Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 57(4): 613-631, 2024 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-38599162

RESUMEN

While largely neglected over decades during which adaptive immunity captured most of the attention, innate immune mechanisms have now become central to our understanding of immunology. Innate immunity provides the first barrier to infection in vertebrates, and it is the sole mechanism of host defense in invertebrates and plants. Innate immunity also plays a critical role in maintaining homeostasis, shaping the microbiota, and in disease contexts such as cancer, neurodegeneration, metabolic syndromes, and aging. The emergence of the field of innate immunity has led to an expanded view of the immune system, which is no longer restricted to vertebrates and instead concerns all metazoans, plants, and even prokaryotes. The study of innate immunity has given rise to new concepts and language. Here, we review the history and definition of the core concepts of innate immunity, discussing their value and fruitfulness in the long run.


Asunto(s)
Inmunidad Innata , Memoria Inmunológica , Animales , Invertebrados , Inmunidad Adaptativa , Vertebrados
2.
Nat Immunol ; 20(4): 433-446, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30804553

RESUMEN

Cells use mitophagy to remove damaged or unwanted mitochondria to maintain homeostasis. Here we report that the intracellular bacterial pathogen Listeria monocytogenes exploits host mitophagy to evade killing. We found that L. monocytogenes induced mitophagy in macrophages through the virulence factor listeriolysin O (LLO). We discovered that NLRX1, the only Nod-like receptor (NLR) family member with a mitochondrial targeting sequence, contains an LC3-interacting region (LIR) and directly associated with LC3 through the LIR. NLRX1 and its LIR motif were essential for L. monocytogenes-induced mitophagy. NLRX1 deficiency and use of a mitophagy inhibitor both increased mitochondrial production of reactive oxygen species and thereby suppressed the survival of L. monocytogenes. Mechanistically, L. monocytogenes and LLO induced oligomerization of NLRX1 to promote binding of its LIR motif to LC3 for induction of mitophagy. Our study identifies NLRX1 as a novel mitophagy receptor and discovers a previously unappreciated strategy used by pathogens to hijack a host cell homeostasis system for their survival.


Asunto(s)
Listeria monocytogenes/fisiología , Proteínas Mitocondriales/fisiología , Mitofagia , Animales , Autofagia , Toxinas Bacterianas/metabolismo , Línea Celular , Femenino , Proteínas de Choque Térmico/metabolismo , Proteínas Hemolisinas/metabolismo , Humanos , Listeria monocytogenes/patogenicidad , Listeriosis/metabolismo , Listeriosis/microbiología , Macrófagos/microbiología , Macrófagos/ultraestructura , Masculino , Ratones , Ratones Noqueados , Viabilidad Microbiana , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Mitocondriales/química , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Dominios Proteicos , Especies Reactivas de Oxígeno/metabolismo , Factores de Virulencia/metabolismo
3.
Mol Cell ; 82(15): 2815-2831.e5, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35752171

RESUMEN

Protein import into mitochondria is a highly regulated process, yet how cells clear mitochondria undergoing dysfunctional protein import remains poorly characterized. Here we showed that mitochondrial protein import stress (MPIS) triggers localized LC3 lipidation. This arm of the mitophagy pathway occurs through the Nod-like receptor (NLR) protein NLRX1 while, surprisingly, without the engagement of the canonical mitophagy protein PINK1. Mitochondrial depolarization, which itself induces MPIS, also required NLRX1 for LC3 lipidation. While normally targeted to the mitochondrial matrix, cytosol-retained NLRX1 recruited RRBP1, a ribosome-binding transmembrane protein of the endoplasmic reticulum, which relocated to the mitochondrial vicinity during MPIS, and the NLRX1/RRBP1 complex in turn controlled the recruitment and lipidation of LC3. Furthermore, NLRX1 controlled skeletal muscle mitophagy in vivo and regulated endurance capacity during exercise. Thus, localization and lipidation of LC3 at the site of mitophagosome formation is a regulated step of mitophagy controlled by NLRX1/RRBP1 in response to MPIS.


Asunto(s)
Proteínas Mitocondriales , Mitofagia , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Transporte de Proteínas
4.
Cell ; 158(2): 288-299, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-25036629

RESUMEN

The etiology of colorectal cancer (CRC) has been linked to deficiencies in mismatch repair and adenomatous polyposis coli (APC) proteins, diet, inflammatory processes, and gut microbiota. However, the mechanism through which the microbiota synergizes with these etiologic factors to promote CRC is not clear. We report that altering the microbiota composition reduces CRC in APC(Min/+)MSH2(-/-) mice, and that a diet reduced in carbohydrates phenocopies this effect. Gut microbes did not induce CRC in these mice through an inflammatory response or the production of DNA mutagens but rather by providing carbohydrate-derived metabolites such as butyrate that fuel hyperproliferation of MSH2(-/-) colon epithelial cells. Further, we provide evidence that the mismatch repair pathway has a role in regulating ß-catenin activity and modulating the differentiation of transit-amplifying cells in the colon. These data thereby provide an explanation for the interaction between microbiota, diet, and mismatch repair deficiency in CRC induction. PAPERCLIP:


Asunto(s)
Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Carbohidratos de la Dieta/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Butiratos/metabolismo , Proliferación Celular , Transformación Celular Neoplásica , Pólipos del Colon/metabolismo , Pólipos del Colon/microbiología , Pólipos del Colon/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/microbiología , Reparación de la Incompatibilidad de ADN , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Inflamación/genética , Inflamación/metabolismo , Inflamación/microbiología , Ratones , Ratones Endogámicos C57BL , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS/genética , Proteínas Nucleares/metabolismo , Organismos Libres de Patógenos Específicos , beta Catenina/metabolismo
5.
Proc Natl Acad Sci U S A ; 120(52): e2306863120, 2023 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-38127978

RESUMEN

The gut microbiota is a considerable source of biologically active compounds that can promote intestinal homeostasis and improve immune responses. Here, we used large expression libraries of cloned metagenomic DNA to identify compounds able to sustain an anti-inflammatory reaction on host cells. Starting with a screen for NF-κB activation, we have identified overlapping clones harbouring a heterodimeric ATP-binding cassette (ABC)-transporter from a Firmicutes. Extensive purification of the clone's supernatant demonstrates that the ABC-transporter allows for the efficient extracellular accumulation of three muropeptide precursor, with anti-inflammatory properties. They induce IL-10 secretion from human monocyte-derived dendritic cells and proved effective in reducing AIEC LF82 epithelial damage and IL-8 secretion in human intestinal resections. In addition, treatment with supernatants containing the muropeptide precursor reduces body weight loss and improves histological parameters in Dextran Sulfate Sodium (DSS)-treated mice. Until now, the source of peptidoglycan fragments was shown to come from the natural turnover of the peptidoglycan layer by endogenous peptidoglycan hydrolases. This is a report showing an ABC-transporter as a natural source of secreted muropeptide precursor and as an indirect player in epithelial barrier strengthening. The mechanism described here might represent an important component of the host immune homeostasis.


Asunto(s)
Colitis , Microbioma Gastrointestinal , Humanos , Ratones , Animales , Peptidoglicano/metabolismo , Intestinos/patología , Inflamación/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Antiinflamatorios/metabolismo , Sulfato de Dextran , Colitis/metabolismo , Modelos Animales de Enfermedad , Colon/metabolismo , Ratones Endogámicos C57BL
6.
J Immunol ; 208(7): 1782-1789, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35256512

RESUMEN

Commensal intestinal protozoa, unlike their pathogenic relatives, are neglected members of the mammalian microbiome. These microbes have a significant impact on the host's intestinal immune homeostasis, typically by elevating anti-microbial host defense. Tritrichomonas musculis, a protozoan gut commensal, strengthens the intestinal host defense against enteric Salmonella infections through Asc- and Il1r1-dependent Th1 and Th17 cell activation. However, the underlying inflammasomes mediating this effect remain unknown. In this study, we report that colonization with T. musculis results in an increase in luminal extracellular ATP that is followed by increased caspase activity, higher cell death, elevated levels of IL-1ß, and increased numbers of IL-18 receptor-expressing Th1 and Th17 cells in the colon. Mice deficient in either Nlrp1b or Nlrp3 failed to display these protozoan-driven immune changes and lost resistance to enteric Salmonella infections even in the presence of T. musculis These findings demonstrate that T. musculis-mediated host protection requires sensors of extracellular and intracellular ATP to confer resistance to enteric Salmonella infections.


Asunto(s)
Proteínas Reguladoras de la Apoptosis , Microbiota , Proteína con Dominio Pirina 3 de la Familia NLR , Tritrichomonas , Animales , Proteínas Reguladoras de la Apoptosis/inmunología , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Mamíferos/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Simbiosis , Tritrichomonas/metabolismo
7.
Proc Natl Acad Sci U S A ; 117(20): 10946-10957, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32350141

RESUMEN

Four decades ago, it was identified that muramyl dipeptide (MDP), a peptidoglycan-derived bacterial cell wall component, could display immunosuppressive functions in animals through mechanisms that remain unexplored. We sought to revisit these pioneering observations because mutations in NOD2, the gene encoding the host sensor of MDP, are associated with increased risk of developing the inflammatory bowel disease Crohn's disease, thus suggesting that the loss of the immunomodulatory functions of NOD2 could contribute to the development of inflammatory disease. Here, we demonstrate that intraperitoneal (i.p.) administration of MDP triggered regulatory T cells and the accumulation of a population of tolerogenic CD103+ dendritic cells (DCs) in the spleen. This was found to occur not through direct sensing of MDP by DCs themselves, but rather via the production of the cytokine GM-CSF, another factor with an established regulatory role in Crohn's disease pathogenesis. Moreover, we demonstrate that populations of CD103-expressing DCs in the gut lamina propria are enhanced by the activation of NOD2, indicating that MDP sensing plays a critical role in shaping the immune response to intestinal antigens by promoting a tolerogenic environment via manipulation of DC populations.


Asunto(s)
Antígenos CD/metabolismo , Células Dendríticas/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Tolerancia Inmunológica , Cadenas alfa de Integrinas/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Acetilmuramil-Alanil-Isoglutamina/genética , Acetilmuramil-Alanil-Isoglutamina/metabolismo , Animales , Enfermedad de Crohn , Citocinas , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Proteína Adaptadora de Señalización NOD2/genética , Linfocitos T Reguladores/metabolismo
8.
J Biol Chem ; 296: 100050, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33168630

RESUMEN

Large cytosolic protein aggregates are removed by two main cellular processes, autophagy and the ubiquitin-proteasome system, and defective clearance of these protein aggregates results in proteotoxicity and cell death. Recently, we found that the eIF2α kinase heme-regulated inhibitory (HRI) induced a cytosolic unfolded protein response to prevent aggregation of innate immune signalosomes, but whether HRI acts as a general sensor of proteotoxicity in the cytosol remains unclear. Here we show that HRI controls autophagy to clear cytosolic protein aggregates when the ubiquitin-proteasome system is inhibited. We further report that silencing the expression of HRI resulted in decreased levels of BAG3 and HSPB8, two proteins involved in chaperone-assisted selective autophagy, suggesting that HRI may control proteostasis in the cytosol at least in part through chaperone-assisted selective autophagy. Moreover, knocking down the expression of HRI resulted in cytotoxic accumulation of overexpressed α-synuclein, a protein known to aggregate in Parkinson's disease, dementia with Lewy bodies, and multiple system atrophy. In agreement with these data, protein aggregate accumulation and microglia activation were observed in the spinal cord white matter of 7-month-old Hri-/- mice as compared with Hri+/+ littermates. Moreover, aged Hri-/- mice showed accumulation of misfolded α-synuclein in the lateral collateral pathway, a region of the sacral spinal cord horn that receives visceral sensory afferents from the bladder and distal colon, a pathological feature common to α-synucleinopathies in humans. Together, these results suggest that HRI contributes to a general cytosolic unfolded protein response that could be leveraged to bolster the clearance of cytotoxic protein aggregates.


Asunto(s)
Autofagia , Microglía/metabolismo , Agregado de Proteínas , Proteínas Serina-Treonina Quinasas/metabolismo , Médula Espinal/metabolismo , Respuesta de Proteína Desplegada , eIF-2 Quinasa/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Células HEK293 , Células HeLa , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/metabolismo , Humanos , Ratones , Ratones Noqueados , Microglía/patología , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Médula Espinal/patología , eIF-2 Quinasa/genética
9.
Nat Immunol ; 11(1): 55-62, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19898471

RESUMEN

Autophagy is emerging as a crucial defense mechanism against bacteria, but the host intracellular sensors responsible for inducing autophagy in response to bacterial infection remain unknown. Here we demonstrated that the intracellular sensors Nod1 and Nod2 are critical for the autophagic response to invasive bacteria. By a mechanism independent of the adaptor RIP2 and transcription factor NF-kappaB, Nod1 and Nod2 recruited the autophagy protein ATG16L1 to the plasma membrane at the bacterial entry site. In cells homozygous for the Crohn's disease-associated NOD2 frameshift mutation, mutant Nod2 failed to recruit ATG16L1 to the plasma membrane and wrapping of invading bacteria by autophagosomes was impaired. Our results link bacterial sensing by Nod proteins to the induction of autophagy and provide a functional link between Nod2 and ATG16L1, which are encoded by two of the most important genes associated with Crohn's disease.


Asunto(s)
Autofagia , Proteínas Portadoras/metabolismo , Membrana Celular/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Animales , Proteínas Relacionadas con la Autofagia , Bacterias/metabolismo , Proteínas Portadoras/genética , Línea Celular , Membrana Celular/microbiología , Membrana Celular/ultraestructura , Células Cultivadas , Femenino , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HeLa , Humanos , Immunoblotting , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica , Microscopía Fluorescente , Mutación , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD2/genética , Transfección
10.
Brain Behav Immun ; 101: 335-345, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35093492

RESUMEN

Functional dyspepsia (FD) affects up to 15% of the population and is characterised by recurring upper gastrointestinal (GI) symptoms occurring in the absence of clinically identifiable pathology. Psychological stress is a key factor associated with the onset of FD and locally acting hypothalamic-pituitary-adrenal (HPA) axis hormones have been implicated in GI motility and barrier dysfunction. Recent pre-clinical work has identified mechanistic pathways linking corticotropin-releasing hormone (CRH) with the innate epithelial immune protein NLRP6, an inflammasome that has been shown to regulate GI mucus secretion. We recruited twelve FD patients and twelve healthy individuals to examine whether dysregulation of hypothalamic-pituitary adrenal (HPA) axis hormones and altered NLRP6 pathways were evident in the duodenal mucosa. Protein expression was assessed by immunoblot and immunohistochemistry in D2 duodenal biopsies. Plasma HPA axis hormones were assayed by ELISA and enteroid and colorectal cancer cell line cultures were used to verify function. FD patients exhibited reduced duodenal CRH-receptor 2, compared to non-GI disease controls, indicating a dysregulation of duodenal HPA signalling. The loss of CRH-receptor 2 correlated with reduced NLRP6 expression and autophagy function, processes critical for maintaining goblet cell homeostasis. In accordance, duodenal goblet cell numbers and mucin exocytosis was reduced in FD patients compared to controls. In vitro studies demonstrated that CRH could reduce NLRP6 in duodenal spheroids and promote mucus secretion in the HT29-MTX-E12 cell line. In conclusion, FD patients exhibit defects in the NLRP6-autophagy axis with decreased goblet cell function that may drive symptoms of disease. These features correlated with loss of CRH receptor 2 and may be driven by dysregulation of HPA signalling in the duodenum of FD patients.


Asunto(s)
Dispepsia , Péptidos y Proteínas de Señalización Intracelular , Sistema Hipófiso-Suprarrenal , Receptores de Hormona Liberadora de Corticotropina , Autofagia , Duodeno/metabolismo , Dispepsia/metabolismo , Células Caliciformes/metabolismo , Homeostasis , Hormonas/metabolismo , Humanos , Sistema Hipotálamo-Hipofisario/metabolismo , Péptidos y Proteínas de Señalización Intracelular/genética , Sistema Hipófiso-Suprarrenal/metabolismo , Receptores de Hormona Liberadora de Corticotropina/genética , Receptores de Hormona Liberadora de Corticotropina/metabolismo
11.
Immunity ; 39(5): 858-73, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24238340

RESUMEN

The peptidoglycan sensor Nod2 and the autophagy protein ATG16L1 have been linked to Crohn's disease (CD). Although Nod2 and the related sensor, Nod1, direct ATG16L1 to initiate anti-bacterial autophagy, whether ATG16L1 affects Nod-driven inflammation has not been examined. Here, we uncover an unanticipated autophagy-independent role for ATG16L1 in negatively regulating Nod-driven inflammatory responses. Knockdown of ATG16L1 expression, but not that of ATG5 or ATG9a, specifically enhanced Nod-driven cytokine production. In addition, autophagy-incompetent truncated forms of ATG16L1 regulated Nod-driven cytokine responses. Mechanistically, we demonstrated that ATG16L1 interfered with poly-ubiquitination of the Rip2 adaptor and recruitment of Rip2 into large signaling complexes. The CD-associated allele of ATG16L1 was impaired in its ability to regulate Nod-driven inflammatory responses. Overall, these results suggest that ATG16L1 is critical for Nod-dependent regulation of cytokine responses and that disruption of this Nod1- or Nod2-ATG16L1 signaling axis could contribute to the chronic inflammation associated with CD.


Asunto(s)
Autofagia/fisiología , Proteínas Portadoras/fisiología , Citocinas/biosíntesis , Proteína Adaptadora de Señalización NOD1/fisiología , Proteína Adaptadora de Señalización NOD2/fisiología , Animales , Proteína 5 Relacionada con la Autofagia , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/química , Proteínas Portadoras/genética , Línea Celular , Enfermedad de Crohn/genética , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/patología , Citocinas/genética , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Predisposición Genética a la Enfermedad , Humanos , Inflamación , Mucosa Intestinal/citología , Ratones , Proteínas Asociadas a Microtúbulos/deficiencia , Proteínas Asociadas a Microtúbulos/fisiología , Procesamiento Proteico-Postraduccional , Interferencia de ARN , ARN Interferente Pequeño/farmacología , Proteína Serina-Treonina Quinasa 2 de Interacción con Receptor , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Ubiquitinación
12.
Mol Cell ; 54(2): 309-20, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24766895

RESUMEN

In recent years, our understanding of the mechanisms underlying colorectal carcinogenesis has vastly expanded. Underlying inflammation within the intestine, diet, and most recently, the gut microbiota, have been demonstrated to influence the development of colorectal cancer. However, since cancer is ultimately a genetic disease, these factors are thought to create genotoxic stress within the intestinal environment to promote genetic and epigenetic alterations leading to cancer. In this review, we will focus on how gut microbes intersect with inflammation, diet, and host genetics to influence the development of colon cancer.


Asunto(s)
Neoplasias del Colon/microbiología , Intestinos/microbiología , Microbiota/inmunología , Carcinogénesis , Colitis/microbiología , Colitis/patología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Daño del ADN , Progresión de la Enfermedad , Humanos , Inmunidad Innata , Inflamación
13.
Mol Cell ; 55(2): 238-52, 2014 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-24954904

RESUMEN

Mammalian cell homeostasis during starvation depends on initiation of autophagy by endoplasmic reticulum-localized phosphatidylinositol 3-phosphate (PtdIns(3)P) synthesis. Formation of double-membrane autophagosomes that engulf cytosolic components requires the LC3-conjugating Atg12-5-16L1 complex. The molecular mechanisms of Atg12-5-16L1 recruitment and significance of PtdIns(3)P synthesis at autophagosome formation sites are unknown. By identifying interacting partners of WIPIs, WD-repeat PtdIns(3)P effector proteins, we found that Atg16L1 directly binds WIPI2b. Mutation experiments and ectopic localization of WIPI2b to plasma membrane show that WIPI2b is a PtdIns(3)P effector upstream of Atg16L1 and is required for LC3 conjugation and starvation-induced autophagy through recruitment of the Atg12-5-16L1 complex. Atg16L1 mutants, which do not bind WIPI2b but bind FIP200, cannot rescue starvation-induced autophagy in Atg16L1-deficient MEFs. WIPI2b is also required for autophagic clearance of pathogenic bacteria. WIPI2b binds the membrane surrounding Salmonella and recruits the Atg12-5-16L1 complex, initiating LC3 conjugation, autophagosomal membrane formation, and engulfment of Salmonella.


Asunto(s)
Proteínas Portadoras/fisiología , Proteínas de la Membrana/fisiología , Proteínas Asociadas a Microtúbulos/metabolismo , Fagosomas/metabolismo , Fosfatos de Fosfatidilinositol/metabolismo , Salmonella typhimurium/fisiología , Secuencia de Aminoácidos , Animales , Autofagia , Proteína 12 Relacionada con la Autofagia , Proteína 5 Relacionada con la Autofagia , Proteínas Relacionadas con la Autofagia , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Secuencia Conservada , Células HEK293 , Interacciones Huésped-Patógeno , Humanos , Membranas Intracelulares/metabolismo , Ratones , Datos de Secuencia Molecular , Fagocitosis , Fagosomas/microbiología , Proteínas de Unión a Fosfato , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Isoformas de Proteínas/fisiología , Procesamiento Proteico-Postraduccional , Transporte de Proteínas , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo
14.
Trends Immunol ; 39(12): 992-1004, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30377046

RESUMEN

The gut microbiota is important in health and disease. Whereas the intestinal immune system has evolved to protect the mucosal barrier against pathogens, there is much interest in understanding how it influences the composition and functions of resident microbial communities. Overall, host innate immunity exerts little influence on the microbiota at homeostasis, but increases upon immune activation and the onset of inflammation, as well as in the presence of certain members of the microbiota. However, many experiments have not adequately incorporated study design to detect such immune influences, including using proper control groups, precise sampling and timing, and measures beyond broad-scale descriptions of dysbiosis for microbial analysis. We discuss these and other challenges in the context of current understanding of chronic inflammatory disease.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Inmunidad Innata/inmunología , Animales , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Ratones , Modelos Animales
15.
J Biol Chem ; 294(22): 9007-9015, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-30996003

RESUMEN

NOD1 and NOD2 are intracellular sensors of bacterial peptidoglycan that belong to the Nod-like receptor family of innate immune proteins. In addition to their role as direct bacterial sensors, it was proposed that the nucleotide-binding oligomerization domain (NOD) proteins could detect endoplasmic reticulum (ER) stress induced by thapsigargin, an inhibitor of the sarcoplasmic or endoplasmic reticulum calcium ATPase family that pumps Ca2+ into the ER, resulting in pro-inflammatory signaling. Here, we confirm that thapsigargin induces NOD-dependent pro-inflammatory signaling in epithelial cells. However, the effect was specific to thapsigargin, as tunicamycin and the subtilase cytotoxin SubAB from Shiga toxigenic Escherichia coli, which induce ER stress by other mechanisms, did not induce cytokine expression. The calcium ionophore A23187 also induced NOD-dependent signaling, and calcium chelators demonstrated a role for both intracellular and extracellular calcium in mediating thapsigargin-induced and NOD-dependent pro-inflammatory signaling, in part through the activation of plasma membrane-associated calcium release-activated channels. Moreover, our results demonstrate that both endocytosis and the addition of serum to the cell culture medium were required for thapsigargin-mediated NOD activation. Finally, we analyzed cell culture grade fetal calf serum as well as serum from laboratory mice using HPLC and MS identified the presence of various peptidoglycan fragments. We propose that cellular perturbations that affect intracellular Ca2+ can trigger internalization of peptidoglycan trace contaminants found in culture serum, thereby stimulating pro-inflammatory signaling. The presence of peptidoglycan in animal serum suggests that a homeostatic function of NOD signaling may have been previously overlooked.


Asunto(s)
Citocinas/metabolismo , Estrés del Retículo Endoplásmico , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Peptidoglicano/sangre , Calcimicina/química , Calcimicina/farmacología , Calcio/química , Calcio/metabolismo , Quimiocina CXCL1/metabolismo , Estrés del Retículo Endoplásmico/efectos de los fármacos , Técnicas de Inactivación de Genes , Células HCT116 , Humanos , Interleucina-8/metabolismo , Proteína Adaptadora de Señalización NOD1/deficiencia , Proteína Adaptadora de Señalización NOD1/genética , Proteína Adaptadora de Señalización NOD2/deficiencia , Proteína Adaptadora de Señalización NOD2/genética , Transducción de Señal/efectos de los fármacos , Tapsigargina/farmacología
16.
Mol Microbiol ; 112(1): 280-301, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31070821

RESUMEN

Campylobacter jejuni is a prevalent enteric pathogen that changes morphology from helical to coccoid under unfavorable conditions. Bacterial peptidoglycan maintains cell shape. As C. jejuni transformed from helical to coccoid, peptidoglycan dipeptides increased and tri- and tetrapeptides decreased. The DL-carboxypeptidase Pgp1 important for C. jejuni helical morphology and putative N-acetylmuramoyl-L-alanyl amidase AmiA were both involved in the coccoid transition. Mutants in pgp1 and amiA showed reduced coccoid formation, with ∆pgp1∆amiA producing minimal coccoids. Both ∆amiA and ∆amiA∆pgp1 lacked flagella and formed unseparated chains of cells consistent with a role for AmiA in cell separation. All strains accumulated peptidoglycan dipeptides over time, but only strains capable of becoming coccoid displayed tripeptide changes. C. jejuni helical shape and corresponding peptidoglycan structure are important for pathogenesis-related attributes. Concomitantly, changing to a coccoid morphology resulted in differences in pathogenic properties; coccoid C. jejuni were non-motile and non-infectious, with minimal adherence and invasion of epithelial cells and an inability to stimulate IL-8. Coccoid peptidoglycan exhibited reduced activation of innate immune receptors Nod1 and Nod2 versus helical peptidoglycan. C. jejuni also transitioned to coccoid within epithelial cells, so the inability of the immune system to detect coccoid C. jejuni may be significant in its pathogenesis.


Asunto(s)
Campylobacter jejuni/metabolismo , Forma de la Célula/fisiología , Peptidoglicano/metabolismo , Proteínas Bacterianas/metabolismo , Infecciones por Campylobacter/microbiología , Campylobacter jejuni/patogenicidad , Campylobacter jejuni/fisiología , Carboxipeptidasas/metabolismo , Pared Celular/metabolismo , Peptidoglicano/química , Peptidoglicano/inmunología
17.
Cell Microbiol ; 21(11): e13079, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31265745

RESUMEN

Inflammasomes are cytosolic, multimeric protein complexes capable of activating pro-inflammatory cytokines such as IL-1ß and IL-18, which play a key role in host defence. Inflammasome components are highly expressed in the intestinal epithelium. In recent years, studies have begun to demonstrate that epithelial-intrinsic inflammasomes play a critical role in regulating epithelial homeostasis, both by defending the epithelium from pathogenic insult and through the regulation of the mucosal environment. However, the majority of research regarding inflammasome activation has focused on professional immune cells, such as macrophages. Here, we present an overview of the current understanding of inflammasome function in epithelial cells and at mucosal surfaces and, in particular, in the intestine.


Asunto(s)
Células Epiteliales/metabolismo , Inflamasomas/metabolismo , Mucosa Intestinal/metabolismo , Animales , Linfocitos T CD4-Positivos/inmunología , Carcinogénesis/genética , Carcinogénesis/inmunología , Carcinogénesis/metabolismo , Regulación de la Expresión Génica/inmunología , Interacciones Microbiota-Huesped , Humanos , Inflamasomas/genética , Inflamasomas/inmunología , Interleucina-18/inmunología , Interleucina-18/metabolismo , Interleucina-1beta/metabolismo , Interleucinas/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/microbiología , Intestinos/inmunología , Intestinos/microbiología , Microbiota/inmunología , Interleucina-22
18.
J Biol Chem ; 293(9): 3073-3087, 2018 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-29317503

RESUMEN

The gut microbiome contributes to inflammatory bowel disease (IBD), in which bacteria can be present within the epithelium. Epithelial barrier function is decreased in IBD, and dysfunctional epithelial mitochondria and endoplasmic reticulum (ER) stress have been individually associated with IBD. We therefore hypothesized that the combination of ER and mitochondrial stresses significantly disrupt epithelial barrier function. Here, we treated human colonic biopsies, epithelial colonoids, and epithelial cells with an uncoupler of oxidative phosphorylation, dinitrophenol (DNP), with or without the ER stressor tunicamycin and assessed epithelial barrier function by monitoring internalization and translocation of commensal bacteria. We also examined barrier function and colitis in mice exposed to dextran sodium sulfate (DSS) or DNP and co-treated with DAPK6, an inhibitor of death-associated protein kinase 1 (DAPK1). Contrary to our hypothesis, induction of ER stress (i.e. the unfolded protein response) protected against decreased barrier function caused by the disruption of mitochondrial function. ER stress did not prevent DNP-driven uptake of bacteria; rather, specific mobilization of the ATF6 arm of ER stress and recruitment of DAPK1 resulted in enhanced autophagic killing (xenophagy) of bacteria. Of note, epithelia with a Crohn's disease-susceptibility mutation in the autophagy gene ATG16L1 exhibited less xenophagy. Systemic delivery of the DAPK1 inhibitor DAPK6 increased bacterial translocation in DSS- or DNP-treated mice. We conclude that promoting ER stress-ATF6-DAPK1 signaling in transporting enterocytes counters the transcellular passage of bacteria evoked by dysfunctional mitochondria, thereby reducing the potential for metabolic stress to reactivate or perpetuate inflammation.


Asunto(s)
Proteínas Quinasas Asociadas a Muerte Celular/metabolismo , Estrés del Retículo Endoplásmico , Mitocondrias/metabolismo , Factor de Transcripción Activador 6/metabolismo , Anciano , Animales , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/efectos de los fármacos , Epitelio/efectos de los fármacos , Epitelio/metabolismo , Escherichia coli/efectos de los fármacos , Escherichia coli/fisiología , Femenino , Humanos , Masculino , Ratones , Mitocondrias/efectos de los fármacos , Fosforilación Oxidativa/efectos de los fármacos , Permeabilidad , Tunicamicina/farmacología
19.
Immunol Cell Biol ; 97(6): 552-562, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30768806

RESUMEN

The NOD-like receptor (NLR) family plays an important role in innate immunity. Class II transactivator and NOD-like receptor caspase activation and recruitment domain CARD containing 5 (NLRC5) are unusual members of the NLR family that instead of recognizing pathogen-associated or damage-associated molecular patterns, form enhanceosomes with adaptor molecules and modulate major histocompatibility complex (MHC) class II and MHC class I expression, respectively. While NLRC5 has been shown to play a role during intracellular pathogen infection and tumor cell immune evasion, its role in regulating antigen-specific CD8+ T-cell responses at the intestinal mucosa has not been investigated. Here, we take advantage of the rotavirus model in adult mice to dissect the impact of NLRC5 on CD8+ T-cell responses to this viral infection at the gut mucosa. We show that while Nlrc5-/- mice exhibited normal proportions of T-cell subpopulations in the intraepithelial and lamina propria compartments, these mice had decreased baseline MHC class I expression on various immune cells in the lamina propria. Upon rotavirus infection, Nlrc5 deficiency resulted in impaired H2-Kb -restricted antigen-specific CD8+ T-cell responses, which were recapitulated in mice deficient for Nlrc5 within the dendritic cell compartment. The impaired CD8+ T-cell response in Nlrc5-/- mice was not significant enough to impact viral titers, suggesting compensation in Nlrc5-/- mice, perhaps as a result of higher numbers of activated B cells in the mesenteric lymph nodes and normal rotavirus-specific immunoglobulin A responses. Collectively, our results demonstrate a minor role for NLRC5 in modulating H2-Kb -restricted antigen-specific CD8+ T-cell responses in the small intestine during rotavirus infection in adult mice.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Mucosa Intestinal/inmunología , Infecciones por Rotavirus/inmunología , Rotavirus/fisiología , Animales , Presentación de Antígeno , Antígenos Virales/inmunología , Células Cultivadas , Antígenos H-2/metabolismo , Epítopos Inmunodominantes/inmunología , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Carga Viral
20.
Arch Biochem Biophys ; 670: 69-81, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-30578751

RESUMEN

NOD1 and NOD2 are related intracellular sensors of bacterial peptidoglycan and belong to the Nod-like receptor (NLR) family of innate immune proteins that play fundamental and pleiotropic roles in host defense against infection and in the control of inflammation. The importance of these proteins is also highlighted by the genetic association between single nucleotide polymorphisms in NOD2 and susceptibility to Crohn's disease, an inflammatory bowel disease. At the cellular level, recent efforts have delineated the signaling pathways triggered following activation of NOD1 and NOD2, and the interplay with various cellular processes, such as autophagy. In vivo studies have revealed the importance of NOD-dependent host defense in models of infection, and a crucial area of investigation focuses on understanding the role of NOD1 and NOD2 at the intestinal mucosa, as this is of prime importance for understanding the etiology of Crohn's disease.


Asunto(s)
Enfermedad , Inmunidad , Inflamación/inmunología , Inflamación/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Proteína Adaptadora de Señalización NOD2/metabolismo , Animales , Humanos , Inflamación/patología , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA