Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 24(12): 2108-2120, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37932457

RESUMEN

Regulatory T cells (Treg cells) are instrumental in establishing immunological tolerance. However, the precise effector mechanisms by which Treg cells control a specific type of immune response in a given tissue remains unresolved. By simultaneously studying Treg cells from different tissue origins under systemic autoimmunity, in the present study we show that interleukin (IL)-27 is specifically produced by intestinal Treg cells to regulate helper T17 cell (TH17 cell) immunity. Selectively increased intestinal TH17 cell responses in mice with Treg cell-specific IL-27 ablation led to exacerbated intestinal inflammation and colitis-associated cancer, but also helped protect against enteric bacterial infection. Furthermore, single-cell transcriptomic analysis has identified a CD83+CD62Llo Treg cell subset that is distinct from previously characterized intestinal Treg cell populations as the main IL-27 producers. Collectively, our study uncovers a new Treg cell suppression mechanism crucial for controlling a specific type of immune response in a particular tissue and provides further mechanistic insights into tissue-specific Treg cell-mediated immune regulation.


Asunto(s)
Interleucina-27 , Linfocitos T Reguladores , Ratones , Animales , Linfocitos T Colaboradores-Inductores , Tolerancia Inmunológica , Inmunidad Celular , Células Th17
2.
Immunity ; 48(5): 839-841, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29768168

RESUMEN

Molecular mechanisms connecting the gut-brain axis to immunity remain elusive. In this issue of Immunity, Labed et al. (2018) demonstrate that two evolutionarily conserved signaling mechanisms, the neuronal muscarinic and the epithelial Wnt pathways, together induce antimicrobial peptide expression that protects Caenorhabditis elegans against intestinal infection.


Asunto(s)
Proteínas de Caenorhabditis elegans , Vía de Señalización Wnt , Acetilcolina , Animales , Antiinfecciosos , Caenorhabditis elegans , Colinérgicos , Emociones
3.
Infect Immun ; 92(5): e0009924, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38557196

RESUMEN

The mouse pathogen Citrobacter rodentium is utilized as a model organism for studying infections caused by the human pathogens enteropathogenic Escherichia coli (EPEC) and enterohemorrhagic E. coli (EHEC) and to elucidate mechanisms of mucosal immunity. In response to C. rodentium infection, innate lymphoid cells and T cells secrete interleukin (IL)-22, a cytokine that promotes mucosal barrier function. IL-22 plays a pivotal role in enabling mice to survive and recover from C. rodentium infection, although the exact mechanisms involved remain incompletely understood. Here, we investigated whether particular components of the host response downstream of IL-22 contribute to the cytokine's protective effects during C. rodentium infection. In line with previous research, mice lacking the IL-22 gene (Il22-/- mice) were highly susceptible to C. rodentium infection. To elucidate the role of specific antimicrobial proteins modulated by IL-22, we infected the following knockout mice: S100A9-/- (calprotectin), Lcn2-/- (lipocalin-2), Reg3b-/- (Reg3ß), Reg3g-/- (Reg3γ), and C3-/- (C3). All knockout mice tested displayed a considerable level of resistance to C. rodentium infection, and none phenocopied the lethality observed in Il22-/- mice. By investigating another arm of the IL-22 response, we observed that C. rodentium-infected Il22-/- mice exhibited an overall decrease in gene expression related to intestinal barrier integrity as well as significantly elevated colonic inflammation, gut permeability, and pathogen levels in the spleen. Taken together, these results indicate that host resistance to lethal C. rodentium infection may depend on multiple antimicrobial responses acting in concert, or that other IL-22-regulated processes, such as tissue repair and maintenance of epithelial integrity, play crucial roles in host defense to attaching and effacing pathogens.


Asunto(s)
Citrobacter rodentium , Infecciones por Enterobacteriaceae , Interleucina-22 , Animales , Ratones , Citrobacter rodentium/inmunología , Modelos Animales de Enfermedad , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/microbiología , Interleucina-22/genética , Interleucina-22/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Pancreatitis/genética , Proteínas Asociadas a Pancreatitis/metabolismo , Proteínas Asociadas a Pancreatitis/inmunología
4.
Proc Natl Acad Sci U S A ; 117(40): 24998-25007, 2020 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-32958643

RESUMEN

Infections elicit immune adaptations to enable pathogen resistance and/or tolerance and are associated with compositional shifts of the intestinal microbiome. However, a comprehensive understanding of how infections with pathogens that exhibit distinct capability to spread and/or persist differentially change the microbiome, the underlying mechanisms, and the relative contribution of individual commensal species to immune cell adaptations is still lacking. Here, we discovered that mouse infection with a fast-spreading and persistent (but not a slow-spreading acute) isolate of lymphocytic choriomeningitis virus induced large-scale microbiome shifts characterized by increased Verrucomicrobia and reduced Firmicute/Bacteroidetes ratio. Remarkably, the most profound microbiome changes occurred transiently after infection with the fast-spreading persistent isolate, were uncoupled from sustained viral loads, and were instead largely caused by CD8 T cell responses and/or CD8 T cell-induced anorexia. Among the taxa enriched by infection with the fast-spreading virus, Akkermansia muciniphila, broadly regarded as a beneficial commensal, bloomed upon starvation and in a CD8 T cell-dependent manner. Strikingly, oral administration of A. muciniphila suppressed selected effector features of CD8 T cells in the context of both infections. Our findings define unique microbiome differences after chronic versus acute viral infections and identify CD8 T cell responses and downstream anorexia as driver mechanisms of microbial dysbiosis after infection with a fast-spreading virus. Our data also highlight potential context-dependent effects of probiotics and suggest a model in which changes in host behavior and downstream microbiome dysbiosis may constitute a previously unrecognized negative feedback loop that contributes to CD8 T cell adaptations after infections with fast-spreading and/or persistent pathogens.


Asunto(s)
Anorexia/inmunología , Antígenos CD8/inmunología , Memoria Inmunológica/inmunología , Coriomeningitis Linfocítica/inmunología , Virosis/inmunología , Akkermansia , Animales , Anorexia/microbiología , Anorexia/virología , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Disbiosis/inmunología , Disbiosis/microbiología , Disbiosis/virología , Firmicutes/inmunología , Firmicutes/metabolismo , Microbioma Gastrointestinal/inmunología , Humanos , Coriomeningitis Linfocítica/microbiología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Ratones , Linfocitos T/inmunología , Linfocitos T/microbiología , Verrucomicrobia/inmunología , Verrucomicrobia/patogenicidad , Virosis/microbiología , Virosis/patología
5.
J Infect Dis ; 223(12 Suppl 2): S307-S313, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-33330928

RESUMEN

Infections caused by Gram-negative bacteria can be challenging to treat due to the outer membrane permeability barrier and the increasing emergence of antibiotic resistance. During infection, Gram-negative pathogens must acquire iron, an essential nutrient, in the host. Many Gram-negative bacteria utilize sophisticated iron acquisition machineries based on siderophores, small molecules that bind iron with high affinity. In this review, we provide an overview of siderophore-mediated iron acquisition in Enterobacteriaceae and show how these systems provide a foundation for the conceptualization and development of approaches to prevent and/or treat bacterial infections. Differences between the siderophore-based iron uptake machineries of pathogenic Enterobacteriaceae and commensal microbes may lead to the development of selective "Trojan-horse" antimicrobials and immunization strategies that will not harm the host microbiota.


Asunto(s)
Antibacterianos/metabolismo , Enterobacteriaceae/efectos de los fármacos , Hierro/metabolismo , Sideróforos/metabolismo , Animales , Bacteriocinas/metabolismo , Bacteriocinas/farmacología , Enterobacteriaceae/crecimiento & desarrollo , Enterobacteriaceae/metabolismo , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Gramnegativas/crecimiento & desarrollo , Bacterias Gramnegativas/metabolismo , Humanos , Inmunización , Microbiota/efectos de los fármacos , Sideróforos/inmunología
6.
Nature ; 580(7805): 594-595, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32313162
7.
Gut ; 67(8): 1434-1444, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-28779026

RESUMEN

OBJECTIVE: Nuclear receptors are known to regulate both immune and barrier functions in the GI tract. The nuclear orphan receptor NR2F6 has been shown to suppress the expression of proinflammatory cytokines in T lymphocytes. NR2F6 gene expression is reduced in patients with IBS or UC, but its functional role and tissue dependency in healthy and inflamed gut have not yet been investigated. DESIGN: Intestinal inflammation was induced in wild-type, Nr2f6-deficient, Rag1-deficient or bone marrow-reconstituted mice by administration of chemical (dextran sodium sulfate (DSS)) and immunogenic (T cell transfer) triggers. Disease phenotypes were investigated by survival, body weight, colon length and analysis of immune cell infiltrates. Additionally, histology, intestinal permeability, tight junction proteins, bacterial fluorescence in situ hybridisation, apoptosis, cell proliferation and mucus production were investigated. RESULTS: Nr2f6-deficient mice were highly susceptible to DSS-induced colitis characterised by enhanced weight loss, increased colonic tissue destruction and immune cell infiltration together with enhanced intestinal permeability and reduced Muc2 expression. T cell transfer colitis and bone marrow reconstitution experiments demonstrated that disease susceptibility was not dependent on the expression of Nr2f6 in the immune compartment but on the protective role of NR2F6 in the intestinal epithelium. Mechanistically, we show that NR2F6 binds to a consensus sequence at -2 kb of the Muc2 promoter and transactivates Muc2 expression. Loss of NR2F6 alters intestinal permeability and results in spontaneous late-onset colitis in Nr2f6-deficient mice. CONCLUSION: We have for the first time identified a fundamental and non-redundant role of NR2F6 in protecting gut barrier homeostasis.


Asunto(s)
Factores de Transcripción COUP/metabolismo , Colitis/metabolismo , Colitis/patología , Animales , Colitis/etiología , Sulfato de Dextran , Modelos Animales de Enfermedad , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Mucina 2/metabolismo , Proteínas Represoras , Proteínas de Uniones Estrechas/metabolismo
8.
Gut ; 67(10): 1813-1823, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-28877980

RESUMEN

OBJECTIVE: Nicotinamide phosphoribosyltransferase (NAMPT, also referred to as pre-B cell colony-enhancing factor or visfatin) is critically required for the maintenance of cellular nicotinamide adenine dinucleotide (NAD) supply catalysing the rate-limiting step of the NAD salvage pathway. NAMPT is strongly upregulated in inflammation including IBD and counteracts an increased cellular NAD turnover mediated by NAD-depleting enzymes. These constitute an important mechanistic link between inflammatory, metabolic and transcriptional pathways and NAD metabolism. DESIGN: We investigated the impact of NAMPT inhibition by the small-molecule inhibitor FK866 in the dextran sulfate sodium (DSS) model of colitis and the azoxymethane/DSS model of colitis-associated cancer. The impact of NAD depletion on differentiation of mouse and human primary monocytes/macrophages was studied in vitro. Finally, we tested the efficacy of FK866 compared with dexamethasone and infliximab in lamina propria mononuclear cells (LPMNC) isolated from patients with IBD. RESULTS: FK866 ameliorated DSS-induced colitis and suppressed inflammation-associated tumorigenesis in mice. FK866 potently inhibited NAMPT activity as demonstrated by reduced mucosal NAD, resulting in reduced abundances and activities of NAD-dependent enzymes including PARP1, Sirt6 and CD38, reduced nuclear factor kappa B activation, and decreased cellular infiltration by inflammatory monocytes, macrophages and activated T cells. Remarkably, FK866 effectively supressed cytokine release from LPMNCs of patients with IBD. As FK866 was also effective in Rag1-/- mice, we mechanistically linked FK866 treatment with altered monocyte/macrophage biology and skewed macrophage polarisation by reducing CD86, CD38, MHC-II and interleukin (IL)-6 and promoting CD206, Egr2 and IL-10. CONCLUSION: Our data emphasise the importance of NAD immunometabolism for mucosal immunity and highlight FK866-mediated NAMPT blockade as a promising therapeutic approach in acute intestinal inflammation.


Asunto(s)
Acrilamidas/farmacología , Diferenciación Celular/efectos de los fármacos , Colitis Ulcerosa , Neoplasias del Colon , Dexametasona/farmacología , Infliximab/farmacología , NAD/metabolismo , Nicotinamida Fosforribosiltransferasa/metabolismo , Piperidinas/farmacología , Animales , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/metabolismo , Neoplasias del Colon/inmunología , Neoplasias del Colon/metabolismo , Metabolismo Energético , Fármacos Gastrointestinales/farmacología , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Monocitos/metabolismo , Monocitos/patología
9.
Gut ; 67(5): 891-901, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-28550049

RESUMEN

OBJECTIVE: Alcoholic liver disease (ALD) is a global health problem with limited therapeutic options. Intestinal barrier integrity and the microbiota modulate susceptibility to ALD. Akkermansia muciniphila, a Gram-negative intestinal commensal, promotes barrier function partly by enhancing mucus production. The aim of this study was to investigate microbial alterations in ALD and to define the impact of A. muciniphila administration on the course of ALD. DESIGN: The intestinal microbiota was analysed in an unbiased approach by 16S ribosomal DNA (rDNA) sequencing in a Lieber-DeCarli ALD mouse model, and faecal A. muciniphila abundance was determined in a cohort of patients with alcoholic steatohepatitis (ASH). The impact of A. muciniphila on the development of experimental acute and chronic ALD was determined in a preventive and therapeutic setting, and intestinal barrier integrity was analysed. RESULTS: Patients with ASH exhibited a decreased abundance of faecal A. muciniphila when compared with healthy controls that indirectly correlated with hepatic disease severity. Ethanol feeding of wild-type mice resulted in a prominent decline in A. muciniphila abundance. Ethanol-induced intestinal A. muciniphila depletion could be restored by oral A. muciniphila supplementation. Furthermore, A. muciniphila administration when performed in a preventive setting decreased hepatic injury, steatosis and neutrophil infiltration. A. muciniphila also protected against ethanol-induced gut leakiness, enhanced mucus thickness and tight-junction expression. In already established ALD, A. muciniphila used therapeutically ameliorated hepatic injury and neutrophil infiltration. CONCLUSION: Ethanol exposure diminishes intestinal A. muciniphila abundance in both mice and humans and can be recovered in experimental ALD by oral supplementation. A. muciniphila promotes intestinal barrier integrity and ameliorates experimental ALD. Our data suggest that patients with ALD might benefit from A. muciniphila supplementation.


Asunto(s)
Etanol/efectos adversos , Microbioma Gastrointestinal/fisiología , Hepatopatías Alcohólicas/microbiología , Verrucomicrobia/efectos de los fármacos , Adulto , Anciano , Animales , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Heces/microbiología , Femenino , Técnica del Anticuerpo Fluorescente , Microbioma Gastrointestinal/genética , Humanos , Inmunohistoquímica , Hígado , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Verrucomicrobia/fisiología
10.
Eur J Immunol ; 45(11): 3073-86, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26332507

RESUMEN

Lipocalin-2 (Lcn2) is an innate immune peptide with pleiotropic effects. Lcn2 binds iron-laden bacterial siderophores, chemo-attracts neutrophils and has immunomodulatory and apoptosis-regulating effects. In this study, we show that upon infection with Salmonella enterica serovar Typhimurium, Lcn2 promotes iron export from Salmonella-infected macrophages, which reduces cellular iron content and enhances the generation of pro-inflammatory cytokines. Lcn2 represses IL-10 production while augmenting Nos2, TNF-α, and IL-6 expression. Lcn2(-/-) macrophages have elevated IL-10 levels as a consequence of increased iron content. The crucial role of Lcn-2/IL-10 interactions was further demonstrated by the greater ability of Lcn2(-/-) IL-10(-/-) macrophages and mice to control intracellular Salmonella proliferation in comparison to Lcn2(-/-) counterparts. Overexpression of the iron exporter ferroportin-1 in Lcn2(-/-) macrophages represses IL-10 and restores TNF-α and IL-6 production to the levels found in wild-type macrophages, so that killing and clearance of intracellular Salmonella is promoted. Our observations suggest that Lcn2 promotes host resistance to Salmonella Typhimurium infection by binding bacterial siderophores and suppressing IL-10 production, and that both functions are linked to its ability to shuttle iron from macrophages.


Asunto(s)
Proteínas de Fase Aguda/inmunología , Homeostasis/inmunología , Hierro/metabolismo , Lipocalinas/inmunología , Macrófagos/metabolismo , Proteínas Oncogénicas/inmunología , Salmonelosis Animal/inmunología , Proteínas de Fase Aguda/metabolismo , Animales , Western Blotting , Lipocalina 2 , Lipocalinas/metabolismo , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Salmonelosis Animal/metabolismo , Salmonella typhimurium , Transfección
11.
J Exp Med ; 221(1)2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-38085267

RESUMEN

Type I interferons (IFNs) exert a broad range of biological effects important in coordinating immune responses, which have classically been studied in the context of pathogen clearance. Yet, whether immunomodulatory bacteria operate through IFN pathways to support intestinal immune tolerance remains elusive. Here, we reveal that the commensal bacterium, Bacteroides fragilis, utilizes canonical antiviral pathways to modulate intestinal dendritic cells (DCs) and regulatory T cell (Treg) responses. Specifically, IFN signaling is required for commensal-induced tolerance as IFNAR1-deficient DCs display blunted IL-10 and IL-27 production in response to B. fragilis. We further establish that IFN-driven IL-27 in DCs is critical in shaping the ensuing Foxp3+ Treg via IL-27Rα signaling. Consistent with these findings, single-cell RNA sequencing of gut Tregs demonstrated that colonization with B. fragilis promotes a distinct IFN gene signature in Foxp3+ Tregs during intestinal inflammation. Altogether, our findings demonstrate a critical role of commensal-mediated immune tolerance via tonic type I IFN signaling.


Asunto(s)
Interferón Tipo I , Interleucina-27 , Ratones , Animales , Interleucina-27/metabolismo , Linfocitos T Reguladores , Interferón Tipo I/metabolismo , Tolerancia Inmunológica , Factores de Transcripción Forkhead/metabolismo , Bacterias/metabolismo , Células Dendríticas
12.
Sci Transl Med ; 16(735): eadi1501, 2024 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-38381845

RESUMEN

Acute graft-versus-host disease (aGVHD) is a life-threatening complication of allogeneic hematopoietic cell transplantation (allo-HCT), for which therapeutic options are limited. Strategies to promote intestinal tissue tolerance during aGVHD may improve patient outcomes. Using single-cell RNA sequencing, we identified a lipocalin-2 (LCN2)-expressing neutrophil population in mice with intestinal aGVHD. Transfer of LCN2-overexpressing neutrophils or treatment with recombinant LCN2 reduced aGVHD severity, whereas the lack of epithelial or hematopoietic LCN2 enhanced aGVHD severity and caused microbiome alterations. Mechanistically, LCN2 induced insulin-like growth factor 1 receptor (IGF-1R) signaling in macrophages through the LCN2 receptor SLC22A17, which increased interleukin-10 (IL-10) production and reduced major histocompatibility complex class II (MHCII) expression. Transfer of LCN2-pretreated macrophages reduced aGVHD severity but did not reduce graft-versus-leukemia effects. Furthermore, LCN2 expression correlated with IL-10 expression in intestinal biopsies in multiple cohorts of patients with aGVHD, and LCN2 induced IGF-1R signaling in human macrophages. Collectively, we identified a LCN2-expressing intestinal neutrophil population that reduced aGVHD severity by decreasing MHCII expression and increasing IL-10 production in macrophages. This work provides the foundation for administration of LCN2 as a therapeutic approach for aGVHD.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Humanos , Animales , Ratones , Neutrófilos/patología , Interleucina-10 , Lipocalina 2/genética , Enfermedad Injerto contra Huésped/genética , Macrófagos/patología , Enfermedad Aguda
13.
J Hepatol ; 59(6): 1315-22, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23928404

RESUMEN

BACKGROUND & AIMS: Severe obesity is associated with a state of chronic inflammation. Sirtuins (SIRT) are a family of conserved enzymes which are able to affect many metabolic and inflammatory pathways thereby potentially improving health and increasing lifespan. METHODS: We investigated the effect of weight loss on subcutaneous adipose tissue and liver mRNA and immunohistochemical expression of SIRT1, SIRT3, and SIRT6. Twenty-nine severely obese patients undergoing laparoscopic adjustable gastric banding (LAGB) were studied. Tissue samples were collected before and 6months after LAGB surgery. Tissue mRNA expression levels of SIRT1, SIRT3, and SIRT6 were correlated with clinical, biochemical, and histological parameters. In vitro, we studied sirtuin expression in native and stimulated monocytes, adipocytes, and hepatocytes. RESULTS: SIRT1, SIRT3, and SIRT6 mRNA expression was higher in the subcutaneous adipose tissue than in the liver. Weight loss resulted in a significant induction of SIRT1, SIRT3, and SIRT6 expression in the subcutaneous adipose tissue. In the liver, a significant increase after weight loss was observed, particularly for SIRT3 and SIRT6 mRNA expression; immunohistochemically, SIRT1 and SIRT3 expression was upregulated. Endotoxin and tumor necrosis factor-alpha suppressed SIRT1, SIRT3, and SIRT6 expression in human monocytes. The same stimuli suppressed total sirtuin deacetylase activity again, mainly in monocytes and less in adipocytes and hepatocytes. CONCLUSIONS: The relative abundance of adipose tissue mRNA expression of certain sirtuins exceeds its expression in the liver. Extensive weight loss increases sirtuin expression significantly both in adipose tissue and liver, probably as a consequence of reduced inflammation.


Asunto(s)
Hígado/metabolismo , Obesidad Mórbida/metabolismo , Sirtuina 1/genética , Sirtuina 3/genética , Sirtuinas/genética , Grasa Subcutánea/metabolismo , Pérdida de Peso , Adulto , Anciano , Femenino , Humanos , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Sirtuina 1/análisis , Sirtuina 3/análisis , Sirtuinas/análisis , Factor de Necrosis Tumoral alfa/farmacología
14.
Can J Physiol Pharmacol ; 91(11): 867-72, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24117253

RESUMEN

The innate immune system and its major mediators, i.e., cytokines, are increasingly recognized as being of crucial importance in metabolic inflammation as observed in morbid obesity and type 2 diabetes (T2D). Morbid obesity is commonly associated with adipose tissue inflammation. Adipose tissue inflammation is characterized by an increased expression of various pro-inflammatory cytokines such as tumor necrosis factor-alpha, interleukin-1 and -6, and by a rather heterogenous cellular infiltrate including monocytes/macrophages, neutrophils, B lymphocytes, T lymphocytes, and others. It has been demonstrated that in patients with severe obesity and fatty liver disease, expression of these pro-inflammatory cytokines in adipose tissue is 100-1000 times higher compared with that in the liver. Therefore, the adipose tissue can be considered in the state of severe obesity as the "cytokine factory" of the body. Rapid weight loss almost entirely eliminates pro-inflammatory cytokines in the adipose tissue, and therefore provides a very potent anti-inflammatory strategy. In conclusion, there is increasing evidence that peripheral tissues such as the adipose tissue may affect disease processes in target organs such as the liver, pancreas, heart, or blood vessels, and may therefore significantly contribute to chronic inflammation as observed in obesity and T2D.


Asunto(s)
Tejido Adiposo/fisiología , Citocinas/fisiología , Inflamación/metabolismo , Inflamación/fisiopatología , Hígado/fisiología , Adipoquinas/fisiología , Tejido Adiposo/fisiopatología , Animales , Humanos , Interleucina-1/fisiología , Interleucina-6/fisiología , Hígado/fisiopatología , Receptor Cross-Talk/fisiología , Factor de Necrosis Tumoral alfa/fisiología
15.
Curr Protoc ; 3(7): e824, 2023 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-37478288

RESUMEN

The pathogen Salmonella enterica encompasses a range of bacterial serovars that cause intestinal inflammation and systemic infections in humans. Mice are a widely used infection model due to their relative simplicity and versatility. Here, we provide standardized protocols for culturing the prolific zoonotic pathogen S. enterica serovar Typhimurium for intragastric inoculation of mice to model colitis or systemic dissemination, along with techniques for direct extraintestinal infection. Furthermore, we present procedures for quantifying pathogen burden and for characterizing the immune response by analyzing tissue pathology, inflammatory markers, and immune cells from intestinal tissues. © 2023 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Murine colitis model utilizing oral streptomycin pretreatment and oral S. Typhimurium administration Basic Protocol 2: Intraperitoneal injection of S. Typhimurium for modeling extraintestinal infection Support Protocol 1: Preparation of S. Typhimurium inoculum Support Protocol 2: Preparation of mixed S. Typhimurium inoculum for competitive infection Basic Protocol 3: Assessment of S. Typhimurium burden Support Protocol 3: Preservation and pathological assessment of S. Typhimurium-infected tissues Support Protocol 4: Measurement of inflammatory marker expression in intestinal tissues by qPCR Support Protocol 5: Preparation of intestinal content for inflammatory marker quantification by ELISA Support Protocol 6: Immune cell isolation from Salmonella-infected intestinal tissues.


Asunto(s)
Colitis , Infecciones por Salmonella , Humanos , Ratones , Animales , Salmonella typhimurium , Modelos Animales de Enfermedad , Infecciones por Salmonella/complicaciones , Infecciones por Salmonella/patología , Intestinos/patología , Colitis/microbiología , Colitis/patología
16.
bioRxiv ; 2023 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-36865314

RESUMEN

Regulatory T (Treg) cells are instrumental in establishing immunological tolerance. However, the precise effector mechanisms by which Treg cells control a specific type of immune response in a given tissue remains unresolved. By simultaneously studying Treg cells from different tissue origins under systemic autoimmunity, here we show that IL-27 is specifically produced by intestinal Treg cells to regulate Th17 immunity. Selectively increased intestinal Th17 responses in mice with Treg cell-specific IL-27 ablation led to exacerbated intestinal inflammation and colitis-associated cancer, but also helped protect against enteric bacterial infection. Furthermore, single-cell transcriptomic analysis has identified a CD83+TCF1+ Treg cell subset that is distinct from previously characterized intestinal Treg cell populations as the main IL-27 producers. Collectively, our study uncovers a novel Treg cell suppression mechanism crucial for controlling a specific type of immune response in a particular tissue, and provides further mechanistic insights into tissue-specific Treg cell-mediated immune regulation.

17.
mBio ; 13(5): e0218422, 2022 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-36094114

RESUMEN

Inflammatory bowel diseases (IBD) are characterized by chronic inflammation of the gastrointestinal tract and profound alterations to the gut microbiome. Adherent-invasive Escherichia coli (AIEC) is a mucosa-associated pathobiont that colonizes the gut of patients with Crohn's disease, a form of IBD. Because AIEC exacerbates gut inflammation, strategies to reduce the AIEC bloom during colitis are highly desirable. To thrive in the inflamed gut, Enterobacteriaceae acquire the essential metal nutrient iron by producing and releasing siderophores. Here, we implemented an immunization-based strategy to target the siderophores enterobactin and its glucosylated derivative salmochelin to reduce the AIEC bloom in the inflamed gut. Using chemical (dextran sulfate sodium) and genetic (Il10-/- mice) IBD mouse models, we showed that immunization with enterobactin conjugated to the mucosal adjuvant cholera toxin subunit B potently elicited mucosal and serum antibodies against these siderophores. Siderophore-immunized mice exhibited lower AIEC gut colonization, diminished AIEC association with the gut mucosa, and reduced colitis severity. Moreover, Peyer's patches and the colonic lamina propria harbored enterobactin-specific B cells that could be identified by flow cytometry. The beneficial effect of siderophore immunization was primarily B cell-dependent because immunized muMT-/- mice, which lack mature B lymphocytes, were not protected during AIEC infection. Collectively, our study identified siderophores as a potential therapeutic target to reduce AIEC colonization and its association with the gut mucosa, which ultimately may reduce colitis exacerbation. Moreover, this work provides the foundation for developing monoclonal antibodies against siderophores, which could provide a narrow-spectrum strategy to target the AIEC bloom in Crohn's disease patients. IMPORTANCE Adherent-invasive Escherichia coli (AIEC) is abnormally prevalent in patients with ileal Crohn's disease and exacerbates intestinal inflammation, but treatment strategies that selectively target AIEC are unavailable. Iron is an essential micronutrient for most living organisms, and bacterial pathogens have evolved sophisticated strategies to capture iron from the host environment. AIEC produces siderophores, small, secreted molecules with a high affinity for iron. Here, we showed that immunization to elicit antibodies against siderophores promoted a reduction of the AIEC bloom, interfered with AIEC association with the mucosa, and mitigated colitis in experimental mouse models. We also established a flow cytometry-based approach to visualize and isolate siderophore-specific B cells, a prerequisite for engineering monoclonal antibodies against these molecules. Together, this work could lead to a more selective and antibiotic-sparing strategy to target AIEC in Crohn's disease patients.


Asunto(s)
Colitis , Enfermedad de Crohn , Infecciones por Escherichia coli , Enfermedades Inflamatorias del Intestino , Ratones , Animales , Sideróforos , Enfermedad de Crohn/microbiología , Interleucina-10 , Enterobactina , Sulfato de Dextran , Toxina del Cólera , Escherichia coli/genética , Adhesión Bacteriana , Colitis/prevención & control , Colitis/microbiología , Mucosa Intestinal/microbiología , Inflamación/complicaciones , Enfermedades Inflamatorias del Intestino/complicaciones , Inmunización , Antibacterianos/farmacología , Hierro , Anticuerpos Monoclonales/farmacología , Micronutrientes
18.
Nat Microbiol ; 7(2): 262-276, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35087228

RESUMEN

Ulcerative colitis (UC) is driven by disruptions in host-microbiota homoeostasis, but current treatments exclusively target host inflammatory pathways. To understand how host-microbiota interactions become disrupted in UC, we collected and analysed six faecal- or serum-based omic datasets (metaproteomic, metabolomic, metagenomic, metapeptidomic and amplicon sequencing profiles of faecal samples and proteomic profiles of serum samples) from 40 UC patients at a single inflammatory bowel disease centre, as well as various clinical, endoscopic and histologic measures of disease activity. A validation cohort of 210 samples (73 UC, 117 Crohn's disease, 20 healthy controls) was collected and analysed separately and independently. Data integration across both cohorts showed that a subset of the clinically active UC patients had an overabundance of proteases that originated from the bacterium Bacteroides vulgatus. To test whether B. vulgatus proteases contribute to UC disease activity, we first profiled B. vulgatus proteases found in patients and bacterial cultures. Use of a broad-spectrum protease inhibitor improved B. vulgatus-induced barrier dysfunction in vitro, and prevented colitis in B. vulgatus monocolonized, IL10-deficient mice. Furthermore, transplantation of faeces from UC patients with a high abundance of B. vulgatus proteases into germfree mice induced colitis dependent on protease activity. These results, stemming from a multi-omics approach, improve understanding of functional microbiota alterations that drive UC and provide a resource for identifying other pathways that could be inhibited as a strategy to treat this disease.


Asunto(s)
Bacteroides/patogenicidad , Colitis Ulcerosa/microbiología , Colitis Ulcerosa/fisiopatología , Microbioma Gastrointestinal/genética , Metagenómica/métodos , Péptido Hidrolasas/genética , Proteómica/métodos , Adulto , Animales , Proteínas Bacterianas/clasificación , Proteínas Bacterianas/genética , Bacteroides/enzimología , Estudios de Cohortes , Heces/microbiología , Femenino , Humanos , Estudios Longitudinales , Masculino , Metagenoma , Ratones , Persona de Mediana Edad , Péptido Hidrolasas/clasificación , Índice de Severidad de la Enfermedad
19.
Nat Commun ; 12(1): 7016, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34853318

RESUMEN

Zinc is an essential cofactor for bacterial metabolism, and many Enterobacteriaceae express the zinc transporters ZnuABC and ZupT to acquire this metal in the host. However, the probiotic bacterium Escherichia coli Nissle 1917 (or "Nissle") exhibits appreciable growth in zinc-limited media even when these transporters are deleted. Here, we show that Nissle utilizes the siderophore yersiniabactin as a zincophore, enabling Nissle to grow in zinc-limited media, to tolerate calprotectin-mediated zinc sequestration, and to thrive in the inflamed gut. We also show that yersiniabactin's affinity for iron or zinc changes in a pH-dependent manner, with increased relative zinc binding as the pH increases. Thus, our results indicate that siderophore metal affinity can be influenced by the local environment and reveal a mechanism of zinc acquisition available to commensal and pathogenic Enterobacteriaceae.


Asunto(s)
Enterobacteriaceae/metabolismo , Sideróforos/metabolismo , Zinc/metabolismo , Transportadoras de Casetes de Unión a ATP , Animales , Proteínas Bacterianas/metabolismo , Proteínas Portadoras , Colon/microbiología , Colon/patología , Escherichia coli/metabolismo , Proteínas de Escherichia coli , Femenino , Complejo de Antígeno L1 de Leucocito , Proteínas de Transporte de Membrana , Ratones , Ratones Endogámicos C57BL , Fenoles , Salmonella typhi , Tiazoles
20.
Mol Aspects Med ; 75: 100895, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32883564

RESUMEN

Iron is an essential micronutrient for nearly all living organisms. In addition to facilitating redox reactions, iron is bound by metalloproteins that participate in a variety of biological processes. As the bioavailability of free iron in host environments is extremely low, iron lies at the center of a battle for nutrients between microbes and their host. Mucosal surfaces such as the respiratory and gastrointestinal tracts are constantly exposed to commensal and pathogenic microorganisms. Whereas a key strategy of mammalian antimicrobial defense is to deprive microbes of iron, pathogens and some commensals have evolved effective strategies to circumvent iron limitation. Here we provide an overview of mechanisms underpinning the tug-of-war for iron between microbes and their host, with a particular focus on mucosal surfaces.


Asunto(s)
Hierro/metabolismo , Animales , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA