Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Front Immunol ; 13: 768076, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35185874

RESUMO

The gastrointestinal tract represents one of the largest body surfaces that is exposed to the outside world. It is the only mucosal surface that is required to simultaneously recognize and defend against pathogens, while allowing nutrients containing foreign antigens to be tolerated and absorbed. It differentiates between these foreign substances through a complex system of pattern recognition receptors expressed on the surface of the intestinal epithelial cells as well as the underlying immune cells. These immune cells actively sample and evaluate microbes and other particles that pass through the lumen of the gut. This local sensing system is part of a broader distributed signaling system that is connected to the rest of the body through the enteric nervous system, the immune system, and the metabolic system. While local tissue homeostasis is maintained by commensal bacteria that colonize the gut, colonization itself may not be required for the activation of distributed signaling networks that can result in modulation of peripheral inflammation. Herein, we describe the ability of a gut-restricted strain of commensal bacteria to drive systemic anti-inflammatory effects in a manner that does not rely upon its ability to colonize the gastrointestinal tract or alter the mucosal microbiome. Orally administered EDP1867, a gamma-irradiated strain of Veillonella parvula, rapidly transits through the murine gut without colonization or alteration of the background microbiome flora. In murine models of inflammatory disease including delayed-type hypersensitivity (DTH), atopic dermatitis, psoriasis, and experimental autoimmune encephalomyelitis (EAE), treatment with EDP1867 resulted in significant reduction in inflammation and immunopathology. Ex vivo cytokine analyses revealed that EDP1867 treatment diminished production of pro-inflammatory cytokines involved in inflammatory cascades. Furthermore, blockade of lymphocyte migration to the gut-associated lymphoid tissues impaired the ability of EDP1867 to resolve peripheral inflammation, supporting the hypothesis that circulating immune cells are responsible for promulgating the signals from the gut to peripheral tissues. Finally, we show that adoptively transferred T cells from EDP1867-treated mice inhibit inflammation induced in recipient mice. These results demonstrate that an orally-delivered, non-viable strain of commensal bacteria can mediate potent anti-inflammatory effects in peripheral tissues through transient occupancy of the gastrointestinal tract, and support the development of non-living bacterial strains for therapeutic applications.


Assuntos
Antibacterianos/farmacologia , Bactérias/imunologia , Citocinas/metabolismo , Microbioma Gastrointestinal/efeitos dos fármacos , Inflamação/imunologia , Animais , Bactérias/efeitos dos fármacos , Bactérias/crescimento & desenvolvimento , Células Epiteliais/efeitos dos fármacos , Feminino , Humanos , Imunidade nas Mucosas , Inflamação/etiologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Simbiose , Linfócitos T/metabolismo
2.
Sci Adv ; 7(40): eabj2485, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34597140

RESUMO

Adaptive changes in lysosomal capacity are driven by the transcription factors TFEB and TFE3 in response to increased autophagic flux and endolysosomal stress, yet the molecular details of their activation are unclear. LC3 and GABARAP members of the ATG8 protein family are required for selective autophagy and sensing perturbation within the endolysosomal system. Here, we show that during the conjugation of ATG8 to single membranes (CASM), Parkin-dependent mitophagy, and Salmonella-induced xenophagy, the membrane conjugation of GABARAP, but not LC3, is required for activation of TFEB/TFE3 to control lysosomal capacity. GABARAP directly binds to a previously unidentified LC3-interacting motif (LIR) in the FLCN/FNIP tumor suppressor complex and mediates sequestration to GABARAP-conjugated membrane compartments. This disrupts FLCN/FNIP GAP function toward RagC/D, resulting in impaired substrate-specific mTOR-dependent phosphorylation of TFEB. Thus, the GABARAP-FLCN/FNIP-TFEB axis serves as a molecular sensor that coordinates lysosomal homeostasis with perturbations and cargo flux within the autophagy-lysosomal network.

3.
J Immunol ; 205(2): 414-424, 2020 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-32522834

RESUMO

Genome-wide association studies have identified common genetic variants impacting human diseases; however, there are indications that the functional consequences of genetic polymorphisms can be distinct depending on cell type-specific contexts, which produce divergent phenotypic outcomes. Thus, the functional impact of genetic variation and the underlying mechanisms of disease risk are modified by cell type-specific effects of genotype on pathological phenotypes. In this study, we extend these concepts to interrogate the interdependence of cell type- and stimulation-specific programs influenced by the core autophagy gene Atg16L1 and its T300A coding polymorphism identified by genome-wide association studies as linked with increased risk of Crohn's disease. We applied a stimulation-based perturbational profiling approach to define Atg16L1 T300A phenotypes in dendritic cells and T lymphocytes. Accordingly, we identified stimulus-specific transcriptional signatures revealing T300A-dependent functional phenotypes that mechanistically link inflammatory cytokines, IFN response genes, steroid biosynthesis, and lipid metabolism in dendritic cells and iron homeostasis and lysosomal biogenesis in T lymphocytes. Collectively, these studies highlight the combined effects of Atg16L1 genetic variation and stimulatory context on immune function.


Assuntos
Proteínas Relacionadas à Autofagia/metabolismo , Doença de Crohn/metabolismo , Células Dendríticas/fisiologia , Genótipo , Linfócitos T/fisiologia , Animais , Proteínas Relacionadas à Autofagia/genética , Células Cultivadas , Doença de Crohn/genética , Predisposição Genética para Doença , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Especificidade de Órgãos , Fenótipo , Polimorfismo Genético , Risco , Ativação Transcricional
4.
Cell Rep ; 24(4): 838-850, 2018 07 24.
Artigo em Inglês | MEDLINE | ID: mdl-30044981

RESUMO

Phagocyte microbiocidal mechanisms and inflammatory cytokine production are temporally coordinated, although their respective interdependencies remain incompletely understood. Here, we identify a nitric-oxide-mediated antioxidant response as a negative feedback regulator of inflammatory cytokine production in phagocytes. In this context, Keap1 functions as a cellular redox sensor that responds to elevated reactive nitrogen intermediates by eliciting an adaptive transcriptional program controlled by Nrf2 and comprised of antioxidant genes, including Prdx5. We demonstrate that engaging the antioxidant response is sufficient to suppress Toll-like receptor (TLR)-induced cytokine production in dendritic cells and that Prdx5 is required for attenuation of inflammatory cytokine production. Collectively, these findings delineate the reciprocal regulation of inflammation and cellular redox systems in myeloid cells.


Assuntos
Óxido Nítrico/metabolismo , Peroxirredoxinas/metabolismo , Fagócitos/metabolismo , Animais , Células da Medula Óssea/metabolismo , Citocinas/biossíntese , Retroalimentação Fisiológica , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/biossíntese , Transdução de Sinais
5.
Oncotarget ; 6(34): 36535-50, 2015 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-26431492

RESUMO

Many host-factors are inducibly expressed during the development of inflammatory bowel disease (IBD), each having their unique properties, such as immune activation, bacterial clearance, and tissue repair/remodeling. Dysregulation/imbalance of these factors may have pathogenic effects that can contribute to colitis-associated cancer (CAC). Previous reports showed that IBD patients inducibly express colonic chitinase 3-like 1 (CHI3L1) that is further upregulated during CAC development. However, little is known about the direct pathogenic involvement of CHI3L1 in vivo. Here we demonstrate that CHI3L1 (aka Brp39) knockout (KO) mice treated with azoxymethane (AOM)/dextran sulphate sodium (DSS) developed severe colitis but lesser incidence of CAC as compared to that in wild-type (WT) mice. Highest CHI3L1 expression was found during the chronic phase of colitis, rather than the acute phase, and is essential to promote intestinal epithelial cell (IEC) proliferation in vivo. This CHI3L1-mediated cell proliferation/survival involves partial downregulation of the pro-apoptotic S100A9 protein that is highly expressed during the acute phase of colitis, by binding to the S100A9 receptor, RAGE (Receptor for Advanced Glycation End products). This interaction disrupts the S100A9-associated expression positive feedback loop during early immune activation, creating a CHI3L1hi S100A9low colonic environment, especially in the later phase of colitis, which promotes cell proliferation/survival of both normal IECs and tumor cells.


Assuntos
Calgranulina B/metabolismo , Proteína 1 Semelhante à Quitinase-3/metabolismo , Neoplasias do Colo/metabolismo , Animais , Proliferação de Células/fisiologia , Proteína 1 Semelhante à Quitinase-3/biossíntese , Proteína 1 Semelhante à Quitinase-3/genética , Doença Crônica , Colite/enzimologia , Colite/genética , Colite/metabolismo , Colite/patologia , Neoplasias do Colo/enzimologia , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Humanos , Mucosa Intestinal/enzimologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Subunidade beta da Proteína Ligante de Cálcio S100/metabolismo
6.
Nat Commun ; 6: 7838, 2015 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-26194095

RESUMO

The phagocyte oxidative burst, mediated by Nox2 NADPH oxidase-derived reactive oxygen species, confers host defense against a broad spectrum of bacterial and fungal pathogens. Loss-of-function mutations that impair function of the Nox2 complex result in a life-threatening immunodeficiency, and genetic variants of Nox2 subunits have been implicated in pathogenesis of inflammatory bowel disease (IBD). Thus, alterations in the oxidative burst can profoundly impact host defense, yet little is known about regulatory mechanisms that fine-tune this response. Here we report the discovery of regulatory nodes controlling oxidative burst by functional screening of genes within loci linked to human inflammatory disease. Implementing a multi-omics approach, we define transcriptional, metabolic and ubiquitin-cycling nodes controlled by Rbpj, Pfkl and Rnf145, respectively. Furthermore, we implicate Rnf145 in proteostasis of the Nox2 complex by endoplasmic reticulum-associated degradation. Consequently, ablation of Rnf145 in murine macrophages enhances bacterial clearance, and rescues the oxidative burst defects associated with Ncf4 haploinsufficiency.


Assuntos
Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , NADPH Oxidases/metabolismo , Fagócitos/metabolismo , Explosão Respiratória , Animais , Sequência de Bases , Linhagem Celular , Genômica , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/metabolismo , Camundongos , Dados de Sequência Molecular , NADPH Oxidase 2 , Fosfofrutoquinase-1/metabolismo , Staphylococcus aureus
7.
Cell Rep ; 11(12): 1905-18, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26095365

RESUMO

The polymorphism ATG16L1 T300A, associated with increased risk of Crohn's disease, impairs pathogen defense mechanisms including selective autophagy, but specific pathway interactions altered by the risk allele remain unknown. Here, we use perturbational profiling of human peripheral blood cells to reveal that CLEC12A is regulated in an ATG16L1-T300A-dependent manner. Antibacterial autophagy is impaired in CLEC12A-deficient cells, and this effect is exacerbated in the presence of the ATG16L1(∗)300A risk allele. Clec12a(-/-) mice are more susceptible to Salmonella infection, supporting a role for CLEC12A in antibacterial defense pathways in vivo. CLEC12A is recruited to sites of bacterial entry, bacteria-autophagosome complexes, and sites of sterile membrane damage. Integrated genomics identified a functional interaction between CLEC12A and an E3-ubiquitin ligase complex that functions in antibacterial autophagy. These data identify CLEC12A as early adaptor molecule for antibacterial autophagy and highlight perturbational profiling as a method to elucidate defense pathways in complex genetic disease.


Assuntos
Proteínas de Transporte/genética , Doença de Crohn/genética , Lectinas Tipo C/genética , Receptores Mitogênicos/genética , Infecções por Salmonella/genética , Alelos , Animais , Autofagia/genética , Proteínas Relacionadas à Autofagia , Doença de Crohn/microbiologia , Doença de Crohn/patologia , Predisposição Genética para Doença , Genômica , Humanos , Lectinas Tipo C/biossíntese , Camundongos , Receptores Mitogênicos/biossíntese , Fatores de Risco , Salmonella/patogenicidade , Infecções por Salmonella/microbiologia
8.
Eur J Clin Microbiol Infect Dis ; 34(5): 963-974, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25579795

RESUMO

The induction of host defense against Candida species is initiated by recognition of the fungi by pattern recognition receptors and activation of downstream pathways that produce inflammatory mediators essential for infection clearance. In this study, we present complementary evidence based on transcriptome analysis, genetics, and immunological studies in knockout mice and humans that the cytosolic RIG-I-like receptor MDA5 (IFIH1) has an important role in the host defense against C. albicans. Firstly, IFIH1 expression in macrophages is specifically induced by invasive C. albicans hyphae, and patients suffering from chronic mucocutaneous candidiasis (CMC) express lower levels of MDA5 than healthy controls. Secondly, there is a strong association between missense variants in the IFIH1 gene (rs1990760 and rs3747517) and susceptibility to systemic Candida infections. Thirdly, cells from Mda5 knockout mice and human peripheral blood mononuclear cells (PBMCs) with different IFIH1 genotypes display an altered cytokine response to C. albicans. These data strongly suggest that MDA5 is involved in immune responses to Candida infection. As a receptor for viral RNA, MDA5 until now has been linked to antiviral host defense, but these novel studies show unexpected effects in antifungal immunity as well. Future studies are warranted to explore the potential of MDA5 as a novel target for immunotherapeutic strategies.


Assuntos
Candida/imunologia , Candidemia/imunologia , RNA Helicases DEAD-box/metabolismo , Adulto , Animais , Células Cultivadas , Estudos de Coortes , RNA Helicases DEAD-box/deficiência , Suscetibilidade a Doenças , Humanos , Helicase IFIH1 Induzida por Interferon , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/microbiologia , Camundongos Knockout , Polimorfismo de Nucleotídeo Único
9.
PLoS Pathog ; 10(10): e1004485, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25356988

RESUMO

The anti-tuberculosis-vaccine Bacillus Calmette-Guérin (BCG) is the most widely used vaccine in the world. In addition to its effects against tuberculosis, BCG vaccination also induces non-specific beneficial effects against certain forms of malignancy and against infections with unrelated pathogens. It has been recently proposed that the non-specific effects of BCG are mediated through epigenetic reprogramming of monocytes, a process called trained immunity. In the present study we demonstrate that autophagy contributes to trained immunity induced by BCG. Pharmacologic inhibition of autophagy blocked trained immunity induced in vitro by stimuli such as ß-glucans or BCG. Single nucleotide polymorphisms (SNPs) in the autophagy genes ATG2B (rs3759601) and ATG5 (rs2245214) influenced both the in vitro and in vivo training effect of BCG upon restimulation with unrelated bacterial or fungal stimuli. Furthermore, pharmacologic or genetic inhibition of autophagy blocked epigenetic reprogramming of monocytes at the level of H3K4 trimethylation. Finally, we demonstrate that rs3759601 in ATG2B correlates with progression and recurrence of bladder cancer after BCG intravesical instillation therapy. These findings identify a key role of autophagy for the nonspecific protective effects of BCG.


Assuntos
Autofagia , Vacina BCG/uso terapêutico , Mycobacterium bovis/imunologia , Polimorfismo de Nucleotídeo Único , Neoplasias da Bexiga Urinária/tratamento farmacológico , Adjuvantes Imunológicos/administração & dosagem , Adjuvantes Imunológicos/uso terapêutico , Administração Intravesical , Antineoplásicos/administração & dosagem , Antineoplásicos/uso terapêutico , Autofagia/genética , Autofagia/imunologia , Proteína 5 Relacionada à Autofagia , Proteínas Relacionadas à Autofagia , Vacina BCG/administração & dosagem , Citocinas/metabolismo , Humanos , Estimativa de Kaplan-Meier , Proteínas Associadas aos Microtúbulos/genética , Monócitos/imunologia , Recidiva Local de Neoplasia , Neoplasias da Bexiga Urinária/imunologia , Neoplasias da Bexiga Urinária/virologia , Vacinação , Proteínas de Transporte Vesicular/genética , beta-Glucanas/metabolismo
10.
Science ; 345(6204): 1250684, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25258083

RESUMO

Epigenetic reprogramming of myeloid cells, also known as trained immunity, confers nonspecific protection from secondary infections. Using histone modification profiles of human monocytes trained with the Candida albicans cell wall constituent ß-glucan, together with a genome-wide transcriptome, we identified the induced expression of genes involved in glucose metabolism. Trained monocytes display high glucose consumption, high lactate production, and a high ratio of nicotinamide adenine dinucleotide (NAD(+)) to its reduced form (NADH), reflecting a shift in metabolism with an increase in glycolysis dependent on the activation of mammalian target of rapamycin (mTOR) through a dectin-1-Akt-HIF-1α (hypoxia-inducible factor-1α) pathway. Inhibition of Akt, mTOR, or HIF-1α blocked monocyte induction of trained immunity, whereas the adenosine monophosphate-activated protein kinase activator metformin inhibited the innate immune response to fungal infection. Mice with a myeloid cell-specific defect in HIF-1α were unable to mount trained immunity against bacterial sepsis. Our results indicate that induction of aerobic glycolysis through an Akt-mTOR-HIF-1α pathway represents the metabolic basis of trained immunity.


Assuntos
Epigênese Genética , Glicólise/imunologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Imunidade Inata/genética , Memória Imunológica/genética , Monócitos/imunologia , Serina-Treonina Quinases TOR/metabolismo , Aerobiose/imunologia , Animais , Candida albicans/imunologia , Candidíase/imunologia , Candidíase/metabolismo , Modelos Animais de Doenças , Feminino , Glucose/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Sepse/genética , Sepse/imunologia , Sepse/metabolismo , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/metabolismo , Staphylococcus aureus , Serina-Treonina Quinases TOR/genética , Transcriptoma , beta-Glucanas/imunologia
11.
Science ; 345(6204): 1251086, 2014 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-25258085

RESUMO

Monocyte differentiation into macrophages represents a cornerstone process for host defense. Concomitantly, immunological imprinting of either tolerance or trained immunity determines the functional fate of macrophages and susceptibility to secondary infections. We characterized the transcriptomes and epigenomes in four primary cell types: monocytes and in vitro-differentiated naïve, tolerized, and trained macrophages. Inflammatory and metabolic pathways were modulated in macrophages, including decreased inflammasome activation, and we identified pathways functionally implicated in trained immunity. ß-glucan training elicits an exclusive epigenetic signature, revealing a complex network of enhancers and promoters. Analysis of transcription factor motifs in deoxyribonuclease I hypersensitive sites at cell-type-specific epigenetic loci unveiled differentiation and treatment-specific repertoires. Altogether, we provide a resource to understand the epigenetic changes that underlie innate immunity in humans.


Assuntos
Diferenciação Celular/genética , Epigênese Genética , Imunidade Inata/genética , Macrófagos/citologia , Monócitos/citologia , Animais , Sítios de Ligação/genética , Desoxirribonuclease I/química , Impressão Genômica , Humanos , Memória Imunológica , Inflamassomos/genética , Inflamassomos/imunologia , Macrófagos/imunologia , Camundongos , Monócitos/imunologia , Fatores de Transcrição/metabolismo , beta-Glucanas/imunologia
12.
Cell Rep ; 1(4): 317-24, 2012 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-22545247

RESUMO

Monocytes serve as a central defense system against infection and injury but can also promote pathological inflammatory responses. Considering the evidence that monocytes exist in at least two subsets committed to divergent functions, we investigated whether distinct factors regulate the balance between monocyte subset responses in vivo. We identified a microRNA (miRNA), miR-146a, which is differentially regulated both in mouse (Ly-6C(hi)/Ly-6C(lo)) and human (CD14(hi)/CD14(lo)CD16(+)) monocyte subsets. The single miRNA controlled the amplitude of the Ly-6C(hi) monocyte response during inflammatory challenge whereas it did not affect Ly-6C(lo) cells. miR-146a-mediated regulation was cell-intrinsic and depended on Relb, a member of the noncanonical NF-κB/Rel family, which we identified as a direct miR-146a target. These observations not only provide mechanistic insights into the molecular events that regulate responses mediated by committed monocyte precursor populations but also identify targets for manipulating Ly-6C(hi) monocyte responses while sparing Ly-6Clo monocyte activity.


Assuntos
MicroRNAs/fisiologia , Monócitos/fisiologia , Fator de Transcrição RelB/fisiologia , Animais , Movimento Celular , Proliferação de Células , Regulação da Expressão Gênica , Humanos , Camundongos , Monócitos/imunologia , Monócitos/metabolismo , Fator de Transcrição RelB/metabolismo
13.
Nat Cell Biol ; 13(12): 1437-42, 2011 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-22037414

RESUMO

Caspase 8 initiates apoptosis downstream of TNF death receptors by undergoing autocleavage and processing the executioner caspase 3 (ref. 1). However, the dominant function of caspase 8 is to transmit a pro-survival signal that suppresses programmed necrosis (or necroptosis) mediated by RIPK1 and RIPK3 (refs 2-6) during embryogenesis and haematopoiesis(7-9). Suppression of necrotic cell death by caspase 8 requires its catalytic activity but not the autocleavage essential for apoptosis(10); however, the key substrate processed by caspase 8 to block necrosis has been elusive. A key substrate must meet three criteria: it must be essential for programmed necrosis; it must be cleaved by caspase 8 in situations where caspase 8 is blocking necrosis; and mutation of the caspase 8 processing site on the substrate should convert a pro-survival response to necrotic death without the need for caspase 8 inhibition. We now identify CYLD as a substrate for caspase 8 that satisfies these criteria. Following TNF stimulation, caspase 8 cleaves CYLD to generate a survival signal. In contrast, loss of caspase 8 prevented CYLD degradation, resulting in necrotic death. A CYLD substitution mutation at Asp 215 that cannot be cleaved by caspase 8 switches cell survival to necrotic cell death in response to TNF.


Assuntos
Caspase 8/metabolismo , Embrião de Mamíferos/patologia , Fibroblastos/patologia , Proteínas Supressoras de Tumor/metabolismo , Animais , Caspase 8/genética , Sobrevivência Celular/genética , Células Cultivadas , Enzima Desubiquitinante CYLD , Embrião de Mamíferos/citologia , Embrião de Mamíferos/enzimologia , Feminino , Fibroblastos/citologia , Fibroblastos/enzimologia , Células HEK293 , Humanos , Células Jurkat , Camundongos , Necrose , Processamento de Proteína Pós-Traducional/genética , Transdução de Sinais/genética , Proteínas Supressoras de Tumor/genética
14.
Autophagy ; 7(9): 1082-4, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21606681

RESUMO

The leucine-rich repeats (LRR)-containing domain is evolutionarily conserved in many proteins associated with innate immunity in plants, invertebrates and vertebrates. Serving as a first line of defense, the innate immune response is initiated through the sensing of pathogen-associated molecular patterns (PAMPs). In plants, NBS (nucleotide-binding site)-LRR proteins provide recognition of pathogen products of avirulence (AVR) genes. LRRs also promote interaction between LRR proteins as observed in receptor-coreceptor complexes. In mammals, toll-like receptors (TLRs) and NOD-like receptors (NLRs) through their LRR domain, sense molecular determinants from a structurally diverse set of bacterial, fungal, parasite and viral-derived components. In humans, at least 34 LRR proteins are implicated in diseases. Most LRR domains consist of 2-45 leucine-rich repeats, with each repeat about 20-30 residues long. Structurally, LRR domains adopt an arc or horseshoe shape, with the concave face consisting of parallel ß-strands and the convex face representing a more variable region of secondary structures including helices. Apart from the TLRs and NLRs, most of the 375 human LRR proteins remain uncharacterized functionally. We incorporated computational and functional analyses to facilitate multifaceted insights into human LRR proteins and outline a few approaches here.


Assuntos
Imunidade/imunologia , Proteínas/metabolismo , Receptores de Reconhecimento de Padrão/imunologia , Transdução de Sinais/imunologia , Animais , Evolução Molecular , Humanos , Proteínas de Repetições Ricas em Leucina , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Modelos Biológicos , Proteínas/genética , Interferência de RNA
15.
Proc Natl Acad Sci U S A ; 108 Suppl 1: 4631-8, 2011 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20616063

RESUMO

In innate immune sensing, the detection of pathogen-associated molecular patterns by recognition receptors typically involve leucine-rich repeats (LRRs). We provide a categorization of 375 human LRR-containing proteins, almost half of which lack other identifiable functional domains. We clustered human LRR proteins by first assigning LRRs to LRR classes and then grouping the proteins based on these class assignments, revealing several of the resulting protein groups containing a large number of proteins with certain non-LRR functional domains. In particular, a statistically significant number of LRR proteins in the typical (T) and bacterial + typical (S+T) categories have transmembrane domains, whereas most of the LRR proteins in the cysteine-containing (CC) category contain an F-box domain (which mediates interactions with the E3 ubiquitin ligase complex). Furthermore, by examining the evolutionary profiles of the LRR proteins, we identified a subset of LRR proteins exhibiting strong conservation in fungi and an enrichment for "nucleic acid-binding" function. Expression analysis of LRR genes identifies a subset of pathogen-responsive genes in human primary macrophages infected with pathogenic bacteria. Using functional RNAi, we show that MFHAS1 regulates Toll-like receptor (TLR)-dependent signaling. By using protein interaction network analysis followed by functional RNAi, we identified LRSAM1 as a component of the antibacterial autophagic response.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Evolução Molecular , Imunidade Inata/genética , Proteínas Oncogênicas/metabolismo , Proteínas/genética , Proteínas/imunologia , Transdução de Sinais/genética , Análise por Conglomerados , Biologia Computacional/métodos , Perfilação da Expressão Gênica , Estudo de Associação Genômica Ampla , Humanos , Imunidade Inata/imunologia , Proteínas de Repetições Ricas em Leucina , Macrófagos/metabolismo , Macrófagos/microbiologia , Proteínas/classificação , Interferência de RNA , Receptores Toll-Like/metabolismo
16.
Dev Cell ; 18(6): 1041-52, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20627085

RESUMO

Autophagy is a cellular catabolic mechanism that plays an essential function in protecting multicellular eukaryotes from neurodegeneration, cancer, and other diseases. However, we still know very little about mechanisms regulating autophagy under normal homeostatic conditions when nutrients are not limiting. In a genome-wide human siRNA screen, we demonstrate that under normal nutrient conditions upregulation of autophagy requires the type III PI3 kinase, but not inhibition of mTORC1, the essential negative regulator of starvation-induced autophagy. We show that a group of growth factors and cytokines inhibit the type III PI3 kinase through multiple pathways, including the MAPK-ERK1/2, Stat3, Akt/Foxo3, and CXCR4/GPCR, which are all known to positively regulate cell growth and proliferation. Our study suggests that the type III PI3 kinase integrates diverse signals to regulate cellular levels of autophagy, and that autophagy and cell proliferation may represent two alternative cell fates that are regulated in a mutually exclusive manner.


Assuntos
Autofagia/genética , Estudo de Associação Genômica Ampla/métodos , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/genética , Fatores de Transcrição/genética , Linhagem Celular Tumoral , Proliferação de Células , Alimentos , Humanos , Sistema de Sinalização das MAP Quinases/genética , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos , Proteínas , RNA Interferente Pequeno/genética , Serina-Treonina Quinases TOR
17.
Genes Dev ; 24(14): 1507-18, 2010 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-20634317

RESUMO

Mitochondria serve a critical role in physiology and disease. The genetic basis of mitochondrial regulation in mammalian cells has not yet been detailed. We performed a large-scale RNAi screen to systematically identify genes that affect mitochondrial abundance and function. This screen revealed previously unrecognized roles for >150 proteins in mitochondrial regulation. We report that increased Wnt signals are a potent activator of mitochondrial biogenesis and reactive oxygen species (ROS) generation, leading to DNA damage and acceleration of cellular senescence in primary cells. The signaling protein insulin receptor substrate-1 (IRS-1), shown here to be a transcriptional target of Wnt, is induced in this setting. The increased level of IRS-1 drives activation of mitochondrial biogenesis; furthermore, in insulin-responsive cell types, it enhances insulin signaling, raising the possibility that Wnt proteins may be used to modulate glucose homeostasis. Our results identify a key component of the mitochondrial regulatory apparatus with a potentially important link to metabolic and degenerative disorders.


Assuntos
Insulina/metabolismo , Mitocôndrias/genética , Mitocôndrias/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo , Animais , Senescência Celular , Humanos , Proteínas Substratos do Receptor de Insulina/metabolismo , Camundongos , Proteínas Proto-Oncogênicas c-myc/metabolismo , Espécies Reativas de Oxigênio/metabolismo
18.
Cell ; 141(7): 1135-45, 2010 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-20602997

RESUMO

It is unclear why disease occurs in only a small proportion of persons carrying common risk alleles of disease susceptibility genes. Here we demonstrate that an interaction between a specific virus infection and a mutation in the Crohn's disease susceptibility gene Atg16L1 induces intestinal pathologies in mice. This virus-plus-susceptibility gene interaction generated abnormalities in granule packaging and unique patterns of gene expression in Paneth cells. Further, the response to injury induced by the toxic substance dextran sodium sulfate was fundamentally altered to include pathologies resembling aspects of Crohn's disease. These pathologies triggered by virus-plus-susceptibility gene interaction were dependent on TNFalpha and IFNgamma and were prevented by treatment with broad spectrum antibiotics. Thus, we provide a specific example of how a virus-plus-susceptibility gene interaction can, in combination with additional environmental factors and commensal bacteria, determine the phenotype of hosts carrying common risk alleles for inflammatory disease.


Assuntos
Proteínas de Transporte/genética , Doença de Crohn/genética , Doença de Crohn/virologia , Predisposição Genética para Doença , Íleo/patologia , Norovirus , Animais , Proteínas Relacionadas à Autofagia , Doença de Crohn/patologia , Perfilação da Expressão Gênica , Humanos , Interferon gama/metabolismo , Camundongos , Celulas de Paneth/metabolismo , Celulas de Paneth/virologia , Fator de Necrose Tumoral alfa/metabolismo
19.
Proc Natl Acad Sci U S A ; 106(38): 16410-5, 2009 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-19717417

RESUMO

Hepatitis C virus (HCV) infection is a major cause of end-stage liver disease and a leading indication for liver transplantation. Current therapy fails in many instances and is associated with significant side effects. HCV encodes only a few proteins and depends heavily on host factors for propagation. Each of these host dependencies is a potential therapeutic target. To find host factors required by HCV, we completed a genome-wide small interfering RNA (siRNA) screen using an infectious HCV cell culture system. We applied a two-part screening protocol to allow identification of host factors involved in the complete viral lifecycle. The candidate genes found included known or previously identified factors, and also implicate many additional host cell proteins in HCV infection. To create a more comprehensive view of HCV and host cell interactions, we performed a bioinformatic meta-analysis that integrates our data with those of previous functional and proteomic studies. The identification of host factors participating in the complete HCV lifecycle will both advance our understanding of HCV pathogenesis and illuminate therapeutic targets.


Assuntos
Genoma Humano/genética , Estudo de Associação Genômica Ampla/métodos , Hepacivirus/fisiologia , Interferência de RNA , Antígenos CD/genética , Antígenos CD/metabolismo , Apolipoproteínas E/genética , Apolipoproteínas E/metabolismo , Linhagem Celular Tumoral , Biologia Computacional/métodos , Hepacivirus/genética , Hepacivirus/metabolismo , Interações Hospedeiro-Patógeno , Humanos , RNA Interferente Pequeno/genética , Tetraspanina 28 , Proteínas Virais/genética , Proteínas Virais/metabolismo
20.
PLoS Genet ; 5(6): e1000534, 2009 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19557189

RESUMO

Translating a set of disease regions into insight about pathogenic mechanisms requires not only the ability to identify the key disease genes within them, but also the biological relationships among those key genes. Here we describe a statistical method, Gene Relationships Among Implicated Loci (GRAIL), that takes a list of disease regions and automatically assesses the degree of relatedness of implicated genes using 250,000 PubMed abstracts. We first evaluated GRAIL by assessing its ability to identify subsets of highly related genes in common pathways from validated lipid and height SNP associations from recent genome-wide studies. We then tested GRAIL, by assessing its ability to separate true disease regions from many false positive disease regions in two separate practical applications in human genetics. First, we took 74 nominally associated Crohn's disease SNPs and applied GRAIL to identify a subset of 13 SNPs with highly related genes. Of these, ten convincingly validated in follow-up genotyping; genotyping results for the remaining three were inconclusive. Next, we applied GRAIL to 165 rare deletion events seen in schizophrenia cases (less than one-third of which are contributing to disease risk). We demonstrate that GRAIL is able to identify a subset of 16 deletions containing highly related genes; many of these genes are expressed in the central nervous system and play a role in neuronal synapses. GRAIL offers a statistically robust approach to identifying functionally related genes from across multiple disease regions--that likely represent key disease pathways. An online version of this method is available for public use (http://www.broad.mit.edu/mpg/grail/).


Assuntos
Doença de Crohn/genética , Deleção de Genes , Genômica , Polimorfismo de Nucleotídeo Único , Esquizofrenia/genética , Doença de Crohn/patologia , Bases de Dados Genéticas , Genoma Humano , Estudo de Associação Genômica Ampla , Humanos , Metanálise como Assunto , Esquizofrenia/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA