Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 142
Filtrar
1.
Annu Rev Immunol ; 36: 755-781, 2018 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-29677472

RESUMEN

Inflammatory bowel disease (IBD) defines a spectrum of complex disorders. Understanding how environmental risk factors, alterations of the intestinal microbiota, and polygenetic and epigenetic susceptibility impact on immune pathways is key for developing targeted therapies. Mechanistic understanding of polygenic IBD is complemented by Mendelian disorders that present with IBD, pharmacological interventions that cause colitis, autoimmunity, and multiple animal models. Collectively, this multifactorial pathogenesis supports a concept of immune checkpoints that control microbial-host interactions in the gut by modulating innate and adaptive immunity, as well as epithelial and mesenchymal cell responses. In addition to classical immunosuppressive strategies, we discuss how resetting the microbiota and restoring innate immune responses, in particular autophagy and epithelial barrier function, might be key for maintaining remission or preventing IBD. Targeting checkpoints in genetically stratified subgroups of patients with Mendelian disorder-associated IBD increasingly directs treatment strategies as part of personalized medicine.


Asunto(s)
Susceptibilidad a Enfermedades/inmunología , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/terapia , Animales , Biomarcadores , Enfermedad Crónica , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos , Disbiosis , Microbioma Gastrointestinal , Predisposición Genética a la Enfermedad , Humanos , Enfermedades Inflamatorias del Intestino/prevención & control , Terapia Molecular Dirigida , Investigación Biomédica Traslacional
2.
Nat Immunol ; 25(5): 886-901, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38609547

RESUMEN

Intestinal immune responses to microbes are controlled by the cytokine IL-10 to avoid immune pathology. Here, we use single-cell RNA sequencing of colon lamina propria leukocytes (LPLs) along with RNA-seq and ATAC-seq of purified CD4+ T cells to show that the transcription factors Blimp-1 (encoded by Prdm1) and c-Maf co-dominantly regulate Il10 while negatively regulating proinflammatory cytokines in effector T cells. Double-deficient Prdm1fl/flMaffl/flCd4Cre mice infected with Helicobacter hepaticus developed severe colitis with an increase in TH1/NK/ILC1 effector genes in LPLs, while Prdm1fl/flCd4Cre and Maffl/flCd4Cre mice exhibited moderate pathology and a less-marked type 1 effector response. LPLs from infected Maffl/flCd4Cre mice had increased type 17 responses with increased Il17a and Il22 expression and an increase in granulocytes and myeloid cell numbers, resulting in increased T cell-myeloid-neutrophil interactions. Genes over-expressed in human inflammatory bowel disease showed differential expression in LPLs from infected mice in the absence of Prdm1 or Maf, revealing potential mechanisms of human disease.


Asunto(s)
Colitis , Helicobacter hepaticus , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Proteínas Proto-Oncogénicas c-maf , Animales , Factor 1 de Unión al Dominio 1 de Regulación Positiva/genética , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Ratones , Proteínas Proto-Oncogénicas c-maf/genética , Colitis/inmunología , Colitis/genética , Humanos , Helicobacter hepaticus/inmunología , Infecciones por Helicobacter/inmunología , Ratones Endogámicos C57BL , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Mucosa Intestinal/microbiología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/genética , Regulación de la Expresión Génica , Modelos Animales de Enfermedad
3.
Cell ; 174(5): 1054-1066, 2018 08 23.
Artículo en Inglés | MEDLINE | ID: mdl-30142344

RESUMEN

Innate lymphoid cells (ILCs) are lymphocytes that do not express the type of diversified antigen receptors expressed on T cells and B cells. ILCs are largely tissue-resident cells and are deeply integrated into the fabric of tissues. The discovery and investigation of ILCs over the past decade has changed our perception of immune regulation and how the immune system contributes to the maintenance of tissue homeostasis. We now know that cytokine-producing ILCs contribute to multiple immune pathways by, for example, sustaining appropriate immune responses to commensals and pathogens at mucosal barriers, potentiating adaptive immunity, and regulating tissue inflammation. Critically, the biology of ILCs also extends beyond classical immunology to metabolic homeostasis, tissue remodeling, and dialog with the nervous system. The last 10 years have also contributed to our greater understanding of the transcriptional networks that regulate lymphocyte commitment and delineation. This, in conjunction with the recent advances in our understanding of the influence of local tissue microenvironments on the plasticity and function of ILCs, has led to a re-evaluation of their existing categorization. In this review, we distill the advances in ILC biology over the past decade to refine the nomenclature of ILCs and highlight the importance of ILCs in tissue homeostasis, morphogenesis, metabolism, repair, and regeneration.


Asunto(s)
Inmunidad Adaptativa/fisiología , Inmunidad Innata , Linfocitos/citología , Animales , Linfocitos B/inmunología , Citocinas/inmunología , Homeostasis , Humanos , Sistema Hipotálamo-Hipofisario , Inflamación/inmunología , Células Asesinas Naturales/citología , Ratones , Fenotipo , Sistema Hipófiso-Suprarrenal , Regeneración , Linfocitos T/inmunología
4.
Nat Immunol ; 21(1): 86-100, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31844327

RESUMEN

By developing a high-density murine immunophenotyping platform compatible with high-throughput genetic screening, we have established profound contributions of genetics and structure to immune variation (http://www.immunophenotype.org). Specifically, high-throughput phenotyping of 530 unique mouse gene knockouts identified 140 monogenic 'hits', of which most had no previous immunologic association. Furthermore, hits were collectively enriched in genes for which humans show poor tolerance to loss of function. The immunophenotyping platform also exposed dense correlation networks linking immune parameters with each other and with specific physiologic traits. Such linkages limit freedom of movement for individual immune parameters, thereby imposing genetically regulated 'immunologic structures', the integrity of which was associated with immunocompetence. Hence, we provide an expanded genetic resource and structural perspective for understanding and monitoring immune variation in health and disease.


Asunto(s)
Infecciones por Enterobacteriaceae/inmunología , Variación Genética/genética , Ensayos Analíticos de Alto Rendimiento/métodos , Inmunofenotipificación/métodos , Infecciones por Salmonella/inmunología , Animales , Citrobacter/inmunología , Infecciones por Enterobacteriaceae/microbiología , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Salmonella/inmunología , Infecciones por Salmonella/microbiología
5.
Nat Immunol ; 20(4): 386-396, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30890797

RESUMEN

The intestine and skin are distinct microenvironments with unique physiological functions and are continually exposed to diverse environmental challenges. Host adaptation at these sites is an active process that involves interaction between immune cells and tissue cells. Regulatory T cells (Treg cells) play a pivotal role in enforcing homeostasis at barrier surfaces, illustrated by the development of intestinal and skin inflammation in diseases caused by primary deficiency in Treg cells. Treg cells at barrier sites are phenotypically distinct from their lymphoid-organ counterparts, and these 'tissue' signatures often reflect their tissue-adapted function. We discuss current understanding of Treg cell adaptation in the intestine and skin, including unique phenotypes, functions and metabolic demands, and how increased knowledge of Treg cells at barrier sites might guide precision medicine therapies.


Asunto(s)
Intestinos/inmunología , Piel/inmunología , Linfocitos T Reguladores/inmunología , Adaptación Fisiológica , Animales , Ambiente , Humanos , Ratones , Timo/inmunología
6.
Nature ; 628(8009): 854-862, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38570678

RESUMEN

The intestinal immune system is highly adapted to maintaining tolerance to the commensal microbiota and self-antigens while defending against invading pathogens1,2. Recognizing how the diverse network of local cells establish homeostasis and maintains it in the complex immune environment of the gut is critical to understanding how tolerance can be re-established following dysfunction, such as in inflammatory disorders. Although cell and molecular interactions that control T regulatory (Treg) cell development and function have been identified3,4, less is known about the cellular neighbourhoods and spatial compartmentalization that shapes microorganism-reactive Treg cell function. Here we used in vivo live imaging, photo-activation-guided single-cell RNA sequencing5-7 and spatial transcriptomics to follow the natural history of T cells that are reactive towards Helicobacter hepaticus through space and time in the settings of tolerance and inflammation. Although antigen stimulation can occur anywhere in the tissue, the lamina propria-but not embedded lymphoid aggregates-is the key microniche that supports effector Treg (eTreg) cell function. eTreg cells are stable once their niche is established; however, unleashing inflammation breaks down compartmentalization, leading to dominance of CD103+SIRPα+ dendritic cells in the lamina propria. We identify and validate the putative tolerogenic interaction between CD206+ macrophages and eTreg cells in the lamina propria and identify receptor-ligand pairs that are likely to govern the interaction. Our results reveal a spatial mechanism of tolerance in the lamina propria and demonstrate how knowledge of local interactions may contribute to the next generation of tolerance-inducing therapies.


Asunto(s)
Mucosa Intestinal , Membrana Mucosa , Linfocitos T Reguladores , Animales , Femenino , Masculino , Ratones , Antígenos CD/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Perfilación de la Expresión Génica , Helicobacter hepaticus/inmunología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/microbiología , Tolerancia Inmunológica/inmunología , Inflamación/inmunología , Inflamación/microbiología , Inflamación/patología , Cadenas alfa de Integrinas/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Membrana Mucosa/citología , Membrana Mucosa/inmunología , Receptores Inmunológicos/metabolismo , Receptores Inmunológicos/inmunología , Análisis de Expresión Génica de una Sola Célula , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/citología , Transcriptoma
7.
Immunity ; 50(4): 992-1006, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30995511

RESUMEN

Inflammatory bowel disease (IBD) is a chronic inflammatory disease of the gastrointestinal tract. Cytokine-targeted therapies have transformed the treatment of IBD, providing control of symptoms and longer relapse-free periods. However, many patients fail to respond, highlighting the need for therapies tailored to the underlying cell and molecular disease drivers. Here we discuss the progression of IBD from the perspective of remodeling of cytokine networks. We place well-established and under-studied cytokine modules in the context of cellular interactions, their dynamic regulation in early and late stages of disease (i.e., fibrosis), and their current and potential use in the clinic. Examining how particular cytokine networks drive distinct features and phases of IBD will shed light on the etiology of IBD and provide a basis for more effective treatments.


Asunto(s)
Citocinas/inmunología , Enfermedades Inflamatorias del Intestino/fisiopatología , Animales , Antirreumáticos/uso terapéutico , Citocinas/genética , Progresión de la Enfermedad , Resistencia a Medicamentos , Células Epiteliales/inmunología , Estudios de Asociación Genética , Homeostasis , Humanos , Inmunidad Innata , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/inmunología , Intestinos/inmunología , Intestinos/patología , Subgrupos Linfocitarios/inmunología , Ratones , Células Mieloides/inmunología , Factor de Transcripción STAT3/biosíntesis , Análisis de la Célula Individual , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores
8.
Immunity ; 50(2): 493-504.e7, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30737144

RESUMEN

Non-lymphoid tissues (NLTs) harbor a pool of adaptive immune cells with largely unexplored phenotype and development. We used single-cell RNA-seq to characterize 35,000 CD4+ regulatory (Treg) and memory (Tmem) T cells in mouse skin and colon, their respective draining lymph nodes (LNs) and spleen. In these tissues, we identified Treg cell subpopulations with distinct degrees of NLT phenotype. Subpopulation pseudotime ordering and gene kinetics were consistent in recruitment to skin and colon, yet the initial NLT-priming in LNs and the final stages of NLT functional adaptation reflected tissue-specific differences. Predicted kinetics were recapitulated using an in vivo melanoma-induction model, validating key regulators and receptors. Finally, we profiled human blood and NLT Treg and Tmem cells, and identified cross-mammalian conserved tissue signatures. In summary, we describe the relationship between Treg cell heterogeneity and recruitment to NLTs through the combined use of computational prediction and in vivo validation.


Asunto(s)
Adaptación Fisiológica/inmunología , Análisis de la Célula Individual/métodos , Linfocitos T Reguladores/inmunología , Transcriptoma/inmunología , Adaptación Fisiológica/genética , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Movimiento Celular/inmunología , Colon/inmunología , Colon/metabolismo , Humanos , Memoria Inmunológica/genética , Memoria Inmunológica/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/metabolismo , Ratones Transgénicos , Neoplasias Experimentales/genética , Neoplasias Experimentales/inmunología , Neoplasias Experimentales/patología , Piel/inmunología , Piel/metabolismo , Bazo/inmunología , Bazo/metabolismo , Linfocitos T Reguladores/metabolismo
9.
Immunity ; 50(2): 432-445.e7, 2019 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-30683619

RESUMEN

Host microbial cross-talk is essential to maintain intestinal homeostasis. However, maladaptation of this response through microbial dysbiosis or defective host defense toward invasive intestinal bacteria can result in chronic inflammation. We have shown that macrophages differentiated in the presence of the bacterial metabolite butyrate display enhanced antimicrobial activity. Butyrate-induced antimicrobial activity was associated with a shift in macrophage metabolism, a reduction in mTOR kinase activity, increased LC3-associated host defense and anti-microbial peptide production in the absence of an increased inflammatory cytokine response. Butyrate drove this monocyte to macrophage differentiation program through histone deacetylase 3 (HDAC3) inhibition. Administration of butyrate induced antimicrobial activity in intestinal macrophages in vivo and increased resistance to enteropathogens. Our data suggest that (1) increased intestinal butyrate might represent a strategy to bolster host defense without tissue damaging inflammation and (2) that pharmacological HDAC3 inhibition might drive selective macrophage functions toward antimicrobial host defense.


Asunto(s)
Antiinfecciosos/farmacología , Butiratos/farmacología , Diferenciación Celular/efectos de los fármacos , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Animales , Diferenciación Celular/genética , Células Cultivadas , Colon/efectos de los fármacos , Colon/metabolismo , Colon/microbiología , Citocinas/genética , Citocinas/metabolismo , Disbiosis/microbiología , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Intestinos/efectos de los fármacos , Intestinos/microbiología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Microbiota/fisiología , Monocitos/metabolismo , Monocitos/microbiología
10.
EMBO J ; 42(6): e112202, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36795015

RESUMEN

Lipids play a major role in inflammatory diseases by altering inflammatory cell functions, either through their function as energy substrates or as lipid mediators such as oxylipins. Autophagy, a lysosomal degradation pathway that limits inflammation, is known to impact on lipid availability, however, whether this controls inflammation remains unexplored. We found that upon intestinal inflammation visceral adipocytes upregulate autophagy and that adipocyte-specific loss of the autophagy gene Atg7 exacerbates inflammation. While autophagy decreased lipolytic release of free fatty acids, loss of the major lipolytic enzyme Pnpla2/Atgl in adipocytes did not alter intestinal inflammation, ruling out free fatty acids as anti-inflammatory energy substrates. Instead, Atg7-deficient adipose tissues exhibited an oxylipin imbalance, driven through an NRF2-mediated upregulation of Ephx1. This shift reduced secretion of IL-10 from adipose tissues, which was dependent on the cytochrome P450-EPHX pathway, and lowered circulating levels of IL-10 to exacerbate intestinal inflammation. These results suggest an underappreciated fat-gut crosstalk through an autophagy-dependent regulation of anti-inflammatory oxylipins via the cytochrome P450-EPHX pathway, indicating a protective effect of adipose tissues for distant inflammation.


Asunto(s)
Ácidos Grasos no Esterificados , Oxilipinas , Humanos , Adipocitos/metabolismo , Autofagia/fisiología , Sistema Enzimático del Citocromo P-450/metabolismo , Sistema Enzimático del Citocromo P-450/farmacología , Ácidos Grasos no Esterificados/metabolismo , Ácidos Grasos no Esterificados/farmacología , Inflamación/genética , Inflamación/metabolismo , Interleucina-10/genética , Oxilipinas/metabolismo
11.
Nature ; 585(7826): 509-517, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32968260

RESUMEN

Colorectal cancer (CRC) is a heterogeneous disease of the intestinal epithelium that is characterized by the accumulation of mutations and a dysregulated immune response. Up to 90% of disease risk is thought to be due to environmental factors such as diet, which is consistent with a growing body of literature that describes an 'oncogenic' CRC-associated microbiota. Whether this dysbiosis contributes to disease or merely represents a bystander effect remains unclear. To prove causation, it will be necessary to decipher which specific taxa or metabolites drive CRC biology and to fully characterize the underlying mechanisms. Here we discuss the host-microbiota interactions in CRC that have been reported so far, with particular focus on mechanisms that are linked to intestinal barrier disruption, genotoxicity and deleterious inflammation. We further comment on unknowns and on the outstanding challenges in the field, and how cutting-edge technological advances might help to overcome these. More detailed mechanistic insights into the complex CRC-associated microbiota would potentially reveal avenues that can be exploited for clinical benefit.


Asunto(s)
Neoplasias Colorrectales/microbiología , Neoplasias Colorrectales/fisiopatología , Microbioma Gastrointestinal , Animales , Neoplasias Colorrectales/inmunología , Neoplasias Colorrectales/terapia , Disbiosis/metabolismo , Disbiosis/microbiología , Microbioma Gastrointestinal/inmunología , Humanos , Inflamación/microbiología , Mutagénesis
12.
Immunity ; 43(1): 187-99, 2015 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-26200014

RESUMEN

The role of intestinal eosinophils in immune homeostasis is enigmatic and the molecular signals that drive them from protective to tissue damaging are unknown. Most commonly associated with Th2 cell-mediated diseases, we describe a role for eosinophils as crucial effectors of the interleukin-23 (IL-23)-granulocyte macrophage colony-stimulating factor (GM-CSF) axis in colitis. Chronic intestinal inflammation was characterized by increased bone marrow eosinopoiesis and accumulation of activated intestinal eosinophils. IL-5 blockade or eosinophil depletion ameliorated colitis, implicating eosinophils in disease pathogenesis. GM-CSF was a potent activator of eosinophil effector functions and intestinal accumulation, and GM-CSF blockade inhibited chronic colitis. By contrast neutrophil accumulation was GM-CSF independent and dispensable for colitis. In addition to TNF secretion, release of eosinophil peroxidase promoted colitis identifying direct tissue-toxic mechanisms. Thus, eosinophils are key perpetrators of chronic inflammation and tissue damage in IL-23-mediated immune diseases and it suggests the GM-CSF-eosinophil axis as an attractive therapeutic target.


Asunto(s)
Colitis/inmunología , Eosinófilos/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Subunidad p19 de la Interleucina-23/inmunología , Animales , Movimiento Celular/inmunología , Subunidad beta Común de los Receptores de Citocinas/genética , Peroxidasa del Eosinófilo/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Inflamación/inmunología , Interleucina-5/antagonistas & inhibidores , Intestinos/citología , Intestinos/inmunología , Intestinos/patología , Procedimientos de Reducción del Leucocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/inmunología , Factores de Necrosis Tumoral/metabolismo
13.
Nature ; 612(7939): 220-221, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36447035
14.
Gut ; 70(6): 1023-1036, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33037057

RESUMEN

OBJECTIVE: Dysregulated immune responses are the cause of IBDs. Studies in mice and humans suggest a central role of interleukin (IL)-23-producing mononuclear phagocytes in disease pathogenesis. Mechanistic insights into the regulation of IL-23 are prerequisite for selective IL-23 targeting therapies as part of personalised medicine. DESIGN: We performed transcriptomic analysis to investigate IL-23 expression in human mononuclear phagocytes and peripheral blood mononuclear cells. We investigated the regulation of IL-23 expression and used single-cell RNA sequencing to derive a transcriptomic signature of hyperinflammatory monocytes. Using gene network correlation analysis, we deconvolved this signature into components associated with homeostasis and inflammation in patient biopsy samples. RESULTS: We characterised monocyte subsets of healthy individuals and patients with IBD that express IL-23. We identified autosensing and paracrine sensing of IL-1α/IL-1ß and IL-10 as key cytokines that control IL-23-producing monocytes. Whereas Mendelian genetic defects in IL-10 receptor signalling induced IL-23 secretion after lipopolysaccharide stimulation, whole bacteria exposure induced IL-23 production in controls via acquired IL-10 signalling resistance. We found a transcriptional signature of IL-23-producing inflammatory monocytes that predicted both disease and resistance to antitumour necrosis factor (TNF) therapy and differentiated that from an IL-23-associated lymphocyte differentiation signature that was present in homeostasis and in disease. CONCLUSION: Our work identifies IL-10 and IL-1 as critical regulators of monocyte IL-23 production. We differentiate homeostatic IL-23 production from hyperinflammation-associated IL-23 production in patients with severe ulcerating active Crohn's disease and anti-TNF treatment non-responsiveness. Altogether, we identify subgroups of patients with IBD that might benefit from IL-23p19 and/or IL-1α/IL-1ß-targeting therapies upstream of IL-23.


Asunto(s)
Resistencia a Medicamentos/genética , Enfermedades Inflamatorias del Intestino/genética , Interleucina-10/genética , Subunidad p19 de la Interleucina-23/biosíntesis , Subunidad p19 de la Interleucina-23/genética , Monocitos/metabolismo , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Comunicación Autocrina , Células Cultivadas , Femenino , Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Homeostasis/genética , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Interleucina-10/metabolismo , Interleucina-1alfa/metabolismo , Interleucina-1beta/metabolismo , Lipopolisacáridos , Masculino , Persona de Mediana Edad , Monocitos/inmunología , Comunicación Paracrina , Receptores de Interleucina-10/antagonistas & inhibidores , Receptores de Interleucina-10/metabolismo , Transducción de Señal/genética , Transcriptoma , Factor de Necrosis Tumoral alfa/efectos adversos , Adulto Joven
15.
Immunity ; 37(6): 1116-29, 2012 Dec 14.
Artículo en Inglés | MEDLINE | ID: mdl-23200826

RESUMEN

In interleukin-23 (IL-23)-dependent colitis, there is excessive accumulation of short-lived neutrophils and inflammatory monocytes in the intestine. It is unknown whether this reflects changes in mature cell populations or whether the IL-23-driven colitogenic T cell program regulates upstream hematopoietic stem and progenitor cells (HSPC). Here we have shown dysregulation of hematopoiesis in colitis mediated by inflammatory cytokines. First, there was an interferon-gamma-dependent accumulation of proliferating hematopoietic stem cells in the bone marrow and spleen. Second, there was a strong skew toward granulocyte-monocyte progenitor (GMP) production at the expense of erythroid and lymphoid progenitors. Extramedullary hematopoiesis was also evident, and granulocyte macrophage-colony stimulating factor (GM-CSF) blockade reduced the accumulation of splenic and colonic GMPs, resulting in amelioration of colitis. Importantly, transfer of GMPs exacerbated colitis. These data identify HSPCs as a major target of the IL-23-driven inflammatory axis suggesting therapeutic strategies for the treatment of inflammatory bowel disease.


Asunto(s)
Colitis/inmunología , Colitis/metabolismo , Células Madre Hematopoyéticas/metabolismo , Interleucina-23/fisiología , Animales , Células de la Médula Ósea/metabolismo , Linaje de la Célula , Proliferación Celular , Colitis/inducido químicamente , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Hematopoyesis Extramedular , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/efectos de los fármacos , Interferón gamma/metabolismo , Interferón gamma/farmacología , Interleucina-23/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/fisiología , Ratones , Ratones Noqueados , Bazo/fisiología
16.
Proc Natl Acad Sci U S A ; 115(40): 10118-10123, 2018 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-30217896

RESUMEN

Intestinal epithelial cells (IECs) play a key role in regulating immune responses and controlling infection. However, the direct role of IECs in restricting pathogens remains incompletely understood. Here, we provide evidence that IL-22 primed intestinal organoids derived from healthy human induced pluripotent stem cells (hIPSCs) to restrict Salmonella enterica serovar Typhimurium SL1344 infection. A combination of transcriptomics, bacterial invasion assays, and imaging suggests that IL-22-induced antimicrobial activity is driven by increased phagolysosomal fusion in IL-22-pretreated cells. The antimicrobial phenotype was absent in hIPSCs derived from a patient harboring a homozygous mutation in the IL10RB gene that inactivates the IL-22 receptor but was restored by genetically complementing the IL10RB deficiency. This study highlights a mechanism through which the IL-22 pathway facilitates the human intestinal epithelium to control microbial infection.


Asunto(s)
Células Epiteliales/inmunología , Células Madre Pluripotentes Inducidas/inmunología , Interleucinas/inmunología , Mucosa Intestinal/inmunología , Fagosomas/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Células Epiteliales/microbiología , Células Epiteliales/patología , Humanos , Células Madre Pluripotentes Inducidas/microbiología , Células Madre Pluripotentes Inducidas/patología , Subunidad beta del Receptor de Interleucina-10/genética , Subunidad beta del Receptor de Interleucina-10/inmunología , Subunidad alfa del Receptor de Interleucina-21/genética , Subunidad alfa del Receptor de Interleucina-21/inmunología , Interleucinas/genética , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Fagosomas/genética , Fagosomas/microbiología , Fagosomas/patología , Infecciones por Salmonella/genética , Infecciones por Salmonella/patología , Salmonella typhimurium/genética , Interleucina-22
17.
Nature ; 513(7519): 564-568, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25043027

RESUMEN

FOXP3(+) regulatory T cells (Treg cells) are abundant in the intestine, where they prevent dysregulated inflammatory responses to self and environmental stimuli. It is now appreciated that Treg cells acquire tissue-specific adaptations that facilitate their survival and function; however, key host factors controlling the Treg response in the intestine are poorly understood. The interleukin (IL)-1 family member IL-33 is constitutively expressed in epithelial cells at barrier sites, where it functions as an endogenous danger signal, or alarmin, in response to tissue damage. Recent studies in humans have described high levels of IL-33 in inflamed lesions of inflammatory bowel disease patients, suggesting a role for this cytokine in disease pathogenesis. In the intestine, both protective and pathological roles for IL-33 have been described in murine models of acute colitis, but its contribution to chronic inflammation remains ill defined. Here we show in mice that the IL-33 receptor ST2 is preferentially expressed on colonic Treg cells, where it promotes Treg function and adaptation to the inflammatory environment. IL-33 signalling in T cells stimulates Treg responses in several ways. First, it enhances transforming growth factor (TGF)-ß1-mediated differentiation of Treg cells and, second, it provides a necessary signal for Treg-cell accumulation and maintenance in inflamed tissues. Strikingly, IL-23, a key pro-inflammatory cytokine in the pathogenesis of inflammatory bowel disease, restrained Treg responses through inhibition of IL-33 responsiveness. These results demonstrate a hitherto unrecognized link between an endogenous mediator of tissue damage and a major anti-inflammatory pathway, and suggest that the balance between IL-33 and IL-23 may be a key controller of intestinal immune responses.


Asunto(s)
Interleucinas/inmunología , Intestinos/citología , Intestinos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Colitis/inmunología , Colitis/patología , Colon/citología , Colon/inmunología , Colon/patología , Modelos Animales de Enfermedad , Femenino , Inmunidad Mucosa , Inflamación/inmunología , Inflamación/metabolismo , Inflamación/patología , Interleucina-23/inmunología , Interleucina-33 , Interleucinas/antagonistas & inhibidores , Interleucinas/metabolismo , Intestinos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina/metabolismo , Transducción de Señal/inmunología , Linfocitos T Reguladores/citología , Timo/citología , Factor de Crecimiento Transformador beta/metabolismo
18.
Immunity ; 32(4): 557-67, 2010 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-20399121

RESUMEN

Dendritic cells (DCs) play a pivotal role in controlling the balance between tolerance and immunity in the intestine. Gut conditioned CD103(+) DCs promote regulatory T (Treg) cell responses; however, little is known about DCs that drive inflammation in the intestine. Here, we show that monocyte-derived inflammatory DCs that express E-cadherin, the receptor for CD103, promote intestinal inflammation. E-cadherin(+) DCs accumulated in the inflamed mesenteric lymph nodes and colon, had high expression of toll-like receptors, and produced colitogenic cytokines, such as IL-6 and IL-23, after activation. Importantly, adoptive transfer of E-cadherin(+) DCs into T cell-restored immunodeficient hosts increased Th17 cell responses in the intestine and led to exacerbation of colitis. These results identify a monocyte-derived inflammatory DC subset that is associated with the pathogenesis of intestinal inflammation, providing a therapeutic target for the treatment of inflammatory bowel disease.


Asunto(s)
Cadherinas/inmunología , Colitis/inmunología , Células Dendríticas/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD/inmunología , Antígeno CD11c/inmunología , Diferenciación Celular , Movimiento Celular , Colitis/genética , Colitis/patología , Células Dendríticas/citología , Regulación de la Expresión Génica , Inmunidad Innata , Inflamación/inmunología , Cadenas alfa de Integrinas/inmunología , Ratones , Ratones Endogámicos BALB C , Monocitos/citología , Monocitos/inmunología
19.
Immunity ; 33(2): 279-88, 2010 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-20732640

RESUMEN

Mutations in the IL23R gene are linked to inflammatory bowel disease susceptibility. Experimental models have shown that interleukin-23 (IL-23) orchestrates innate and T cell-dependent colitis; however, the cell populations it acts on to induce intestinal immune pathology are unknown. Here, using Il23r(-/-) T cells, we demonstrated that T cell reactivity to IL-23 was critical for development of intestinal pathology, but not for systemic inflammation. Through direct signaling into T cells, IL-23 drove intestinal T cell proliferation, promoted intestinal Th17 cell accumulation, and enhanced the emergence of an IL-17A(+)IFN-gamma(+) population of T cells. Furthermore, IL-23R signaling in intestinal T cells suppressed the differentiation of Foxp3(+) cells and T cell IL-10 production. Although Il23r(-/-) T cells displayed unimpaired Th1 cell differentiation, these cells showed impaired proliferation and failed to accumulate in the intestine. Together, these results highlight the multiple functions of IL-23 signaling in T cells that contribute to its colitogenic activity.


Asunto(s)
Colitis/inmunología , Interleucina-23/inmunología , Linfocitos T/inmunología , Animales , Proliferación Celular , Células Cultivadas , Colitis/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Interleucina/deficiencia , Receptores de Interleucina/inmunología , Linfocitos T/citología
20.
Immunity ; 32(5): 605-15, 2010 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-20493732

RESUMEN

STAT3, an essential transcription factor with pleiotropic functions, plays critical roles in the pathogenesis of autoimmunity. Despite recent data linking STAT3 with inflammatory bowel disease, exactly how it contributes to chronic intestinal inflammation is not known. Using a T cell transfer model of colitis, we found that STAT3 expression in T cells was essential for the induction of both colitis and systemic inflammation. STAT3 was critical in modulating the balance of T helper 17 (Th17) and regulatory T (Treg) cells, as well as in promoting CD4(+) T cell proliferation. We used chromatin immunoprecipitation and massive parallel sequencing (ChIP-Seq) to define the genome-wide targets of STAT3 in CD4(+) T cells. We found that STAT3 bound to multiple genes involved in Th17 cell differentiation, cell activation, proliferation, and survival, regulating both expression and epigenetic modifications. Thus, STAT3 orchestrates multiple critical aspects of T cell function in inflammation and homeostasis.


Asunto(s)
Colitis/inmunología , Colitis/fisiopatología , Homeostasis/inmunología , Factor de Transcripción STAT3/inmunología , Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Interleucina-17/inmunología , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Noqueados , Linfocitos T/citología , Linfocitos T Reguladores/inmunología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda