Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Res Sq ; 2023 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-37961128

RESUMEN

Chronic stress underlies the etiology of both major depressive disorder (MDD) and irritable bowel syndrome (IBS), two highly prevalent and debilitating conditions with high rates of co-morbidity. However, it is not fully understood how the brain and gut bi-directionally communicate during stress to impact intestinal homeostasis and stress-relevant behaviours. Using the chronic social defeat stress (CSDS) model, we find that stressed mice display greater intestinal permeability and circulating levels of the endotoxin lipopolysaccharide (LPS) compared to unstressed control (CON) mice. Interestingly, the microbiota in the colon also exhibit elevated LPS biosynthesis gene expression following CSDS. Additionally, CSDS triggers an increase in pro-inflammatory colonic IFNγ+ Th1 cells and a decrease in IL4+ Th2 cells compared to CON mice, and this gut inflammation contributes to stress-induced intestinal barrier permeability and social avoidance behaviour. We next investigated the role of enteric neurons and identified that noradrenergic dopamine beta-hydroxylase (DBH)+ neurons in the colon are activated by CSDS, and that their ablation protects against gut pathophysiology and disturbances in social behaviour. Retrograde tracing from the colon identified a population of corticotropin-releasing hormone-expressing (CRH+) neurons in the paraventricular nucleus of the hypothalamus (PVH) that innervate the colon and are activated by stress. Chemogenetically activating these PVH CRH+ neurons is sufficient to induce gut inflammation, barrier permeability, and social avoidance behaviour, while inhibiting these cells prevents these effects following exposure to CSDS. Thus, we define a stress-activated brain-to-gut circuit that confers colonic inflammation, leading to impaired intestinal barrier function, and consequent behavioural deficits.

2.
Cell Mol Immunol ; 19(7): 777-790, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35468944

RESUMEN

The food colorant Red 40 is an environmental risk factor for colitis development in mice with increased expression of interleukin (IL)-23. This immune response is mediated by CD4+ T cells, but mechanistic insights into how these CD4+ T cells trigger and perpetuate colitis have remained elusive. Here, using single-cell transcriptomic analysis, we found that several CD4+ T-cell subsets are present in the intestines of colitic mice, including an interferon (IFN)-γ-producing subset. In vivo challenge of primed mice with Red 40 promoted rapid activation of CD4+ T cells and caused marked intestinal epithelial cell (IEC) apoptosis that was attenuated by depletion of CD4+ cells and blockade of IFN-γ. Ex vivo experiments showed that intestinal CD4+ T cells from colitic mice directly promoted apoptosis of IECs and intestinal enteroids. CD4+ T cell-mediated cytotoxicity was contact-dependent and required FasL, which promoted caspase-dependent cell death in target IECs. Genetic ablation of IFN-γ constrained IL-23- and Red 40-induced colitis development, and blockade of IFN-γ inhibited epithelial cell death in vivo. These results advance the understanding of the mechanisms regulating colitis development caused by IL-23 and food colorants and identify IFN-γ+ cytotoxic CD4+ T cells as a new potential therapeutic target for colitis.


Asunto(s)
Linfocitos T CD4-Positivos , Colitis , Colorantes de Alimentos , Interleucina-23 , Animales , Linfocitos T CD4-Positivos/inmunología , Colitis/inducido químicamente , Colitis/inmunología , Colorantes de Alimentos/efectos adversos , Interferón gamma/metabolismo , Interleucina-23/efectos adversos , Ratones , Ratones Endogámicos C57BL
3.
Cell ; 184(23): 5715-5727.e12, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34717799

RESUMEN

The enteric nervous system (ENS) controls several intestinal functions including motility and nutrient handling, which can be disrupted by infection-induced neuropathies or neuronal cell death. We investigated possible tolerance mechanisms preventing neuronal loss and disruption in gut motility after pathogen exposure. We found that following enteric infections, muscularis macrophages (MMs) acquire a tissue-protective phenotype that prevents neuronal loss, dysmotility, and maintains energy balance during subsequent challenge with unrelated pathogens. Bacteria-induced neuroprotection relied on activation of gut-projecting sympathetic neurons and signaling via ß2-adrenergic receptors (ß2AR) on MMs. In contrast, helminth-mediated neuroprotection was dependent on T cells and systemic production of interleukin (IL)-4 and IL-13 by eosinophils, which induced arginase-expressing MMs that prevented neuronal loss from an unrelated infection located in a different intestinal region. Collectively, these data suggest that distinct enteric pathogens trigger a state of disease or tissue tolerance that preserves ENS number and functionality.


Asunto(s)
Sistema Nervioso Entérico/microbiología , Sistema Nervioso Entérico/parasitología , Infecciones/microbiología , Infecciones/parasitología , Neuronas/patología , Neuroprotección , Especificidad de Órganos , Yersinia pseudotuberculosis/fisiología , Animales , Eosinófilos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Inmunidad , Infecciones/inmunología , Interleucina-13/metabolismo , Interleucina-4/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Strongyloides/fisiología , Estrongiloidiasis/genética , Estrongiloidiasis/inmunología , Estrongiloidiasis/parasitología , Transcriptoma/genética , Infecciones por Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/inmunología , Infecciones por Yersinia pseudotuberculosis/microbiología
4.
Cell Metab ; 33(7): 1358-1371.e5, 2021 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-33989521

RESUMEN

Both genetic predisposition and environmental factors appear to play a role in inflammatory bowel disease (IBD) development. Genetic studies in humans have linked the interleukin (IL)-23 signaling pathway with IBD, but the environmental factors contributing to disease have remained elusive. Here, we show that the azo dyes Red 40 and Yellow 6, the most abundant food colorants in the world, can trigger an IBD-like colitis in mice conditionally expressing IL-23, or in two additional animal models in which IL-23 expression was augmented. Increased IL-23 expression led to generation of activated CD4+ T cells that expressed interferon-γ and transferred disease to mice exposed to Red 40. Colitis induction was dependent on the commensal microbiota promoting the azo reduction of Red 40 and generation of a metabolite, 1-amino-2-naphthol-6-sulfonate sodium salt. Together these findings suggest that specific food colorants represent novel risk factors for development of colitis in mice with increased IL-23 signaling.


Asunto(s)
Bacterias/metabolismo , Colitis , Colorantes de Alimentos/metabolismo , Interleucina-23/genética , Mucosa Intestinal/microbiología , Animales , Colitis/genética , Colitis/metabolismo , Colitis/microbiología , Modelos Animales de Enfermedad , Colorantes de Alimentos/efectos adversos , Predisposición Genética a la Enfermedad , Proteínas de Homeodominio/genética , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/microbiología , Interferón gamma/genética , Interleucina-23/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Simbiosis
5.
Theranostics ; 11(4): 1594-1608, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33408769

RESUMEN

The Notch pathway is highly active in almost all patients with T-cell acute lymphoblastic leukemia (T-ALL), but the implication of Notch ligands in T-ALL remains underexplored. Methods: We used a genetic mouse model of Notch ligand delta like 4 (DLL4)-driven T-ALL and performed thymectomies and splenectomies in those animals. We also used several patient-derived T-ALL (PDTALL) models, including one with DLL4 expression on the membrane and we treated PDTALL cells in vitro and in vivo with demcizumab, a blocking antibody against human DLL4 currently being tested in clinical trials in patients with solid cancer. Results: We show that surgical removal of the spleen abrogated T-ALL development in our preclinical DLL4-driven T-ALL mouse model. Mechanistically, we found that the spleen, and not the thymus, promoted the accumulation of circulating CD4+CD8+ T cells before T-ALL onset, suggesting that DLL4-driven T-ALL derives from these cells. Then, we identified a small subset of T-ALL patients showing higher levels of DLL4 expression. Moreover, in mice xenografted with a DLL4-positive PDTALL model, treatment with demcizumab had the same therapeutic effect as global Notch pathway inhibition using the potent γ-secretase inhibitor dibenzazepine. This result demonstrates that, in this PDTALL model, Notch pathway activity depends on DLL4 signaling, thus validating our preclinical mouse model. Conclusion: DLL4 expression in human leukemic cells can be a source of Notch activity in T-ALL, and the spleen plays a major role in a genetic mouse model of DLL4-driven T-ALL.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Biomarcadores de Tumor/metabolismo , Proteínas de Unión al Calcio/metabolismo , Regulación Neoplásica de la Expresión Génica , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , Receptores Notch/metabolismo , Bazo/patología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Proliferación Celular , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Leucemia-Linfoma Linfoblástico de Células T Precursoras/etiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Receptores Notch/genética , Bazo/metabolismo , Bazo/cirugía , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Sci Rep ; 10(1): 8259, 2020 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-32427877

RESUMEN

Psoriasis (PS) is a chronic skin inflammation. Up to 30% of the patients with PS develop psoriatic arthritis (PsA), a condition characterized by inflammatory arthritis that affects joints or entheses. Although there is mounting evidence for a critical role of interleukin-23 (IL-23) signaling in the pathogenesis of both PS and PsA, it remains unclear whether IL-23-induced skin inflammation drives joint disease. Here, we show that mice expressing increased levels of IL-23 in the skin (K23 mice) develop a PS-like disease that is characterized by acanthosis, parakeratosis, hyperkeratosis, and inflammatory infiltrates in the dermis. Skin disease preceded development of PsA, including enthesitis, dactylitis, and bone destruction. The development of enthesitis and dactylitis was not due to high circulating levels of IL-23, as transgenic animals and controls had similar levels of this cytokine in circulation. IL-22, a downstream cytokine of IL-23, was highly increased in the serum of K23 mice. Although IL-22 deficiency did not affect skin disease development, IL-22 deficiency aggravated the PsA-like disease in K23 mice. Our results demonstrate a central role for skin expressed IL-23 in the initiation of PS and on pathogenic processes leading to PsA.


Asunto(s)
Artritis Psoriásica/genética , Interleucina-23/genética , Psoriasis/genética , Piel/inmunología , Animales , Artritis Psoriásica/inmunología , Femenino , Humanos , Interleucina-23/inmunología , Interleucinas/genética , Interleucinas/inmunología , Masculino , Ratones , Psoriasis/inmunología , Interleucina-22
7.
Nat Commun ; 11(1): 291, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31941899

RESUMEN

Clonal evolution of a tumor ecosystem depends on different selection pressures that are principally immune and treatment mediated. We integrate RNA-seq, DNA sequencing, TCR-seq and SNP array data across multiple regions of liver cancer specimens to map spatio-temporal interactions between cancer and immune cells. We investigate how these interactions reflect intra-tumor heterogeneity (ITH) by correlating regional neo-epitope and viral antigen burden with the regional adaptive immune response. Regional expression of passenger mutations dominantly recruits adaptive responses as opposed to hepatitis B virus and cancer-testis antigens. We detect different clonal expansion of the adaptive immune system in distant regions of the same tumor. An ITH-based gene signature improves single-biopsy patient survival predictions and an expression survey of 38,553 single cells across 7 regions of 2 patients further reveals heterogeneity in liver cancer. These data quantify transcriptomic ITH and how the different components of the HCC ecosystem interact during cancer evolution.


Asunto(s)
Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Evolución Clonal , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/virología , Variaciones en el Número de Copia de ADN , Epítopos/genética , Epítopos/inmunología , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Heterogeneidad Genética , Antígenos de la Hepatitis B/genética , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/inmunología , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Estimación de Kaplan-Meier , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/virología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Linfocitos Infiltrantes de Tumor/virología , Polimorfismo de Nucleótido Simple , Análisis de la Célula Individual
8.
Nat Commun ; 10(1): 4517, 2019 10 04.
Artículo en Inglés | MEDLINE | ID: mdl-31586069

RESUMEN

Neonatal inflammatory diseases are associated with severe morbidity, but the inflammatory factors underlying them and their potential effector mechanisms are poorly defined. Here we show that necrotizing enterocolitis in neonate mice is accompanied by elevation of IL-23 and IL-22 and decreased production of pancreatic enzymes. These phenotypes are mirrored in neonate mice overexpressing IL-23 in CX3CR1+ myeloid cells or in keratinocytes. The mice fail to grow and die prematurely, displaying systemic inflammation, nutrient malabsorption and decreased expression of intestinal and pancreatic genes mediating digestion and absorption of carbohydrates, proteins, and lipids. Germ-free environment improves, and genetic ablation of IL-22 restores normal growth in mice overexpressing IL-23. Mechanistically, IL-22 acts directly at the level of pancreatic acinar cells to decrease expression of the pancreas associated transcription factor 1a (PTF1a). These results show that augmented production of IL-23 and IL-22 in early life has a negative impact on pancreatic enzyme secretion and food absorption.


Asunto(s)
Enterocolitis Necrotizante/inmunología , Interleucina-23/metabolismo , Interleucinas/metabolismo , Páncreas/enzimología , Factores de Transcripción/metabolismo , Células Acinares/enzimología , Animales , Animales Recién Nacidos , Células Cultivadas , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/patología , Humanos , Interleucina-23/genética , Interleucina-23/inmunología , Interleucinas/genética , Interleucinas/inmunología , Absorción Intestinal/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Queratinocitos , Ratones , Ratones Noqueados , Células Mieloides , Páncreas/citología , Cultivo Primario de Células , Interleucina-22
9.
Gastroenterology ; 157(6): 1572-1583.e8, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31470007

RESUMEN

BACKGROUND & AIMS: Transgenic mice (HBUS) that express the epidermal growth factor receptor (EGFR) ligand HBEGF (heparin-binding epidermal growth factor-like growth factor) and a constitutively active G protein-coupled receptor (US28) in intestinal epithelial cells develop serrated polyps in the cecum. Development of serrated polyps depends on the composition of the gut microbiota and is associated with bacterial invasion of the lamina propria, accompanied by induction of inflammation and up-regulation of interleukin 1 beta (IL1B) and matrix metalloproteinase (MMP) 3 in the cecum. We investigated the mechanisms by which these changes contribute to development of serrated polyps. METHODS: We performed studies with C57BL/6 (control) and HBUS mice. To accelerate polyp development, we increased the exposure of the bacteria to the lamina propria by injecting HBUS mice with diphtheria toxin, which binds transgenic HBEGF expressed by the epithelial cells and causes apoptosis. Mice were given injections of IL1B-neutralizing antibody and the MMP inhibitor N-isobutyl-N-(4-methoxyphenylsulfonyl)glycyl hydroxamic acid. Intestinal tissues were collected from mice and analyzed by histology, reverse-transcription polymerase chain reaction, enzyme-linked immunosorbent assay, immunofluorescence, and flow cytometry. We examined fibroblast subsets in polyps using single-cell RNA sequencing. RESULTS: Administration of diphtheria toxin to HBUS mice accelerated development of serrated polyps (95% of treated mice developed polyps before 100 days of age, compared with 53% given vehicle). IL1B stimulated subsets of platelet-derived growth factor receptor alpha+ (PDGRFA+) fibroblasts isolated from cecum, resulting in increased expression of MMP3. Neutralizing antibodies against IL1B or administration of the MMP inhibitor reduced the number of serrated polyps that formed in the HBUS mice. Single-cell RNA sequencing analysis showed subsets of fibroblasts in serrated polyps that express genes that regulate matrix fibroblasts and inflammation. CONCLUSIONS: In studies of mice, we found that barrier breakdown and expression of inflammatory factors contribute to development of serrated polyps. Subsets of cecal PDGFRA+ fibroblasts are activated by release of IL1B from myeloid cells during the early stages of serrated polyp development. MMP3 produced by PDGFRA+ fibroblasts is important for serrated polyp development. Our findings confirm the functions of previously identified serrated polyp-associated molecules and indicate roles for immune and stromal cells in serrated polyp development.


Asunto(s)
Pólipos del Colon/inmunología , Receptores ErbB/metabolismo , Interleucina-1beta/metabolismo , Mucosa Intestinal/patología , Metaloproteinasa 3 de la Matriz/metabolismo , Animales , Apoptosis/inmunología , Ciego/citología , Ciego/inmunología , Ciego/patología , Toxina Diftérica/administración & dosificación , Toxina Diftérica/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/patología , Receptores ErbB/antagonistas & inhibidores , Fibroblastos/inmunología , Fibroblastos/metabolismo , Gefitinib/farmacología , Factor de Crecimiento Similar a EGF de Unión a Heparina/genética , Factor de Crecimiento Similar a EGF de Unión a Heparina/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Interleucina-1beta/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Metaloproteinasa 3 de la Matriz/inmunología , Inhibidores de la Metaloproteinasa de la Matriz/farmacología , Ratones , Ratones Transgénicos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Sulfonamidas/farmacología
10.
Sci Transl Med ; 10(461)2018 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-30282696

RESUMEN

Gut homing CD4+ T cells expressing the integrin α4ß7 are early viral targets and contribute to HIV-1 pathogenesis, likely by seeding the gastrointestinal (GI) tract with HIV. Although simianized anti-α4ß7 monoclonal antibodies have shown promise in preventing or attenuating the disease course of simian immunodeficiency virus in nonhuman primate studies, the mechanisms of drug action remain elusive. We present a cohort of individuals with mild inflammatory bowel disease and concomitant HIV-1 infection receiving anti-α4ß7 treatment. By sampling the immune inductive and effector sites of the GI tract, we have discovered that anti-α4ß7 therapy led to a significant and unexpected attenuation of lymphoid aggregates, most notably in the terminal ileum. Given that lymphoid aggregates serve as important sanctuary sites for maintaining viral reservoirs, their attrition by anti-α4ß7 therapy has important implications for HIV-1 therapeutics and eradication efforts and defines a rational basis for the use of anti-α4ß7 therapy in HIV-1 infection.


Asunto(s)
Tracto Gastrointestinal/patología , Tracto Gastrointestinal/virología , Infecciones por VIH/terapia , Integrinas/antagonistas & inhibidores , Tejido Linfoide/patología , Adulto , Anticuerpos Monoclonales Humanizados/efectos adversos , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Linfocitos B/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Femenino , Infecciones por VIH/sangre , Infecciones por VIH/inmunología , Humanos , Integrinas/metabolismo , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Subgrupos Linfocitarios/efectos de los fármacos , Subgrupos Linfocitarios/inmunología , Masculino , Persona de Mediana Edad , Empalme del ARN/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
Cytokine ; 111: 146-153, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30145369

RESUMEN

Interleukin 33 (IL33) is a cytokine found in the extracellular space (mature IL33) or in the cell nucleus (full-length IL33). Nuclear accumulation of IL33 has been reported in intestinal epithelial cells (IEC) during intestinal inflammation and cancer, but a functional role for this nuclear form remains unclear. To study the role of nuclear IL33 in IEC, we generated transgenic mice expressing full-length IL33 in the intestinal epithelium (Vfl33 mice). Expression of full-length IL33 in the epithelium resulted in accumulation of IL33 protein in the nucleus and secretion of IL33. Over-expression of full-length IL33 by IEC did not promote gut inflammation, but induced expression of genes in the IEC and lamina propria lymphocytes (LPL) that correlated negatively with genes expressed in inflammatory bowel diseases (IBD). Because the IL33 receptor ST2 is expressed by IEC, there was the potential that both the mature and full-length forms could mediate this effect. To specifically interrogate the transcriptional role of nuclear IL33, we intercrossed the Vfl33 mice with ST2- deficient mice. ST2 deficiency completely abrogated the transcriptional effects elicited by IL33 expression, suggesting that the transcriptional effects of IL33 on IEC are mediated by its mature, not its nuclear form.


Asunto(s)
Núcleo Celular/inmunología , Enterocitos/inmunología , Regulación de la Expresión Génica/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Interleucina-33/inmunología , Animales , Núcleo Celular/genética , Núcleo Celular/patología , Enterocitos/patología , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Proteína 1 Similar al Receptor de Interleucina-1/genética , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-33/genética , Linfocitos/inmunología , Linfocitos/patología , Ratones , Ratones Noqueados
12.
Gastroenterology ; 155(4): 1177-1191.e16, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29909020

RESUMEN

BACKGROUND & AIMS: Several studies have shown that signaling via the interleukin 23 (IL23) receptor is required for development of colitis. We studied the roles of IL23, dietary factors, alterations to the microbiota, and T cells in the development and progression of colitis in mice. METHODS: All mice were maintained on laboratory diet 5053, unless otherwise noted. We generated mice that express IL23 in CX3CR1-positive myeloid cells (R23FR mice) upon cyclic administration of tamoxifen dissolved in diet 2019. Diets 2019 and 5053 have minor differences in the overall composition of protein, fat, fiber, minerals, and vitamins. CX3CR1CreER mice (FR mice) were used as controls. Some mice were given antibiotics, and others were raised in a germ-free environment. Intestinal tissues were collected and analyzed by histology and flow cytometry. Feces were collected and analyzed by 16S rDNA sequencing. Feces from C57/Bl6, R23FR, or FR mice were fed to FR and R23FR germ-free mice in microbiota transplant experiments. We also performed studies with R23FR/Rag-/-, R23FR/Mu-/-, and R23FR/Tcrd-/- mice. R23FR mice were given injections of antibodies against CD4 or CD8 to deplete T cells. Mesenteric lymph nodes and large intestine CD4+ cells from R23FR or FR mice in remission from colitis were transferred into Rag-/- mice. CD4+ cells were isolated from donor R23FR mice and recipient Rag-/- mice, and T-cell receptor sequences were determined. RESULTS: Expression of IL23 led to development of a relapsing-remitting colitis that was dependent on the microbiota and CD4+ T cells. The relapses were caused by switching from the conventional diet used in our facility (diet 5053) to the diet 2019 and were not dependent on tamoxifen after the first cycle. The switch in the diet modified the microbiota but did not alter levels of IL23 in intestinal tissues compared with mice that remained on the conventional diet. Mesenteric lymph nodes and large intestine CD4+ cells from R23FR mice in remission, but not from FR mice, induced colitis after transfer into Rag-/- mice, but only when these mice were placed on the diet 2019. The CD4+ T-cell receptor repertoire of Rag-/- mice with colitis (fed the 2019 diet) was less diverse than that from donor mice and Rag-/- mice without colitis (fed the 5053 diet) because of expansion of dominant T-cell clones. CONCLUSIONS: We developed mice that express IL23 in CX3CR1-positive myeloid cells (R23FR mice) and found that they are more susceptible to diet-induced colitis than mice that do not express IL23. The R23FR mice have a population of CD4+ T cells that becomes activated in response to dietary changes and alterations to the intestinal microbiota. The results indicate that alterations in the diet, intestinal microbiota, and IL23 signaling can contribute to pathogenesis of inflammatory bowel disease.


Asunto(s)
Alimentación Animal , Linfocitos T CD4-Positivos/metabolismo , Colitis/dietoterapia , Colon/metabolismo , Microbioma Gastrointestinal , Interleucina-23/metabolismo , Células Mieloides/metabolismo , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/trasplante , Receptor 1 de Quimiocinas CX3C/metabolismo , Colitis/inmunología , Colitis/metabolismo , Colitis/microbiología , Colon/inmunología , Colon/microbiología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Heces/microbiología , Interacción Gen-Ambiente , Interacciones Huésped-Patógeno , Interleucina-23/genética , Ratones Endogámicos C57BL , Ratones Transgénicos , Valor Nutritivo , Transducción de Señal , Factores de Tiempo
13.
Nature ; 557(7706): 580-584, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29769727

RESUMEN

Somatic mutations in tet methylcytosine dioxygenase 2 (TET2), which encodes an epigenetic modifier enzyme, drive the development of haematopoietic malignancies1-7. In both humans and mice, TET2 deficiency leads to increased self-renewal of haematopoietic stem cells with a net developmental bias towards the myeloid lineage1,4,8,9. However, pre-leukaemic myeloproliferation (PMP) occurs in only a fraction of Tet2-/- mice8,9 and humans with TET2 mutations1,3,5-7, suggesting that extrinsic non-cell-autonomous factors are required for disease onset. Here we show that bacterial translocation and increased interleukin-6 production, resulting from dysfunction of the small-intestinal barrier, are critical for the development of PMP in mice that lack Tet2 expression in haematopoietic cells. Furthermore, in symptom-free Tet2-/- mice, PMP can be induced by disrupting intestinal barrier integrity, or in response to systemic bacterial stimuli such as the toll-like receptor 2 agonist. PMP was reversed by antibiotic treatment and failed to develop in germ-free Tet2-/- mice, which illustrates the importance of microbial signals in the development of this condition. Our findings demonstrate the requirement for microbial-dependent inflammation in the development of PMP and provide a mechanistic basis for the variation in PMP penetrance observed in Tet2-/- mice. This study will prompt new lines of investigation that may profoundly affect the prevention and management of haematopoietic malignancies.


Asunto(s)
Enfermedades Asintomáticas , Fenómenos Fisiológicos Bacterianos , Proliferación Celular , Proteínas de Unión al ADN/deficiencia , Leucemia/microbiología , Leucemia/patología , Proteínas Proto-Oncogénicas/deficiencia , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Fenómenos Fisiológicos Bacterianos/inmunología , Proteínas de Unión al ADN/genética , Dioxigenasas , Femenino , Vida Libre de Gérmenes , Inflamación/microbiología , Interleucina-6/inmunología , Mucosa Intestinal/metabolismo , Lactobacillus/química , Lactobacillus/citología , Lactobacillus/inmunología , Masculino , Ratones , Penetrancia , Permeabilidad , Proteínas Proto-Oncogénicas/genética , Receptor Toll-Like 2/agonistas
14.
Sci Rep ; 7(1): 5520, 2017 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-28710436

RESUMEN

Increased expression of Interleukin (IL)-33 has been detected in intestinal samples of patients with ulcerative colitis, a condition associated with increased risk for colon cancer, but its role in the development of colorectal cancer has yet to be fully examined. Here, we investigated the role of epithelial expressed IL-33 during development of intestinal tumors. IL-33 expression was detected in epithelial cells in colorectal cancer specimens and in the Apc Min/+ mice. To better understand the role of epithelial-derived IL-33 in the intestinal tumorigenesis, we generated transgenic mice expressing IL-33 in intestinal epithelial cells (V33 mice). V33 Apc Min/+ mice, resulting from the cross of V33 with Apc Min/+ mice, had increased intestinal tumor burden compared with littermate Apc Min/+ mice. Consistently, Apc Min/+ mice deficient for IL-33 receptor (ST2), had reduced polyp burden. Mechanistically, overexpression of IL-33 promoted expansion of ST2+ regulatory T cells, increased Th2 cytokine milieu, and induced alternatively activated macrophages in the gut. IL-33 promoted marked changes in the expression of antimicrobial peptides, and antibiotic treatment of V33 Apc Min/+ mice abrogated the tumor promoting-effects of IL-33 in the colon. In conclusion, elevated IL-33 signaling increases tumor development in the Apc Min/+ mice.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Transformación Celular Neoplásica/inmunología , Células Epiteliales/metabolismo , Interleucina-33/metabolismo , Neoplasias Intestinales/inmunología , Animales , Transformación Celular Neoplásica/genética , Células Cultivadas , Citocinas/metabolismo , Células Epiteliales/citología , Regulación Neoplásica de la Expresión Génica , Humanos , Proteína 1 Similar al Receptor de Interleucina-1/metabolismo , Interleucina-33/genética , Neoplasias Intestinales/genética , Activación de Macrófagos , Macrófagos/inmunología , Ratones , Ratones Transgénicos , Transducción de Señal , Linfocitos T Reguladores/inmunología , Células Th2/inmunología , Regulación hacia Arriba
15.
Nature ; 539(7630): 565-569, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27828940

RESUMEN

Recognition and removal of apoptotic cells by professional phagocytes, including dendritic cells and macrophages, preserves immune self-tolerance and prevents chronic inflammation and autoimmune pathologies. The diverse array of phagocytes that reside within different tissues, combined with the necessarily prompt nature of apoptotic cell clearance, makes it difficult to study this process in situ. The full spectrum of functions executed by tissue-resident phagocytes in response to homeostatic apoptosis, therefore, remains unclear. Here we show that mouse apoptotic intestinal epithelial cells (IECs), which undergo continuous renewal to maintain optimal barrier and absorptive functions, are not merely extruded to maintain homeostatic cell numbers, but are also sampled by a single subset of dendritic cells and two macrophage subsets within a well-characterized network of phagocytes in the small intestinal lamina propria. Characterization of the transcriptome within each subset before and after in situ sampling of apoptotic IECs revealed gene expression signatures unique to each phagocyte, including macrophage-specific lipid metabolism and amino acid catabolism, and a dendritic-cell-specific program of regulatory CD4+ T-cell activation. A common 'suppression of inflammation' signature was noted, although the specific genes and pathways involved varied amongst dendritic cells and macrophages, reflecting specialized functions. Apoptotic IECs were trafficked to mesenteric lymph nodes exclusively by the dendritic cell subset and served as critical determinants for the induction of tolerogenic regulatory CD4+ T-cell differentiation. Several of the genes that were differentially expressed by phagocytes bearing apoptotic IECs overlapped with susceptibility genes for inflammatory bowel disease. Collectively, these findings provide new insights into the consequences of apoptotic cell sampling, advance our understanding of how homeostasis is maintained within the mucosa and set the stage for development of novel therapeutics to alleviate chronic inflammatory diseases such as inflammatory bowel disease.


Asunto(s)
Apoptosis , Células Epiteliales/citología , Células Epiteliales/inmunología , Homeostasis , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Fagocitos/citología , Fagocitos/inmunología , Aminoácidos/metabolismo , Animales , Antígenos CD/metabolismo , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Diferenciación Celular , Movimiento Celular , Células Dendríticas/citología , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Femenino , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología , Cadenas alfa de Integrinas/metabolismo , Metabolismo de los Lípidos , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Macrófagos/citología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Fagocitos/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Transcripción Genética
16.
J Leukoc Biol ; 100(5): 1125-1134, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27286792

RESUMEN

During intraocular bacterial infections, the primary innate responders are neutrophils, which may cause bystander damage to the retina or perturb the clarity of the visual axis. We hypothesized that cytokine IL-6 and chemokine CXCL1 contributed to rapid neutrophil recruitment during Bacillus cereus endophthalmitis, a severe form of intraocular infection that is characterized by explosive inflammation and retinal damage that often leads to rapid vision loss. To test this hypothesis, we compared endophthalmitis pathogenesis in C57BL/6J, IL-6-/-, and CXCL1-/- mice. Bacterial growth in eyes of CXCL1-/-, IL-6-/-, and C67BL/6J mice was similar. Retinal function retention was greater in eyes of IL-6-/- and CXCL1-/- mice compared with that of C57BL/6J, despite these eyes having similar bacterial burdens. Neutrophil influx into eyes of CXCL1-/- mice was reduced to a greater degree compared with that of eyes of IL6-/- mice. Histology confirmed significantly less inflammation in eyes of CXCL1-/- mice, but similar degrees of inflammation in IL6-/- and C57BL/6J eyes. Because inflammation was reduced in eyes of infected CXCL1-/- mice, we tested the efficacy of anti-CXCL1 in B. cereus endophthalmitis. Retinal function was retained to a greater degree and there was less overall inflammation in eyes treated with anti-CXCL1, which suggested that anti-CXCL1 may have therapeutic efficacy in limiting inflammation during B. cereus endophthalmitis. Taken together, our results indicate that absence of IL-6 did not affect overall pathogenesis of endophthalmitis. In contrast, absence of CXCL1, in CXCL1-/- mice or after anti-CXCL1 treatment, led to an improved clinical outcome. Our findings suggest a potential benefit in targeting CXCL1 to control inflammation during B. cereus and perhaps other types of intraocular infections.


Asunto(s)
Bacillus cereus , Quimiocina CXCL1/fisiología , Quimiotaxis de Leucocito/fisiología , Endoftalmitis/inmunología , Infecciones Bacterianas del Ojo/inmunología , Interleucina-6/fisiología , Neutrófilos/inmunología , Animales , Anticuerpos Monoclonales/farmacología , Bacillus cereus/crecimiento & desarrollo , Bacillus cereus/aislamiento & purificación , Carga Bacteriana , Quimiocina CXCL1/antagonistas & inhibidores , Quimiocina CXCL1/deficiencia , Quimiocina CXCL1/genética , Electrorretinografía , Endoftalmitis/microbiología , Infecciones Bacterianas del Ojo/microbiología , Mediadores de Inflamación/análisis , Interleucina-6/deficiencia , Interleucina-6/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Peroxidasa/análisis , Retina/patología
17.
Mucosal Immunol ; 8(2): 390-402, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25160819

RESUMEN

Interleukin-23 (IL-23) responsive group 3 innate lymphoid cells (ILC3s) have been implicated in immune homeostasis and pathogenesis in the adult, but little is known about their roles in the newborn. Here we show that IL-23 promotes conversion of embryonic intestinal Lin(-)IL-23R(+)Thy1(+) cells into IL-22-producing Thy1(+)Sca-1(hi) ILC3s in vitro. Gut-specific expression of IL-23 also activated and expanded Thy1(+)Sca-1(hi) ILC3s, which produced IL-22, IL-17, interferon gamma (IFN-γ), and granulocyte-macrophage colony-stimulating factor (GM-CSF) and were distinct from canonical CD4(+) lymphoid tissue inducer (LTi) cells. These ILC3s accumulated under the epithelium in intercellular adhesion molecule (ICAM)-1-positive cell aggregates together with neutrophils that disrupted the epithelium, leading to the formation of discrete intestinal erosions, bleeding, and neonatal death. Genetic and antibody depletion of ILC3s rescued the mice from neonatal death. Antibiotic treatment of pregnant mothers and offspring prolonged survival of IL-23 transgenic mice, suggesting a role for the commensal flora on ILC3-induced pathogenesis. Our results reveal a novel role for the IL-23-ILC3s axis in the pathogenesis of neonatal intestinal inflammation.


Asunto(s)
Inmunidad Innata , Interleucina-23/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Animales , Antígenos de Superficie/metabolismo , Citocinas/metabolismo , Expresión Génica , Humanos , Inmunidad Innata/genética , Inmunofenotipificación , Interleucina-23/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Intestinos/microbiología , Intestinos/patología , Ratones , Ratones Transgénicos , Infiltración Neutrófila , Neutrófilos/inmunología , Neutrófilos/metabolismo , Permeabilidad , Fenotipo
18.
J Exp Med ; 211(3): 457-72, 2014 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-24590763

RESUMEN

The preferential localization of some neoplasms, such as serrated polyps (SPs), in specific areas of the intestine suggests that nongenetic factors may be important for their development. To test this hypothesis, we took advantage of transgenic mice that expressed HB-EGF throughout the intestine but developed SPs only in the cecum. Here we show that a host-specific microbiome was associated with SPs and that alterations of the microbiota induced by antibiotic treatment or by embryo transfer rederivation markedly inhibited the formation of SPs in the cecum. Mechanistically, development of SPs was associated with a local decrease in epithelial barrier function, bacterial invasion, production of antimicrobials, and increased expression of several inflammatory factors such as IL-17, Cxcl2, Tnf-α, and IL-1. Increased numbers of neutrophils were found within the SPs, and their depletion significantly reduced polyp growth. Together these results indicate that nongenetic factors contribute to the development of SPs and suggest that the development of these intestinal neoplasms in the cecum is driven by the interplay between genetic changes in the host, an inflammatory response, and a host-specific microbiota.


Asunto(s)
Ciego/patología , Epitelio/fisiología , Regulación Neoplásica de la Expresión Génica/inmunología , Pólipos Intestinales/patología , Microbiota/fisiología , Modelos Moleculares , Animales , Antibacterianos/farmacología , Citocinas/metabolismo , Factor de Crecimiento Similar a EGF de Unión a Heparina , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Pólipos Intestinales/metabolismo , Ratones , Ratones Transgénicos , Conformación Proteica , Especificidad de la Especie
19.
Immunity ; 38(3): 581-95, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-23395676

RESUMEN

CD103+ dendritic cells (DCs) carry bacteria from the small intestine and can present antigens to T cells. Yet they have not been recorded sampling luminal bacteria or presenting bacterial antigens in mesentery lymph nodes. We used 2-photon microscopy in live Cx3cr1(+/gfp) ×Cd11c-YFP mice to study these processes. At steady state, sparse CD103+ DCs occupied the epithelium. They patrolled among enterocytes while extending dendrites toward the lumen, likely using tight-junction proteins to penetrate the epithelium. Challenge with Salmonella triggered chemokine- and toll-like receptor (TLR)-dependent recruitment of additional DCs from the lamina propria (LP). The DCs efficiently phagocytosed the bacteria using intraepithelial dendrites. Noninvasive bacteria were similarly sampled. In contrast, CD103+ DCs sampled soluble luminal antigen inefficiently. In mice harboring CD103+ DCs, antigen-specific CD8 T cells were subsequently activated in MLNs. Intestinal CD103+ DCs are therefore equipped with unique mechanisms to independently complete the processes of uptake, transportation, and presentation of bacterial antigens.


Asunto(s)
Presentación de Antígeno/inmunología , Antígenos Bacterianos/inmunología , Antígenos CD/inmunología , Células Dendríticas/inmunología , Cadenas alfa de Integrinas/inmunología , Mucosa Intestinal/inmunología , Animales , Antígenos CD/metabolismo , Antígeno CD11c/genética , Antígeno CD11c/inmunología , Antígeno CD11c/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Receptor 1 de Quimiocinas CX3C , Línea Celular Tumoral , Movimiento Celular/inmunología , Células Cultivadas , Células Dendríticas/metabolismo , Citometría de Flujo , Interacciones Huésped-Patógeno/inmunología , Cadenas alfa de Integrinas/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía de Fluorescencia por Excitación Multifotónica , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Membrana Mucosa/microbiología , Receptores de Quimiocina/genética , Receptores de Quimiocina/inmunología , Receptores de Quimiocina/metabolismo , Salmonella typhi/inmunología , Salmonella typhi/fisiología , Salmonella typhimurium/inmunología , Salmonella typhimurium/fisiología , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo
20.
Nat Immunol ; 13(8): 744-52, 2012 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-22729248

RESUMEN

Langerhans cells (LCs) are epidermal dendritic cells with incompletely understood origins that associate with hair follicles for unknown reasons. Here we show that in response to external stress, mouse hair follicles recruited Gr-1(hi) monocyte-derived precursors of LCs whose epidermal entry was dependent on the chemokine receptors CCR2 and CCR6, whereas the chemokine receptor CCR8 inhibited the recruitment of LCs. Distinct hair-follicle regions had differences in their expression of ligands for CCR2 and CCR6. The isthmus expressed the chemokine CCL2; the infundibulum expressed the chemokine CCL20; and keratinocytes in the bulge produced the chemokine CCL8, which is the ligand for CCR8. Thus, distinct hair-follicle keratinocyte subpopulations promoted or inhibited repopulation with LCs via differences in chemokine production, a feature also noted in humans. Pre-LCs failed to enter hairless skin in mice or humans, which establishes hair follicles as portals for LCs.


Asunto(s)
Quimiocinas/biosíntesis , Folículo Piloso/inmunología , Células de Langerhans/fisiología , Estrés Fisiológico , Alopecia , Animales , Movimiento Celular , Quimiocina CCL20/biosíntesis , Quimiocina CCL8/biosíntesis , Quimiocinas/metabolismo , Folículo Piloso/metabolismo , Humanos , Queratinocitos/metabolismo , Células de Langerhans/inmunología , Ratones , Ratones Pelados , Receptores CCR2/metabolismo , Receptores CCR6/metabolismo , Receptores CCR8/metabolismo , Piel/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA