Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 55
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 180(1): 50-63.e12, 2020 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-31923399

RESUMEN

Mucosal barrier immunity is essential for the maintenance of the commensal microflora and combating invasive bacterial infection. Although immune and epithelial cells are thought to be the canonical orchestrators of this complex equilibrium, here, we show that the enteric nervous system (ENS) plays an essential and non-redundant role in governing the antimicrobial protein (AMP) response. Using confocal microscopy and single-molecule fluorescence in situ mRNA hybridization (smFISH) studies, we observed that intestinal neurons produce the pleiotropic cytokine IL-18. Strikingly, deletion of IL-18 from the enteric neurons alone, but not immune or epithelial cells, rendered mice susceptible to invasive Salmonella typhimurium (S.t.) infection. Mechanistically, unbiased RNA sequencing and single-cell sequencing revealed that enteric neuronal IL-18 is specifically required for homeostatic goblet cell AMP production. Together, we show that neuron-derived IL-18 signaling controls tissue-wide intestinal immunity and has profound consequences on the mucosal barrier and invasive bacterial killing.


Asunto(s)
Inmunidad Mucosa/inmunología , Interleucina-18/inmunología , Mucosa Intestinal/inmunología , Animales , Citocinas/inmunología , Sistema Nervioso Entérico/inmunología , Sistema Nervioso Entérico/metabolismo , Células Epiteliales/inmunología , Femenino , Células Caliciformes/inmunología , Interleucina-18/biosíntesis , Mucosa Intestinal/metabolismo , Intestino Delgado/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/inmunología , Ratas , Ratas Sprague-Dawley , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Transducción de Señal/inmunología
2.
Cell ; 163(6): 1444-56, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26638073

RESUMEN

The intestinal mucosal barrier controlling the resident microbiome is dependent on a protective mucus layer generated by goblet cells, impairment of which is a hallmark of the inflammatory bowel disease, ulcerative colitis. Here, we show that IL-18 is critical in driving the pathologic breakdown of barrier integrity in a model of colitis. Deletion of Il18 or its receptor Il18r1 in intestinal epithelial cells (Δ/EC) conferred protection from colitis and mucosal damage in mice. In contrast, deletion of the IL-18 negative regulator Il18bp resulted in severe colitis associated with loss of mature goblet cells. Colitis and goblet cell loss were rescued in Il18bp(-/-);Il18r(Δ/EC) mice, demonstrating that colitis severity is controlled at the level of IL-18 signaling in intestinal epithelial cells. IL-18 inhibited goblet cell maturation by regulating the transcriptional program instructing goblet cell development. These results inform on the mechanism of goblet cell dysfunction that underlies the pathology of ulcerative colitis.


Asunto(s)
Colitis Ulcerosa/patología , Colitis Ulcerosa/fisiopatología , Interleucina-18/inmunología , Animales , Colitis Ulcerosa/inducido químicamente , Colitis Ulcerosa/metabolismo , Sulfato de Dextran , Células Endoteliales/metabolismo , Células Epiteliales/citología , Femenino , Células Caliciformes/metabolismo , Células Caliciformes/patología , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Subunidad alfa del Receptor de Interleucina-18/genética , Subunidad alfa del Receptor de Interleucina-18/metabolismo , Mucosa Intestinal/fisiopatología , Masculino , Ratones , Transducción de Señal
4.
Cell ; 159(7): 1563-77, 2014 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-25525875

RESUMEN

The mechanism by which cells undergo death determines whether dying cells trigger inflammatory responses or remain immunologically silent. Mitochondria play a central role in the induction of cell death, as well as in immune signaling pathways. Here, we identify a mechanism by which mitochondria and downstream proapoptotic caspases regulate the activation of antiviral immunity. In the absence of active caspases, mitochondrial outer membrane permeabilization by Bax and Bak results in the expression of type I interferons (IFNs). This induction is mediated by mitochondrial DNA-dependent activation of the cGAS/STING pathway and results in the establishment of a potent state of viral resistance. Our results show that mitochondria have the capacity to simultaneously expose a cell-intrinsic inducer of the IFN response and to inactivate this response in a caspase-dependent manner. This mechanism provides a dual control, which determines whether mitochondria initiate an immunologically silent or a proinflammatory type of cell death.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Interferón Tipo I/metabolismo , Transducción de Señal , Animales , ADN Mitocondrial/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Interferón Tipo I/inmunología , Ratones , Ratones Noqueados , Virosis/inmunología
5.
Cell ; 158(5): 1000-1010, 2014 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-25171403

RESUMEN

Specific members of the intestinal microbiota dramatically affect inflammatory bowel disease (IBD) in mice. In humans, however, identifying bacteria that preferentially affect disease susceptibility and severity remains a major challenge. Here, we used flow-cytometry-based bacterial cell sorting and 16S sequencing to characterize taxa-specific coating of the intestinal microbiota with immunoglobulin A (IgA-SEQ) and show that high IgA coating uniquely identifies colitogenic intestinal bacteria in a mouse model of microbiota-driven colitis. We then used IgA-SEQ and extensive anaerobic culturing of fecal bacteria from IBD patients to create personalized disease-associated gut microbiota culture collections with predefined levels of IgA coating. Using these collections, we found that intestinal bacteria selected on the basis of high coating with IgA conferred dramatic susceptibility to colitis in germ-free mice. Thus, our studies suggest that IgA coating identifies inflammatory commensals that preferentially drive intestinal disease. Targeted elimination of such bacteria may reduce, reverse, or even prevent disease development.


Asunto(s)
Colitis Ulcerosa/inmunología , Enfermedad de Crohn/inmunología , Inmunoglobulina A/inmunología , Microbiota , Animales , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/patología , Enfermedad de Crohn/microbiología , Enfermedad de Crohn/patología , ADN Bacteriano/análisis , Disbiosis/inmunología , Disbiosis/microbiología , Humanos , Inflamasomas/inmunología , Inflamación/inmunología , Inflamación/microbiología , Intestinos/inmunología , Intestinos/microbiología , Ratones , Ratones Endogámicos C57BL , ARN Ribosómico 16S/análisis , Organismos Libres de Patógenos Específicos
6.
Nature ; 573(7772): 69-74, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31435009

RESUMEN

Direct recognition of invading pathogens by innate immune cells is a critical driver of the inflammatory response. However, cells of the innate immune system can also sense their local microenvironment and respond to physiological fluctuations in temperature, pH, oxygen and nutrient availability, which are altered during inflammation. Although cells of the immune system experience force and pressure throughout their life cycle, little is known about how these mechanical processes regulate the immune response. Here we show that cyclical hydrostatic pressure, similar to that experienced by immune cells in the lung, initiates an inflammatory response via the mechanically activated ion channel PIEZO1. Mice lacking PIEZO1 in innate immune cells showed ablated pulmonary inflammation in the context of bacterial infection or fibrotic autoinflammation. Our results reveal an environmental sensory axis that stimulates innate immune cells to mount an inflammatory response, and demonstrate a physiological role for PIEZO1 and mechanosensation in immunity.


Asunto(s)
Presión Hidrostática , Inmunidad Innata , Canales Iónicos/metabolismo , Mecanotransducción Celular/inmunología , Animales , Endotelina-1/metabolismo , Femenino , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Transducción de Señal
8.
PLoS Pathog ; 17(8): e1009787, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34339468

RESUMEN

The Gram-negative bacterium Campylobacter jejuni is a major cause of foodborne disease in humans. After infection, C. jejuni rapidly colonizes the mucus layer of the small and large intestine and induces a potent pro-inflammatory response characterized by the production of a large repertoire of cytokines, chemokines, and innate effector molecules, resulting in (bloody) diarrhea. The virulence mechanisms by which C. jejuni causes this intestinal response are still largely unknown. Here we show that C. jejuni releases a potent pro-inflammatory compound into its environment, which activates an NF-κB-mediated pro-inflammatory response including the induction of CXCL8, CXCL2, TNFAIP2 and PTGS2. This response was dependent on a functional ALPK1 receptor and independent of Toll-like Receptor and Nod-like Receptor signaling. Chemical characterization, inactivation of the heptose-biosynthesis pathway by the deletion of the hldE gene and in vitro engineering identified the released factor as the LOS-intermediate ADP-heptose and/or related heptose phosphates. During C. jejuni infection of intestinal cells, the ALPK1-NF-κB axis was potently activated by released heptose metabolites without the need for a type III or type IV injection machinery. Our results classify ADP-heptose and/or related heptose phosphates as a major virulence factor of C. jejuni that may play an important role during Campylobacter infection in humans.


Asunto(s)
Infecciones por Campylobacter/inmunología , Campylobacter jejuni/inmunología , Células Epiteliales/inmunología , Inflamación/inmunología , Intestinos/inmunología , FN-kappa B/metabolismo , Proteínas Quinasas/metabolismo , Infecciones por Campylobacter/metabolismo , Infecciones por Campylobacter/microbiología , Citocinas , Células Epiteliales/metabolismo , Células Epiteliales/microbiología , Células HeLa , Humanos , Inmunidad Innata/inmunología , Inflamación/metabolismo , Inflamación/microbiología , Intestinos/microbiología , FN-kappa B/genética , Proteínas Quinasas/genética , Transducción de Señal , Virulencia , Factores de Virulencia/metabolismo
9.
Artículo en Inglés | MEDLINE | ID: mdl-36748696

RESUMEN

As part of a culturomics study to identify bacterial species associated with inflammatory bowel disease, a large collection of bacteria was isolated from patients with ulcerative colitis. Two of these isolates were tentatively identified as members of the family Erysipelotrichaceae. Following phylogenetic analysis based on 16S rRNA gene sequence and genome sequences, both strain 128T and 539T were found to be most closely related to Allobaculum stercoricanis, with G+C contents of 48.6 and 50.5 mol%, respectively, and the genome sizes of 2 864 314 and 2 580 362 base pairs, respectively. Strains 128T and 539T were strict anaerobe rods that grew in long chains between 37 and 42 °C. Scanning electron microscopy did not reveal flagella, fimbriae or visible endospores. Biochemical analysis showed nearly identical results for both strains with enzymatic activity of C4 and C8 esterases, acid phosphatase, naphthol-AS-BI-phosphohydrolase, ß-glucuronidase, N-acetyl-ß-glucosaminidase and arginine arylamidase. In addition, both strains produced indole and reduced nitrate. Major fatty acids were identified as C18:1 ω9c (oleic acid, 64.06% in 128T and 74.35% in 539T), C18:1 ω7c/C18:1 ω9t/C18:1 ω12t/UN17.834 (16.18 % in 128T and 6.22% in 539T) and C16:0 (6.23% in 128T and 7.37% in 538T). Based on these analyses two novel species are proposed, Allobaculum mucilyticum sp. nov. with the type strain 128T (=NCTC 14626T=DSM 112815T) and Allobaculum fili sp. nov. with the type strain 539T (=NCTC 14627T=DSM 112814T).


Asunto(s)
Bacilos Grampositivos , Filogenia , Humanos , Técnicas de Tipificación Bacteriana , Composición de Base , ADN Bacteriano/genética , Ácidos Grasos/química , Bacilos Grampositivos/clasificación , Bacilos Grampositivos/aislamiento & purificación , ARN Ribosómico 16S/genética , Análisis de Secuencia de ADN , Intestinos/microbiología , Colitis Ulcerosa
10.
Nature ; 546(7660): 667-670, 2017 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-28636595

RESUMEN

Rotavirus, a leading cause of severe gastroenteritis and diarrhoea in young children, accounts for around 215,000 deaths annually worldwide. Rotavirus specifically infects the intestinal epithelial cells in the host small intestine and has evolved strategies to antagonize interferon and NF-κB signalling, raising the question as to whether other host factors participate in antiviral responses in intestinal mucosa. The mechanism by which enteric viruses are sensed and restricted in vivo, especially by NOD-like receptor (NLR) inflammasomes, is largely unknown. Here we uncover and mechanistically characterize the NLR Nlrp9b that is specifically expressed in intestinal epithelial cells and restricts rotavirus infection. Our data show that, via RNA helicase Dhx9, Nlrp9b recognizes short double-stranded RNA stretches and forms inflammasome complexes with the adaptor proteins Asc and caspase-1 to promote the maturation of interleukin (Il)-18 and gasdermin D (Gsdmd)-induced pyroptosis. Conditional depletion of Nlrp9b or other inflammasome components in the intestine in vivo resulted in enhanced susceptibility of mice to rotavirus replication. Our study highlights an important innate immune signalling pathway that functions in intestinal epithelial cells and may present useful targets in the modulation of host defences against viral pathogens.


Asunto(s)
Células Epiteliales/inmunología , Células Epiteliales/virología , Inflamasomas/metabolismo , Intestinos/citología , Receptores Acoplados a Proteínas G/metabolismo , Infecciones por Rotavirus/inmunología , Infecciones por Rotavirus/virología , Rotavirus/inmunología , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Proteínas Adaptadoras de Señalización CARD/metabolismo , Caspasa 1/metabolismo , ARN Helicasas DEAD-box/metabolismo , Células Epiteliales/metabolismo , Femenino , Inmunidad Innata , Inflamasomas/química , Inflamasomas/genética , Interleucina-18/inmunología , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Péptidos y Proteínas de Señalización Intracelular , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión a Fosfato , Piroptosis , ARN Bicatenario/metabolismo , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/inmunología , Rotavirus/crecimiento & desarrollo
11.
Int J Mol Sci ; 24(9)2023 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-37175726

RESUMEN

The impact of bacterial members of the microbiota on the development of colorectal cancer (CRC) has become clear in recent years. However, exactly how bacteria contribute to the development of cancer is often still up for debate. The impact of bacteria-derived metabolites, which can influence the development of CRC either in a promoting or inhibiting manner, is undeniable. Here, we discuss the effects of the most well-studied bacteria-derived metabolites associated with CRC, including secondary bile acids, short-chain fatty acids, trimethylamine-N-oxide and indoles. We show that the effects of individual metabolites on CRC development are often nuanced and dose- and location-dependent. In the coming years, the array of metabolites involved in CRC development will undoubtedly increase further, which will emphasize the need to focus on causation and mechanisms and the clearly defined roles of bacterial species within the microbiota.


Asunto(s)
Bacterias , Neoplasias Colorrectales , Microbioma Gastrointestinal , Animales , Humanos , Bacterias/metabolismo , Ácidos y Sales Biliares/metabolismo , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Ácidos Grasos Volátiles/metabolismo , Sulfuro de Hidrógeno/metabolismo , Indoles/metabolismo , Poliaminas/metabolismo , Especies Reactivas de Oxígeno/metabolismo
12.
Gastroenterology ; 160(6): 1970-1985, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33476671

RESUMEN

BACKGROUND & AIMS: It is currently unclear whether reported changes in the gut microbiome are cause or consequence of inflammatory bowel disease (IBD). Therefore, we studied the gut microbiome of IBD-discordant and -concordant twin pairs, which offers the unique opportunity to assess individuals at increased risk of developing IBD, namely healthy cotwins from IBD-discordant twin pairs. METHODS: Fecal samples were obtained from 99 twins (belonging to 51 twin pairs), 495 healthy age-, sex-, and body mass index-matched controls, and 99 unrelated patients with IBD. Whole-genome metagenomic shotgun sequencing was performed. Taxonomic and functional (pathways) composition was compared among healthy cotwins, IBD-twins, unrelated patients with IBD, and healthy controls with multivariable (ie, adjusted for potential confounding) generalized linear models. RESULTS: No significant differences were observed in the relative abundance of species and pathways between healthy cotwins and their IBD-twins (false discovery rate <0.10). Compared with healthy controls, 13, 19, and 18 species, and 78, 105, and 153 pathways were found to be differentially abundant in healthy cotwins, IBD-twins, and unrelated patients with IBD, respectively (false discovery rate <0.10). Of these, 8 (42.1%) of 19 and 1 (5.6%) of 18 species, and 37 (35.2%) of 105 and 30 (19.6%) of 153 pathways overlapped between healthy cotwins and IBD-twins, and healthy cotwins and unrelated patients with IBD, respectively. Many of the shared species and pathways have previously been associated with IBD. The shared pathways include potentially inflammation-related pathways, for example, an increase in propionate degradation and L-arginine degradation pathways. CONCLUSIONS: The gut microbiome of healthy cotwins from IBD-discordant twin pairs displays IBD-like signatures. These IBD-like microbiome signatures might precede the onset of IBD. However, longitudinal follow-up studies are needed to infer a causal relationship.


Asunto(s)
Microbioma Gastrointestinal , Neoplasias Inflamatorias de la Mama/epidemiología , Neoplasias Inflamatorias de la Mama/microbiología , Adulto , Antígenos Bacterianos/biosíntesis , Estudios de Casos y Controles , Estudios Transversales , Heces/microbiología , Femenino , Microbioma Gastrointestinal/fisiología , Humanos , Masculino , Metagenómica , Persona de Mediana Edad , Países Bajos/epidemiología , Fenotipo , Factores de Riesgo , Sideróforos/biosíntesis , Gemelos Dicigóticos , Gemelos Monocigóticos , Adulto Joven
13.
Immunity ; 37(6): 950-2, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23244714

RESUMEN

NLRP1 has long remained an elusive member of the NOD-like-receptor family of innate immune sensors. In this issue of Immunity, Masters et al. (2012) describe its role in immune responses to stress and infection.

14.
Nature ; 523(7559): 221-5, 2015 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-25924064

RESUMEN

Inflammation is a beneficial host response to infection but can contribute to inflammatory disease if unregulated. The Th17 lineage of T helper (Th) cells can cause severe human inflammatory diseases. These cells exhibit both instability (they can cease to express their signature cytokine, IL-17A) and plasticity (they can start expressing cytokines typical of other lineages) upon in vitro re-stimulation. However, technical limitations have prevented the transcriptional profiling of pre- and post-conversion Th17 cells ex vivo during immune responses. Thus, it is unknown whether Th17 cell plasticity merely reflects change in expression of a few cytokines, or if Th17 cells physiologically undergo global genetic reprogramming driving their conversion from one T helper cell type to another, a process known as transdifferentiation. Furthermore, although Th17 cell instability/plasticity has been associated with pathogenicity, it is unknown whether this could present a therapeutic opportunity, whereby formerly pathogenic Th17 cells could adopt an anti-inflammatory fate. Here we used two new fate-mapping mouse models to track Th17 cells during immune responses to show that CD4(+) T cells that formerly expressed IL-17A go on to acquire an anti-inflammatory phenotype. The transdifferentiation of Th17 into regulatory T cells was illustrated by a change in their signature transcriptional profile and the acquisition of potent regulatory capacity. Comparisons of the transcriptional profiles of pre- and post-conversion Th17 cells also revealed a role for canonical TGF-ß signalling and consequently for the aryl hydrocarbon receptor (AhR) in conversion. Thus, Th17 cells transdifferentiate into regulatory cells, and contribute to the resolution of inflammation. Our data suggest that Th17 cell instability and plasticity is a therapeutic opportunity for inflammatory diseases.


Asunto(s)
Transdiferenciación Celular , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Células Th17/citología , Células Th17/inmunología , Animales , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Helmintiasis/inmunología , Masculino , Ratones , Nippostrongylus/inmunología , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus/inmunología
15.
Proc Natl Acad Sci U S A ; 114(45): E9626-E9634, 2017 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-29078283

RESUMEN

Immunodeficient mice reconstituted with a human immune system represent a promising tool for translational research as they may allow modeling and therapy of human diseases in vivo. However, insufficient development and function of human natural killer (NK) cells and T cell subsets limit the applicability of humanized mice for studying cancer biology and therapy. Here, we describe a human interleukin 15 (IL15) and human signal regulatory protein alpha (SIRPA) knock-in mouse on a Rag2-/- Il2rg-/- background (SRG-15). Transplantation of human hematopoietic stem and progenitor cells into SRG-15 mice dramatically improved the development and functional maturation of circulating and tissue-resident human NK and CD8+ T cells and promoted the development of tissue-resident innate lymphoid cell (ILC) subsets. Profiling of human NK cell subsets by mass cytometry revealed a highly similar expression pattern of killer inhibitory receptors and other candidate molecules in NK cell subpopulations between SRG-15 mice and humans. In contrast to nonobese diabetic severe combined immunodeficient Il2rg-/- (NSG) mice, human NK cells in SRG-15 mice did not require preactivation but infiltrated a Burkitt's lymphoma xenograft and efficiently inhibited tumor growth following treatment with the therapeutic antibody rituximab. Our humanized mouse model may thus be useful for preclinical testing of novel human NK cell-targeted and combinatory cancer immunotherapies and for studying how they elicit human antitumor immune responses in vivo.


Asunto(s)
Células Asesinas Naturales/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunidad Innata/inmunología , Subunidad gamma Común de Receptores de Interleucina/inmunología , Interleucina-15/inmunología , Linfocitos/inmunología , Ratones , Ratones SCID , Receptores Inmunológicos/inmunología , Rituximab/inmunología
16.
Immunology ; 156(4): 339-355, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30472727

RESUMEN

Sound evidence supports a role for interleukin-17 (IL-17) -producing γδ T cells and IL-17-producing helper T (Th17) cells in intestinal homeostasis, especially in intestinal barrier integrity. In the present study, we aimed to evaluate the role of IL-17 cytokine in the regulation of intestinal immunity and obesity-induced metabolic syndrome (MetS) in an experimental murine model. C57BL/6 wild-type (WT) mice and mice lacking the IL-17 cytokine receptor (IL-17RA-/- ) were fed either a control diet (CD) or a high-fat diet (HFD) for 9 weeks. Our data demonstrate that IL-17RA-/- mice are protected against obesity, but develop hyperglycemia, hyperinsulinemia and insulin resistance. In parallel, HFD-fed IL-17RA-/- mice display intense inflammation in the ileum compared with WT mice on the HFD. IL-17RA-/- mice fed the HFD exhibit impaired neutrophil migration to the intestinal mucosa and reduced gene expression of the CXCL-1 chemokine and CXCR-2 receptor in the ileum. Interestingly, the populations of neutrophils (CD11b+  Ly6G+ ) and anti-inflammatory macrophages (CD11b+  CX3CR1+ ) are increased in the mesenteric lymph nodes of these mice. IL-17RA-/- mice on the HFD also display increased commensal bacterial translocation into the bloodstream and elevated lipopolysaccharide (LPS) levels in the visceral adipose tissue (VAT). Metagenomic analysis of bacterial 16S gene revealed increased Proteobacteria and Bacteroidetes phyla, the main representatives of Gram-negative bacteria, and reduced Akkermansia muciniphila in the fecal samples of IL-17RA-/- mice fed the HFD. Together, these data indicate that the IL-17/IL-17R axis drives intestinal neutrophil migration, limits gut dysbiosis and attenuates LPS translocation to VAT, resulting in protection to MetS.


Asunto(s)
Movimiento Celular , Dieta Alta en Grasa/efectos adversos , Disbiosis/inmunología , Interleucina-17/inmunología , Intestinos/inmunología , Lipopolisacáridos/metabolismo , Síndrome Metabólico/inmunología , Neutrófilos/inmunología , Receptores de Interleucina-17/inmunología , Animales , Movimiento Celular/inmunología , Modelos Animales de Enfermedad , Masculino , Síndrome Metabólico/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/citología
17.
Immunology ; 2018 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-29722014

RESUMEN

We addressed the role of interleukin-23 (IL-23) in driving the intestinal T helper type 17 (Th17) response during obesity and metabolic syndrome progression induced by a high-fat diet (HFD). Diet-induced obese and lean mice received HFD or control diet (CTD), respectively, for 20 weeks. The nutritional, metabolic and immune parameters were examined at weeks 9 and 20. Gene and protein IL-23p19 and IL-23 receptor expression was increased in the ileum of obese wild-type mice (WT) fed the HFD for 9 weeks. Mice lacking IL-23 and fed the HFD exhibited greater weight gain, higher fat accumulation, adipocyte hypertrophy and hepatic steatosis. Notably, these mice had more glucose intolerance, insulin resistance and associated metabolic alterations, such as hyperinsulinaemia and hyperlipidaemia. IL-23 deficiency also significantly reduced protein levels of IL-17, CCL20 and neutrophil elastase in the ileum and reduced Th17 cell expansion in the mesenteric lymph nodes of the HFD mice. Of importance, IL-23-deficient mice exhibited increased gut permeability and blood bacterial translocation compared with WT mice fed HFD. Finally, metagenomics analysis of gut microbiota revealed a dramatic outgrowth of Bacteroidetes over Firmicutes phylum with the prevalence of Bacteroides genera in the faeces of IL-23-deficient mice after HFD. In summary, IL-23 appears to maintain the Th17 response and neutrophil migration into the intestinal mucosa, minimizing the gut dysbiosis and protecting against obesity and metabolic disease development in mice.

18.
J Immunol ; 193(9): 4548-57, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25267974

RESUMEN

The Gram-negative pathogen Campylobacter jejuni is the most common cause of bacterial foodborne disease worldwide. The mechanisms that lead to bacterial invasion of eukaryotic cells and massive intestinal inflammation are still unknown. In this study, we report that C. jejuni infection of mouse macrophages induces upregulation of pro-IL-1ß transcript and secretion of IL-1ß without eliciting cell death. Immunoblotting indicated cleavage of caspase-1 and IL-1ß in infected cells. In bone marrow-derived macrophages from different knockout mice, IL-1ß secretion was found to require NLRP3, ASC, and caspase-1/11 but not NLRC4. In contrast to NLRP3 activation by ATP, C. jejuni activation did not require priming of these macrophages. C. jejuni also activated the NLRP3 inflammasome in human macrophages as indicated by the presence of ASC foci and caspase-1-positive cells. Analysis of a vast array of C. jejuni mutants with defects in capsule formation, LPS biosynthesis, chemotaxis, flagella synthesis and flagellin (-like) secretion, type 6 secretion system needle protein, or cytolethal distending toxin revealed a direct correlation between the number of intracellular bacteria and NLRP3 inflammasome activation. The C. jejuni invasion-related activation of the NLRP3 inflammasome without cytotoxicity and even in nonprimed cells extends the known repertoire of bacterial inflammasome activation and likely contributes to C. jejuni-induced intestinal inflammation.


Asunto(s)
Campylobacter jejuni/inmunología , Inflamasomas/metabolismo , Animales , Infecciones por Campylobacter/genética , Infecciones por Campylobacter/inmunología , Infecciones por Campylobacter/metabolismo , Campylobacter jejuni/genética , Proteínas Portadoras/metabolismo , Caspasa 1/metabolismo , Células Cultivadas , Humanos , Interleucina-1beta/biosíntesis , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR
19.
Proc Natl Acad Sci U S A ; 110(5): 1851-6, 2013 Jan 29.
Artículo en Inglés | MEDLINE | ID: mdl-23307811

RESUMEN

A flagellin-independent caspase-1 activation pathway that does not require NAIP5 or NRLC4 is induced by the intracellular pathogen Legionella pneumophila. Here we demonstrate that this pathway requires caspase-11. Treatment of macrophages with LPS up-regulated the host components required for this caspase-11 activation pathway. Activation by Legionella differed from caspase-11 activation using previously described agonists in that Legionella caspase-11 activation was rapid and required bacteria with a functional type IV secretion system called Dot/Icm. Legionella activation of caspase-11 induced pyroptosis by a mechanism independent of the NAIP/NLRC4 and caspase-1 axis. Legionella activation of caspase-11 stimulated activation of caspase-1 through NLRP3 and ASC. Induction of caspase-11-dependent responses occurred in macrophages deficient in the adapter proteins TRIF or MyD88 but not in macrophages deficient in both signaling factors. Although caspase-11 was produced in macrophages deficient in the type-I IFN receptor, there was a severe defect in caspase-11-dependent pyroptosis in these cells. These data indicate that macrophages respond to microbial signatures to produce proteins that mediate a capsase-11 response and that the caspase-11 system provides an alternative pathway for rapid detection of an intracellular pathogen capable of evading the canonical caspase-1 activation system that responds to bacterial flagellin.


Asunto(s)
Apoptosis , Caspasas/metabolismo , Flagelina/metabolismo , Legionella pneumophila/metabolismo , Macrófagos/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/microbiología , Proteínas Adaptadoras de Señalización CARD , Proteínas de Unión al Calcio/genética , Proteínas de Unión al Calcio/metabolismo , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 1/genética , Caspasa 1/metabolismo , Caspasas/genética , Caspasas Iniciadoras , Células Cultivadas , Citocinas/metabolismo , Proteínas del Citoesqueleto/genética , Proteínas del Citoesqueleto/metabolismo , Activación Enzimática , Flagelina/genética , Interacciones Huésped-Patógeno , Immunoblotting , Legionella pneumophila/genética , Legionella pneumophila/fisiología , Macrófagos/citología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Necrosis , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo
20.
Infect Immun ; 83(12): 4740-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26416908

RESUMEN

Interferon (IFN)-inducible guanylate binding proteins (GBPs) mediate cell-autonomous host resistance to bacterial pathogens and promote inflammasome activation. The prevailing model postulates that these two GBP-controlled activities are directly linked through GBP-dependent vacuolar lysis. It was proposed that the rupture of pathogen-containing vacuoles (PVs) by GBPs destroyed the microbial refuge and simultaneously contaminated the host cell cytosol with microbial activators of inflammasomes. Here, we demonstrate that GBP-mediated host resistance and GBP-mediated inflammatory responses can be uncoupled. We show that PVs formed by the rodent pathogen Chlamydia muridarum, so-called inclusions, remain free of GBPs and that C. muridarum is impervious to GBP-mediated restrictions on bacterial growth. Although GBPs neither bind to C. muridarum inclusions nor restrict C. muridarum growth, we find that GBPs promote inflammasome activation in C. muridarum-infected macrophages. We demonstrate that C. muridarum infections induce GBP-dependent pyroptosis through both caspase-11-dependent noncanonical and caspase-1-dependent canonical inflammasomes. Among canonical inflammasomes, we find that C. muridarum and the human pathogen Chlamydia trachomatis activate not only NLRP3 but also AIM2. Our data show that GBPs support fast-kinetics processing and secretion of interleukin-1ß (IL-1ß) and IL-18 by the NLRP3 inflammasome but are dispensable for the secretion of the same cytokines at later times postinfection. Because IFN-γ fails to induce IL-1ß transcription, GBP-dependent fast-kinetics inflammasome activation can drive the preferential processing of constitutively expressed IL-18 in IFN-γ-primed macrophages in the absence of prior Toll-like receptor stimulation. Together, our results reveal that GBPs control the kinetics of inflammasome activation and thereby shape macrophage responses to Chlamydia infections.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia muridarum/inmunología , Proteínas de Unión al GTP/inmunología , Inflamasomas/inmunología , Macrófagos/inmunología , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/inmunología , Caspasas/genética , Caspasas/inmunología , Caspasas Iniciadoras , Infecciones por Chlamydia/genética , Infecciones por Chlamydia/microbiología , Infecciones por Chlamydia/patología , Chlamydia muridarum/genética , Chlamydia muridarum/patogenicidad , Chlamydia trachomatis/genética , Chlamydia trachomatis/inmunología , Chlamydia trachomatis/patogenicidad , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Fibroblastos/inmunología , Fibroblastos/microbiología , Proteínas de Unión al GTP/genética , Regulación de la Expresión Génica , Interacciones Huésped-Patógeno , Cuerpos de Inclusión/inmunología , Cuerpos de Inclusión/microbiología , Inflamasomas/genética , Interferón gamma/genética , Interferón gamma/inmunología , Interleucina-18/genética , Interleucina-18/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Cultivo Primario de Células , Transducción de Señal , Vacuolas/inmunología , Vacuolas/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA