segregate with protective immunity but with loss of oral tolerance, in which interferon regulatory factor 1 is critical. These data show that viral capacity to trigger specific inflammatory pathways at sites where T cell responses to dietary antigens take place interferes with the development of tolerance to an oral antigen. Collectively, these data provide a foundation for the development of therapeutic strategies to prevent TH1-mediated complex immune disorders triggered by viral infections.


Assuntos
Infecções por Caliciviridae/imunologia , Dieta , Norovirus/imunologia , Norovirus/patogenicidade , Ovalbumina/imunologia , Células Th1/imunologia , Administração Oral , Animais , Infecções por Caliciviridae/virologia , Proteínas do Capsídeo/imunologia , Doença Celíaca/imunologia , Modelos Animais de Doenças , Feminino , Células HEK293 , Humanos , Imunidade , Inflamação , Fator Regulador 1 de Interferon/imunologia , Linfonodos , Camundongos , Camundongos Endogâmicos C57BL , Ovalbumina/administração & dosagem , Eliminação de Partículas Virais
9.
Front Immunol ; 9: 2311, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30349541

RESUMO

Genome-wide association studies as well as lymphatic expression analyses have linked both Cbl-b and GM-CSF to human multiple sclerosis as well as other autoimmune diseases. Both Cbl-b and GM-CSF have been shown to play a prominent role in the development of murine encephalomyelitis; however, no functional connection between the two has yet been established. In this study, we show that Cblb knockout mice demonstrated significantly exacerbated severity of experimental autoimmune encephalomyelitis (EAE), augmented T cell infiltration into the central nervous system (CNS) and strongly increased production of GM-CSF in T cells in vitro and in vivo.GM-CSF neutralization demonstrated that the increased susceptibility of Cblb-/- mice to EAE was dependent on GM-CSF. Mechanistically, p50 binding to the GM-CSF promoter and the IL-3/GM-CSF enhancer element "CNSa" was strongly increased in nuclear extracts from Cbl-b-deficient T cells. This study suggests that Cbl-b limits autoimmunity by preventing the pathogenic effects of GM-CSF overproduction in T cells.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Linfócitos T CD4-Positivos/metabolismo , Encefalomielite Autoimune Experimental/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Proteínas Proto-Oncogênicas c-cbl/fisiologia , Animais , Autoimunidade/genética , Regulação da Expressão Gênica , Interleucina-3/genética , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas
10.
Sci Immunol ; 3(24)2018 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-29934344

RESUMO

Epithelial-resident T lymphocytes, such as intraepithelial lymphocytes (IELs) located at the intestinal barrier, can offer swift protection against invading pathogens. Lymphocyte activation is strictly regulated because of its potential harmful nature and metabolic cost, and most lymphocytes are maintained in a quiescent state. However, IELs are kept in a heightened state of activation resembling effector T cells but without cytokine production or clonal proliferation. We show that this controlled activation state correlates with alterations in the IEL mitochondrial membrane, especially the cardiolipin composition. Upon inflammation, the cardiolipin composition is altered to support IEL proliferation and effector function. Furthermore, we show that cardiolipin makeup can particularly restrict swift IEL proliferation and effector functions, reducing microbial containment capability. These findings uncover an alternative mechanism to control cellular activity, special to epithelial-resident T cells, and a novel role for mitochondria, maintaining cells in a metabolically poised state while enabling rapid progression to full functionality.


Assuntos
Coccidiose/imunologia , Mucosa Intestinal/citologia , Linfócitos Intraepiteliais/imunologia , Mitocôndrias/metabolismo , Linfócitos T/imunologia , Animais , Cardiolipinas/metabolismo , Células Cultivadas , Coccidiose/parasitologia , Modelos Animais de Doenças , Eimeria/imunologia , Feminino , Humanos , Mucosa Intestinal/imunologia , Linfócitos Intraepiteliais/citologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Mitocôndrias/imunologia , Mitocôndrias/ultraestrutura , Membranas Mitocondriais/imunologia , Membranas Mitocondriais/metabolismo , Membranas Mitocondriais/ultraestrutura , Cultura Primária de Células , Linfócitos T/citologia
11.
Nature ; 557(7706): 580-584, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29769727

RESUMO

Somatic mutations in tet methylcytosine dioxygenase 2 (TET2), which encodes an epigenetic modifier enzyme, drive the development of haematopoietic malignancies1-7. In both humans and mice, TET2 deficiency leads to increased self-renewal of haematopoietic stem cells with a net developmental bias towards the myeloid lineage1,4,8,9. However, pre-leukaemic myeloproliferation (PMP) occurs in only a fraction of Tet2-/- mice8,9 and humans with TET2 mutations1,3,5-7, suggesting that extrinsic non-cell-autonomous factors are required for disease onset. Here we show that bacterial translocation and increased interleukin-6 production, resulting from dysfunction of the small-intestinal barrier, are critical for the development of PMP in mice that lack Tet2 expression in haematopoietic cells. Furthermore, in symptom-free Tet2-/- mice, PMP can be induced by disrupting intestinal barrier integrity, or in response to systemic bacterial stimuli such as the toll-like receptor 2 agonist. PMP was reversed by antibiotic treatment and failed to develop in germ-free Tet2-/- mice, which illustrates the importance of microbial signals in the development of this condition. Our findings demonstrate the requirement for microbial-dependent inflammation in the development of PMP and provide a mechanistic basis for the variation in PMP penetrance observed in Tet2-/- mice. This study will prompt new lines of investigation that may profoundly affect the prevention and management of haematopoietic malignancies.


Assuntos
Doenças Assintomáticas , Fenômenos Fisiológicos Bacterianos , Proliferação de Células , Proteínas de Ligação a DNA/deficiência , Leucemia/microbiologia , Leucemia/patologia , Proteínas Proto-Oncogênicas/deficiência , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Fenômenos Fisiológicos Bacterianos/imunologia , Proteínas de Ligação a DNA/genética , Dioxigenases , Feminino , Vida Livre de Germes , Inflamação/microbiologia , Interleucina-6/imunologia , Mucosa Intestinal/metabolismo , Lactobacillus/química , Lactobacillus/citologia , Lactobacillus/imunologia , Masculino , Camundongos , Penetrância , Permeabilidade , Proteínas Proto-Oncogênicas/genética , Receptor 2 Toll-Like/agonistas
12.
J Nutr Biochem ; 54: 95-104, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29331497

RESUMO

Inflammatory bowel diseases (IBD) are caused by the convergence of microbial, environmental, and genetic factors. Diet significantly alters these interactions by affecting both the host and microbiome. Using a mucosal inflammatory model that resembles the human condition of ileal pouchitis, we investigated the effects of Control (CONT) or Antioxidant (AOX) diet, containing pharmacologically relevant levels of 4 micronutrients, on disease risk in wild-type and IL-10-/- animals following surgical self-filling (SF) ileal blind loop placement. Although no differences were found in body weight change or survival, IL-10-/- CONT animals had significantly larger lymphoid organs compared with IL-10-/- AOX or with WT. SF loops from IL-10-/- CONT loop mucosa demonstrated histological inflammation, characterized by goblet cell depletion, increased mucosal myeloperoxidase (MPO), and elevated IFNγ, TNFα, and IL-17α gene expression, which AOX attenuated. AOX elevated luminal IgA in IL-10-/- animals, but not significantly in WT. In IL-10-/- animals, AOX significantly decreased the percentage of CD4 + T-bet and CD4 + RORγ T-cells compared with CONT, with no changes in CD4 + Foxp3+ Treg cells. 16S rRNA gene sequencing demonstrated AOX increased microbial alpha diversity compared with CONT in both genotypes. Notably, colonizing germ-free IL-10-/- hosts with CONT bacterial communities, but not AOX, recapitulated the inflammatory phenotype. Collectively, these findings highlight that common dietary antioxidant micronutrients reshape the gut microbial community to mitigate intestinal inflammatory profiles in genetically susceptible hosts. Insights into the dietary-immune-microbial nexus may improve understanding for recurrent inflammatory episodes in susceptible patient populations and opportunities for practical therapeutics to restore immune and microbial homeostasis.


Assuntos
Antioxidantes/farmacologia , Doenças Inflamatórias Intestinais/dietoterapia , Mucosa Intestinal/efeitos dos fármacos , Micronutrientes/farmacologia , Animais , Peso Corporal/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Microbioma Gastrointestinal , Predisposição Genética para Doença , Imunoglobulinas/metabolismo , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/microbiologia , Interleucina-10/genética , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Selênio/farmacologia , Linfócitos T/efeitos dos fármacos , Vitaminas/farmacologia
13.
Sci Rep ; 7(1): 10411, 2017 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-28874832

RESUMO

Recent evidence suggests the commensal microbiome regulates host immunity and influences brain function; findings that have ramifications for neurodegenerative diseases. In the context of Alzheimer's disease (AD), we previously reported that perturbations in microbial diversity induced by life-long combinatorial antibiotic (ABX) selection pressure in the APPSWE/PS1ΔE9 mouse model of amyloidosis is commensurate with reductions in amyloid-ß (Aß) plaque pathology and plaque-localised gliosis. Considering microbiota-host interactions, specifically during early post-natal development, are critical for immune- and neuro-development we now examine the impact of microbial community perturbations induced by acute ABX exposure exclusively during this period in APPSWE/PS1ΔE9 mice. We show that early post-natal (P) ABX treatment (P14-P21) results in long-term alterations of gut microbial genera (predominantly Lachnospiraceae and S24-7) and reduction in brain Aß deposition in aged APPSWE/PS1ΔE9 mice. These mice exhibit elevated levels of blood- and brain-resident Foxp3+ T-regulatory cells and display an alteration in the inflammatory milieu of the serum and cerebrospinal fluid. Finally, we confirm that plaque-localised microglia and astrocytes are reduced in ABX-exposed mice. These findings suggest that ABX-induced microbial diversity perturbations during post-natal stages of development coincide with altered host immunity mechanisms and amyloidosis in a murine model of AD.


Assuntos
Doença de Alzheimer/etiologia , Precursor de Proteína beta-Amiloide/genética , Amiloidose/genética , Antibacterianos/farmacologia , Microbiota/efeitos dos fármacos , Doença de Alzheimer/metabolismo , Doença de Alzheimer/patologia , Precursor de Proteína beta-Amiloide/metabolismo , Amiloidose/metabolismo , Amiloidose/patologia , Animais , Biodiversidade , Biomarcadores , Encéfalo/metabolismo , Encéfalo/patologia , Modelos Animais de Doenças , Microbioma Gastrointestinal , Mediadores da Inflamação/metabolismo , Masculino , Metagenoma , Metagenômica/métodos , Camundongos , Camundongos Transgênicos , Neuroimunomodulação/efeitos dos fármacos , Neuroimunomodulação/genética , Neuroimunomodulação/imunologia , Placa Amiloide/etiologia , Placa Amiloide/metabolismo , Placa Amiloide/patologia , RNA Ribossômico 16S/genética
14.
Science ; 356(6333): 44-50, 2017 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-28386004

RESUMO

Viral infections have been proposed to elicit pathological processes leading to the initiation of T helper 1 (TH1) immunity against dietary gluten and celiac disease (CeD). To test this hypothesis and gain insights into mechanisms underlying virus-induced loss of tolerance to dietary antigens, we developed a viral infection model that makes use of two reovirus strains that infect the intestine but differ in their immunopathological outcomes. Reovirus is an avirulent pathogen that elicits protective immunity, but we discovered that it can nonetheless disrupt intestinal immune homeostasis at inductive and effector sites of oral tolerance by suppressing peripheral regulatory T cell (pTreg) conversion and promoting TH1 immunity to dietary antigen. Initiation of TH1 immunity to dietary antigen was dependent on interferon regulatory factor 1 and dissociated from suppression of pTreg conversion, which was mediated by type-1 interferon. Last, our study in humans supports a role for infection with reovirus, a seemingly innocuous virus, in triggering the development of CeD.


Assuntos
Antígenos/imunologia , Doença Celíaca/imunologia , Doença Celíaca/virologia , Glutens/imunologia , Inflamação/virologia , Infecções por Reoviridae/complicações , Infecções por Reoviridae/imunologia , Células Th1/imunologia , Animais , Dieta/efeitos adversos , Modelos Animais de Doenças , Engenharia Genética , Humanos , Tolerância Imunológica , Inflamação/imunologia , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/imunologia , Interferon Tipo I/genética , Interferon Tipo I/imunologia , Intestinos/imunologia , Intestinos/patologia , Intestinos/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor de Interferon alfa e beta/genética , Reoviridae/genética
15.
ISME J ; 11(1): 15-30, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27648810

RESUMO

Dysbiosis resulting in gut-microbiome alterations with reduced butyrate production are thought to disrupt intestinal immune homeostasis and promote complex immune disorders. However, whether and how dysbiosis develops before the onset of overt pathology remains poorly defined. Interleukin-15 (IL-15) is upregulated in distressed tissue and its overexpression is thought to predispose susceptible individuals to and have a role in the pathogenesis of celiac disease and inflammatory bowel disease (IBD). Although the immunological roles of IL-15 have been largely studied, its potential impact on the microbiota remains unexplored. Analysis of 16S ribosomal RNA-based inventories of bacterial communities in mice overexpressing IL-15 in the intestinal epithelium (villin-IL-15 transgenic (v-IL-15tg) mice) shows distinct changes in the composition of the intestinal bacteria. Although some alterations are specific to individual intestinal compartments, others are found across the ileum, cecum and feces. In particular, IL-15 overexpression restructures the composition of the microbiota with a decrease in butyrate-producing bacteria that is associated with a reduction in luminal butyrate levels across all intestinal compartments. Fecal microbiota transplant experiments of wild-type and v-IL-15tg microbiota into germ-free mice further indicate that diminishing butyrate concentration observed in the intestinal lumen of v-IL-15tg mice is the result of intrinsic alterations in the microbiota induced by IL-15. This reconfiguration of the microbiota is associated with increased susceptibility to dextran sodium sulfate-induced colitis. Altogether, this study reveals that IL-15 impacts butyrate-producing bacteria and lowers butyrate levels in the absence of overt pathology, which represent events that precede and promote intestinal inflammatory diseases.


Assuntos
Bactérias/metabolismo , Butiratos/metabolismo , Colite/metabolismo , Disbiose/microbiologia , Microbioma Gastrointestinal , Interleucina-15/metabolismo , Intestinos/microbiologia , Animais , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Colite/genética , Colite/microbiologia , Colite/terapia , Suscetibilidade a Doenças , Disbiose/genética , Disbiose/metabolismo , Transplante de Microbiota Fecal , Fezes/microbiologia , Feminino , Vida Livre de Germes , Humanos , Interleucina-15/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
17.
Nature ; 507(7493): 508-12, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24553136

RESUMO

Tumour metastasis is the primary cause of mortality in cancer patients and remains the key challenge for cancer therapy. New therapeutic approaches to block inhibitory pathways of the immune system have renewed hopes for the utility of such therapies. Here we show that genetic deletion of the E3 ubiquitin ligase Cbl-b (casitas B-lineage lymphoma-b) or targeted inactivation of its E3 ligase activity licenses natural killer (NK) cells to spontaneously reject metastatic tumours. The TAM tyrosine kinase receptors Tyro3, Axl and Mer (also known as Mertk) were identified as ubiquitylation substrates for Cbl-b. Treatment of wild-type NK cells with a newly developed small molecule TAM kinase inhibitor conferred therapeutic potential, efficiently enhancing anti-metastatic NK cell activity in vivo. Oral or intraperitoneal administration using this TAM inhibitor markedly reduced murine mammary cancer and melanoma metastases dependent on NK cells. We further report that the anticoagulant warfarin exerts anti-metastatic activity in mice via Cbl-b/TAM receptors in NK cells, providing a molecular explanation for a 50-year-old puzzle in cancer biology. This novel TAM/Cbl-b inhibitory pathway shows that it might be possible to develop a 'pill' that awakens the innate immune system to kill cancer metastases.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Células Matadoras Naturais/imunologia , Neoplasias Mamárias Experimentais/patologia , Melanoma Experimental/patologia , Metástase Neoplásica/imunologia , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Anticoagulantes/farmacologia , Anticoagulantes/uso terapêutico , Feminino , Células Matadoras Naturais/efeitos dos fármacos , Células Matadoras Naturais/metabolismo , Masculino , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/imunologia , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Metástase Neoplásica/tratamento farmacológico , Metástase Neoplásica/prevenção & controle , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-cbl/deficiência , Proteínas Proto-Oncogênicas c-cbl/genética , Receptores Proteína Tirosina Quinases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/deficiência , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Varfarina/farmacologia , Varfarina/uso terapêutico , c-Mer Tirosina Quinase , Receptor Tirosina Quinase Axl
18.
J Mol Cell Biol ; 5(6): 358-68, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23709694

RESUMO

T cell-intrinsic transforming growth factor ß (TGFß) receptor signaling plays an essential role in controlling immune responses. The RING-type E3 ligase Cbl-b has been shown to mediate the sensitivity of T cells to TGFß; however, the mechanism underlying this process is unknown. This study shows that SMAD7, an established negative regulator of TGFß receptor (TGFßR) signaling, is a key downstream effector target of Cbl-b. SMAD7 protein levels, but not SMAD7 mRNA levels, are upregulated in cblb(-/-) T cells. Cbl-b directly interacts with and ubiquitinates SMAD7, suggesting that Cbl-b posttranscriptionally regulates SMAD7. In support of this notion, concomitant genetic loss of SMAD7 in cblb(-/-) mice restored TGFß sensitivity on T cell cytokine responses and abrogated the tumor rejection phenotype of cblb(-/-) mice. These results demonstrate an essential and non-redundant role for Cbl-b in controlling TGFßR signaling by directly targeting SMAD7 for degradation during T cell responses in vitro and in vivo.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Proteína Smad7/metabolismo , Linfócitos T/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Imunoprecipitação da Cromatina , Interferon gama/metabolismo , Camundongos , Camundongos Knockout , Microscopia Confocal , Proteínas Proto-Oncogênicas c-cbl/genética , Proteína Smad7/genética , Proteínas com Domínio T/genética , Proteínas com Domínio T/metabolismo , Fator de Crescimento Transformador beta/genética , Ubiquitinação
19.
Oncoimmunology ; 2(2): e22893, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23525844

RESUMO

The adoptive transfer of synthetic siRNA-mediated cblb-depleted autologous CD8+ T cells acts as a potent adjuvant for dendritic cell (DC) vaccination and provides a significant therapeutic benefit. Our proof-of-concept study validates the strategy of inhibiting CBLB as a rational approach to augment the effectiveness of adoptively transferred immune cells.

20.
Immunity ; 38(1): 41-52, 2013 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-23290522

RESUMO

Transforming growth-factor ß (TGFß) has been implicated in T helper 17 (Th17) cell biology and in triggering expression of interleukin-17A (IL-17A), which is a key Th17 cell cytokine. Deregulated TGFß receptor (TGFßR) signaling has been implicated in Th17-cell-mediated autoimmune pathogenesis. Nevertheless, the full molecular mechanisms involved in the activation of the TGFßR pathway in driving IL-17A expression remain unknown. Here, we identified protein kinase C α (PKCα) as a signaling intermediate specific to the Th17 cell subset in the activation of TGFßRI. We have shown that PKCα physically interacts and functionally cooperates with TGFßRI to promote robust SMAD2-3 activation. Furthermore, PKCα-deficient (Prkca(-/-)) cells demonstrated a defect in SMAD-dependent IL-2 suppression, as well as decreased STAT3 DNA binding within the Il17a promoter. Consistently, Prkca(-/-) cells failed to mount appropriate IL-17A, but not IL-17F, responses in vitro and were resistant to induction of Th17-cell-dependent experimental autoimmune encephalomyelitis in vivo.


Assuntos
Interleucina-17/metabolismo , Proteína Quinase C-alfa/metabolismo , Células Th17/imunologia , Células Th17/metabolismo , Animais , Encefalomielite Autoimune Experimental/induzido quimicamente , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/imunologia , Regulação da Expressão Gênica , Interleucina-17/imunologia , Camundongos , Camundongos Knockout , Glicoproteína Mielina-Oligodendrócito/efeitos adversos , Fragmentos de Peptídeos/efeitos adversos , Proteína Quinase C-alfa/genética , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteínas Smad/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA