Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
Semin Immunol ; 25(5): 352-7, 2013 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-24184013

RESUMEN

The intestine represents one of the most challenging sites for the immune system as immune cells must be able to mount an efficient response to invading pathogens while tolerating the large number and diverse array of resident commensal bacteria. Foxp3(+) regulatory T-cells (Tregs) play a non-redundant role at maintaining this balance. At the same time Treg cell differentiation and function can be modulated by the intestinal microbiota. In this review, we will discuss effector mechanisms of Treg cells in the intestine and how these cells can be influenced by the intestinal microbiota.


Asunto(s)
Bacterias/inmunología , Intestinos/inmunología , Intestinos/microbiología , Linfocitos T Reguladores/inmunología , Animales , Homeostasis , Humanos , Microbiota/inmunología , Timo/inmunología
2.
Nat Med ; 27(11): 1970-1981, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34675383

RESUMEN

Current inflammatory bowel disease (IBD) therapies are ineffective in a high proportion of patients. Combining bulk and single-cell transcriptomics, quantitative histopathology and in situ localization across three cohorts of patients with IBD (total n = 376), we identify coexpressed gene modules within the heterogeneous tissular inflammatory response in IBD that map to distinct histopathological and cellular features (pathotypes). One of these pathotypes is defined by high neutrophil infiltration, activation of fibroblasts and vascular remodeling at sites of deep ulceration. Activated fibroblasts in the ulcer bed display neutrophil-chemoattractant properties that are IL-1R, but not TNF, dependent. Pathotype-associated neutrophil and fibroblast signatures are increased in nonresponders to several therapies across four independent cohorts (total n = 343). The identification of distinct, localized, tissular pathotypes will aid precision targeting of current therapeutics and provides a biological rationale for IL-1 signaling blockade in ulcerating disease.


Asunto(s)
Enfermedades Inflamatorias del Intestino/patología , Interleucina-1/metabolismo , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Células del Estroma/inmunología , Adulto , Anciano , Femenino , Fibroblastos/metabolismo , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/genética , Masculino , Persona de Mediana Edad , Receptores de Interleucina-1/metabolismo , Transducción de Señal/fisiología , Remodelación Vascular/fisiología
3.
Sci Immunol ; 5(47)2020 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-32444476

RESUMEN

Mononuclear phagocytes (MNPs) are vital for maintaining intestinal homeostasis but, in response to acute microbial stimulation, can also trigger immunopathology, accelerating recruitment of Ly6Chi monocytes to the gut. The regulators that control monocyte tissue adaptation in the gut remain poorly understood. Interferon regulatory factor 5 (IRF5) is a transcription factor previously shown to play a key role in maintaining the inflammatory phenotype of macrophages. Here, we investigate the impact of IRF5 on the MNP system and physiology of the gut at homeostasis and during inflammation. We demonstrate that IRF5 deficiency has a limited impact on colon physiology at steady state but ameliorates immunopathology during Helicobacter hepaticus-induced colitis. Inhibition of IRF5 activity in MNPs phenocopies global IRF5 deficiency. Using a combination of bone marrow chimera and single-cell RNA-sequencing approaches, we examined the intrinsic role of IRF5 in controlling colonic MNP development. We demonstrate that IRF5 promotes differentiation of Ly6Chi monocytes into CD11c+ macrophages and controls the production of antimicrobial and inflammatory mediators by these cells. Thus, we identify IRF5 as a key transcriptional regulator of the colonic MNP system during intestinal inflammation.


Asunto(s)
Antígenos CD11/inmunología , Inflamación/inmunología , Factores Reguladores del Interferón/inmunología , Macrófagos/inmunología , Monocitos/inmunología , Animales , Helicobacter hepaticus/inmunología , Inflamación/patología , Factores Reguladores del Interferón/deficiencia , Macrófagos/patología , Ratones , Ratones Noqueados , Monocitos/patología , Fenotipo
4.
Elife ; 92020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32716298

RESUMEN

T cell cross-reactivity ensures that diverse pathogen-derived epitopes encountered during a lifetime are recognized by the available TCR repertoire. A feature of cross-reactivity where previous exposure to one microbe can alter immunity to subsequent, non-related pathogens has been mainly explored for viruses. Yet cross-reactivity to additional microbes is important to consider, especially in HIV infection where gut-intestinal barrier dysfunction could facilitate T cell exposure to commensal/pathogenic microbes. Here we evaluated the cross-reactivity of a 'public', HIV-specific, CD8 T cell-derived TCR (AGA1 TCR) using MHC class I yeast display technology. Via screening of MHC-restricted libraries comprising ~2×108 sequence-diverse peptides, AGA1 TCR specificity was mapped to a central peptide di-motif. Using the top TCR-enriched library peptides to probe the non-redundant protein database, bacterial peptides that elicited functional responses by AGA1-expressing T cells were identified. The possibility that in context-specific settings, MHC class I proteins presenting microbial peptides influence virus-specific T cell populations in vivo is discussed.


Asunto(s)
Antígenos Bacterianos/inmunología , Antígenos de Histocompatibilidad Clase I , Receptores de Antígenos de Linfocitos T/metabolismo , Reacciones Cruzadas , Células HL-60 , Humanos
5.
Artículo en Inglés | MEDLINE | ID: mdl-23818502

RESUMEN

A fundamental role of the mammalian immune system is to eradicate pathogens while minimizing immunopathology. Instigating and maintaining immunological tolerance within the intestine represents a unique challenge to the mucosal immune system. Regulatory T cells are critical for continued immune tolerance in the intestine through active control of innate and adaptive immune responses. Dynamic adaptation of regulatory T-cell populations to the intestinal tissue microenvironment is key in this process. Here, we discuss specialization of regulatory T-cell responses in the intestine, and how a breakdown in these processes can lead to chronic intestinal inflammation.


Asunto(s)
Tolerancia Inmunológica , Intestinos/inmunología , Modelos Inmunológicos , Linfocitos T Reguladores/inmunología , Células Presentadoras de Antígenos/inmunología , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/inmunología , Humanos , Inmunidad Celular , Interleucina-10/inmunología , Intestinos/citología , Microbiota/fisiología , Factor de Crecimiento Transformador beta/fisiología
6.
J Exp Med ; 209(7): 1309-24, 2012 Jul 02.
Artículo en Inglés | MEDLINE | ID: mdl-22734048

RESUMEN

Chronic inflammation of the intestine has been associated with an elevated risk of developing colorectal cancer. Recent association studies have highlighted the role of genetic predisposition in the etiology of colitis and started to unravel its complexity. However, the genetic factors influencing the progression from colon inflammation to tumorigenesis are not known. We report the identification of a genetic interval Hiccs that regulates Helicobacter hepaticus-induced colitis and associated cancer susceptibility in a 129.RAG(-/-) mouse model. The 1.7-Mb congenic interval on chromosome 3, containing eight genes and five microRNAs, renders susceptible mice resistant to colitis and reduces tumor incidence and multiplicity. Bone marrow chimera experiments showed that resistance is conferred by the hematopoietic compartment. Moreover, the Hiccs locus controls the induction of the innate inflammatory response by regulating cytokine expression and granulocyte recruitment by Thy1(+) innate lymphoid cells. Using a tumor-promoting model combining chronic Helicobacter hepaticus infection and the carcinogen azoxymethane, we found that Hiccs also regulates the frequency of colitis-associated neoplasia. Our study highlights the importance of innate immune cells and their genetic configuration in driving progression from inflammation toward cancer and opens the door for analysis of these pathways in human inflammatory disorders and associated cancers.


Asunto(s)
Colitis/genética , Neoplasias Colorrectales/genética , Sitios Genéticos/genética , Predisposición Genética a la Enfermedad/genética , Infecciones por Helicobacter/genética , Animales , Azoximetano/toxicidad , Carcinógenos/toxicidad , Mapeo Cromosómico , Cromosomas de los Mamíferos/genética , Colitis/inmunología , Colitis/microbiología , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/microbiología , Resistencia a la Enfermedad/efectos de los fármacos , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Helicobacter hepaticus/inmunología , Helicobacter hepaticus/fisiología , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Innata/genética , Inmunidad Innata/inmunología , Inflamación/genética , Inflamación/inmunología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Polimorfismo de Nucleótido Simple , Telómero/genética
7.
Eur J Immunol ; 36(11): 2832-6, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17051620

RESUMEN

Regulatory T cells (Treg) provide protection from autoimmune disease, graft-versus-host disease, transplant rejection and overwhelming tissue destruction during infections. Conversely, high Treg numbers enable cancer cells to evade the host immune response. Thus, Treg are seen as an important tool to manipulate the immune response. However, as the immunological community is trying to move this knowledge from mice to humans, contradictory results regarding the number and function of Treg in various diseases are appearing. This problem arises because we cannot clearly define Treg populations on the basis of expression of CD25 and other cell surface markers in humans. This review addresses the utility of the FOXP3 forkhead transcription factor for the identification of Treg populations and summarizes recent data on the expression of FOXP3 in lymphomas. It is crucial to really understand Treg biology before attempting therapies, including (i) the injection of expanded Treg to cure autoimmune disease or prevent graft-versus-host disease or (ii) the depletion or inhibition of Treg in cancer therapy. For instance, new data arising from the study of haematological malignancies highlight the additional complexity of systems where malignant cell populations may also be direct Treg targets.


Asunto(s)
Factores de Transcripción Forkhead/análisis , Factores de Transcripción Forkhead/metabolismo , Neoplasias/inmunología , Linfocitos T Reguladores/química , Linfocitos T Reguladores/inmunología , Animales , Biomarcadores/análisis , Factores de Transcripción Forkhead/agonistas , Humanos , Depleción Linfocítica , Ratones , Neoplasias/química , Neoplasias/terapia , Linfocitos T Reguladores/efectos de los fármacos
8.
Gastroenterology ; 131(5): 1475-85, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17064701

RESUMEN

BACKGROUND & AIMS: Many models of autoimmunity are associated with lymphopenia. Most involve a T-helper cell (Th)1-type disease, including the diabetic BioBreeding (BB) rat. To investigate the roles of identified susceptibility loci in disease pathogenesis, we bred PVG-RT1(u), lymphopenia (lyp)/lyp rats, congenic for the iddm1 (RT1(u)) and iddm2 (lyp, Gimap5(-/-)) diabetes susceptibility loci on the PVG background. Surprisingly, these rats developed a spontaneous, progressive, inflammatory bowel disease. To understand the disease pathogenesis, we undertook investigations at the genetic, histologic, and cellular levels. METHODS: Genetically lymphopenic rats and congenic wild-type partners were compared for gross pathologic, histologic, and immunologic parameters, the latter including cytokines and autoantibodies. RESULTS: Genetic analysis demonstrated that homozygosity at the lyp locus was required for disease. All rats developed disease, and the median age at humane killing was approximately 36 weeks. This panintestinal disease showed a conspicuous eosinophilic infiltrate in the submucosa and muscle layers, but the villi were unaffected. Diseased rats showed splenomegaly and massive enlargement of the mesenteric lymph nodes. This pathology resembles human eosinophilic gastroenteritis, and several further features indicate a Th2 basis. The rats developed high serum IgE and made IgG autoantibodies that detected a nonleukocytic cell present in the intestinal wall of all rats (including germ free). CONCLUSIONS: The T-lymphopenic state associated with GIMAP5 deficiency renders rats generally susceptible to T-cell-mediated autoimmunity, but the immunoregulatory bias (Th1/Th2) of any disease depends on other genetic (or environmental) factors. In the present model, we suggest that defective peripheral tolerance to an intestine-specific autoantigen leads to uncontrolled inflammation of the intestinal wall.


Asunto(s)
Modelos Animales de Enfermedad , Enteritis/genética , Eosinofilia/genética , Linfopenia/genética , Células Th2/inmunología , Animales , Autoanticuerpos/sangre , Linfocitos B/inmunología , Enteritis/inmunología , Eosinofilia/inmunología , Haplotipos , Antígenos de Histocompatibilidad/genética , Inmunoglobulina E/sangre , Intestinos/inmunología , Ratas , Ratas Endogámicas BB
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA