Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 126
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 184(19): 5015-5030.e16, 2021 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-34407392

RESUMEN

Group 3 innate lymphoid cells (ILC3s) regulate immunity and inflammation, yet their role in cancer remains elusive. Here, we identify that colorectal cancer (CRC) manifests with altered ILC3s that are characterized by reduced frequencies, increased plasticity, and an imbalance with T cells. We evaluated the consequences of these changes in mice and determined that a dialog between ILC3s and T cells via major histocompatibility complex class II (MHCII) is necessary to support colonization with microbiota that subsequently induce type-1 immunity in the intestine and tumor microenvironment. As a result, mice lacking ILC3-specific MHCII develop invasive CRC and resistance to anti-PD-1 immunotherapy. Finally, humans with dysregulated intestinal ILC3s harbor microbiota that fail to induce type-1 immunity and immunotherapy responsiveness when transferred to mice. Collectively, these data define a protective role for ILC3s in cancer and indicate that their inherent disruption in CRC drives dysfunctional adaptive immunity, tumor progression, and immunotherapy resistance.


Asunto(s)
Neoplasias del Colon/inmunología , Neoplasias del Colon/terapia , Progresión de la Enfermedad , Inmunidad Innata , Inmunoterapia , Linfocitos/inmunología , Animales , Comunicación Celular/efectos de los fármacos , Plasticidad de la Célula/efectos de los fármacos , Neoplasias del Colon/microbiología , Heces/microbiología , Antígenos de Histocompatibilidad Clase II/metabolismo , Humanos , Inhibidores de Puntos de Control Inmunológico/farmacología , Inmunidad Innata/efectos de los fármacos , Inflamación/inmunología , Inflamación/patología , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/microbiología , Enfermedades Inflamatorias del Intestino/patología , Intestinos/patología , Linfocitos/efectos de los fármacos , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , Invasividad Neoplásica , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología , Donantes de Tejidos
2.
Nat Immunol ; 23(2): 251-261, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35102343

RESUMEN

Tumor necrosis factor (TNF) drives chronic inflammation and cell death in the intestine, and blocking TNF is a therapeutic approach in inflammatory bowel disease (IBD). Despite this knowledge, the pathways that protect the intestine from TNF are incompletely understood. Here we demonstrate that group 3 innate lymphoid cells (ILC3s) protect the intestinal epithelium from TNF-induced cell death. This occurs independent of interleukin-22 (IL-22), and we identify that ILC3s are a dominant source of heparin-binding epidermal growth factor-like growth factor (HB-EGF). ILC3s produce HB-EGF in response to prostaglandin E2 (PGE2) and engagement of the EP2 receptor. Mice lacking ILC3-derived HB-EGF exhibit increased susceptibility to TNF-mediated epithelial cell death and experimental intestinal inflammation. Finally, human ILC3s produce HB-EGF and are reduced from the inflamed intestine. These results define an essential role for ILC3-derived HB-EGF in protecting the intestine from TNF and indicate that disruption of this pathway contributes to IBD.


Asunto(s)
Factor de Crecimiento Similar a EGF de Unión a Heparina/inmunología , Inmunidad Innata/inmunología , Inflamación/inmunología , Intestinos/inmunología , Linfocitos/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Células Epiteliales/inmunología , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos C57BL , Transducción de Señal/inmunología
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 ; 20(3): 257-264, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30778250

RESUMEN

Post-translational modification of chemokines mediated by the dipeptidyl peptidase DPP4 (CD26) has been shown to negatively regulate lymphocyte trafficking, and its inhibition enhances T cell migration and tumor immunity by preserving functional chemokine CXCL10. By extending those initial findings to pre-clinical models of hepatocellular carcinoma and breast cancer, we discovered a distinct mechanism by which inhibition of DPP4 improves anti-tumor responses. Administration of the DPP4 inhibitor sitagliptin resulted in higher concentrations of the chemokine CCL11 and increased migration of eosinophils into solid tumors. Enhanced tumor control was preserved in mice lacking lymphocytes and was ablated after depletion of eosinophils or treatment with degranulation inhibitors. We further demonstrated that tumor-cell expression of the alarmin IL-33 was necessary and sufficient for eosinophil-mediated anti-tumor responses and that this mechanism contributed to the efficacy of checkpoint-inhibitor therapy. These findings provide insight into IL-33- and eosinophil-mediated tumor control, revealed when endogenous mechanisms of DPP4 immunoregulation are inhibited.


Asunto(s)
Dipeptidil Peptidasa 4/inmunología , Eosinófilos/inmunología , Interleucina-33/inmunología , Neoplasias Experimentales/inmunología , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Movimiento Celular/inmunología , Quimiocina CCL11/inmunología , Quimiocina CCL11/metabolismo , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Modelos Animales de Enfermedad , Eosinófilos/efectos de los fármacos , Eosinófilos/metabolismo , Humanos , Interleucina-33/metabolismo , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales/metabolismo , Neoplasias Experimentales/prevención & control , Fosfato de Sitagliptina/farmacología
5.
Cell ; 166(5): 1231-1246.e13, 2016 Aug 25.
Artículo en Inglés | MEDLINE | ID: mdl-27545347

RESUMEN

Innate lymphoid cells (ILCs) are critical modulators of mucosal immunity, inflammation, and tissue homeostasis, but their full spectrum of cellular states and regulatory landscapes remains elusive. Here, we combine genome-wide RNA-seq, ChIP-seq, and ATAC-seq to compare the transcriptional and epigenetic identity of small intestinal ILCs, identifying thousands of distinct gene profiles and regulatory elements. Single-cell RNA-seq and flow and mass cytometry analyses reveal compartmentalization of cytokine expression and metabolic activity within the three classical ILC subtypes and highlight transcriptional states beyond the current canonical classification. In addition, using antibiotic intervention and germ-free mice, we characterize the effect of the microbiome on the ILC regulatory landscape and determine the response of ILCs to microbial colonization at the single-cell level. Together, our work characterizes the spectrum of transcriptional identities of small intestinal ILCs and describes how ILCs differentially integrate signals from the microbial microenvironment to generate phenotypic and functional plasticity.


Asunto(s)
Microbioma Gastrointestinal , Inmunidad Innata/genética , Intestinos/inmunología , Intestinos/microbiología , Linfocitos/inmunología , Linfocitos/microbiología , Animales , Secuencia de Bases , Cromatina/metabolismo , Citocinas/inmunología , Epigénesis Genética , Regulación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Análisis de la Célula Individual , Transcripción Genética
6.
Immunity ; 50(5): 1276-1288.e5, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30902637

RESUMEN

Microbes colonize all body surfaces at birth and participate in the development of the immune system. In newborn mammals, the intestinal microbiota is first shaped by the dietary and immunological components of milk and then changes upon the introduction of solid food during weaning. Here, we explored the reactivity of the mouse intestinal immune system during the first weeks after birth and into adulthood. At weaning, the intestinal microbiota induced a vigorous immune response-a "weaning reaction"-that was programmed in time. Inhibition of the weaning reaction led to pathological imprinting and increased susceptibility to colitis, allergic inflammation, and cancer later in life. Prevention of this pathological imprinting was associated with the generation of RORγt+ regulatory T cells, which required bacterial and dietary metabolites-short-chain fatty acids and retinoic acid. Thus, the weaning reaction to microbiota is required for immune ontogeny, the perturbation of which leads to increased susceptibility to immunopathologies later in life.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Linfocitos T Reguladores/inmunología , Destete , Animales , Animales Recién Nacidos/inmunología , Animales Recién Nacidos/microbiología , Ácidos Grasos Volátiles/metabolismo , Ratones , Ratones Endogámicos C57BL , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Tretinoina/metabolismo
7.
Nature ; 609(7925): 159-165, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35831503

RESUMEN

RORγt is a lineage-specifying transcription factor that is expressed by immune cells that are enriched in the gastrointestinal tract and promote immunity, inflammation and tissue homeostasis1-15. However, fundamental questions remain with regard to the cellular heterogeneity among these cell types, the mechanisms that control protective versus inflammatory properties and their functional redundancy. Here we define all RORγt+ immune cells in the intestine at single-cell resolution and identify a subset of group 3 innate lymphoid cells (ILC3s) that expresses ZBTB46, a transcription factor specifying conventional dendritic cells16-20. ZBTB46 is robustly expressed by CCR6+ lymphoid-tissue-inducer-like ILC3s that are developmentally and phenotypically distinct from conventional dendritic cells, and its expression is imprinted by RORγt, fine-tuned by microbiota-derived signals and increased by pro-inflammatory cytokines. ZBTB46 restrains the inflammatory properties of ILC3s, including the OX40L-dependent expansion of T helper 17 cells and the exacerbated intestinal inflammation that occurs after enteric infection. Finally, ZBTB46+ ILC3s are a major source of IL-22, and selective depletion of this population renders mice susceptible to enteric infection and associated intestinal inflammation. These results show that ZBTB46 is a transcription factor that is shared between conventional dendritic cells and ILC3s, and identify a cell-intrinsic function for ZBTB46 in restraining the pro-inflammatory properties of ILC3s and a non-redundant role for ZBTB46+ ILC3s in orchestrating intestinal health.


Asunto(s)
Inmunidad Innata , Intestinos , Linfocitos , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares , Factores de Transcripción , Animales , Inflamación/inmunología , Inflamación/patología , Interleucinas , Intestinos/citología , Intestinos/inmunología , Intestinos/patología , Linfocitos/citología , Linfocitos/inmunología , Ratones , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Ligando OX40/metabolismo , Receptores CCR6/metabolismo , Células Th17/citología , Células Th17/inmunología , Factores de Transcripción/metabolismo , Interleucina-22
8.
Nature ; 610(7933): 744-751, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36071169

RESUMEN

Microbial colonization of the mammalian intestine elicits inflammatory or tolerogenic T cell responses, but the mechanisms controlling these distinct outcomes remain poorly understood, and accumulating evidence indicates that aberrant immunity to intestinal microbiota is causally associated with infectious, inflammatory and malignant diseases1-8. Here we define a critical pathway controlling the fate of inflammatory versus tolerogenic T cells that respond to the microbiota and express the transcription factor RORγt. We profiled all RORγt+ immune cells at single-cell resolution from the intestine-draining lymph nodes of mice and reveal a dominant presence of T regulatory (Treg) cells and lymphoid tissue inducer-like group 3 innate lymphoid cells (ILC3s), which co-localize at interfollicular regions. These ILC3s are distinct from extrathymic AIRE-expressing cells, abundantly express major histocompatibility complex class II, and are necessary and sufficient to promote microbiota-specific RORγt+ Treg cells and prevent their expansion as inflammatory T helper 17 cells. This occurs through ILC3-mediated antigen presentation, αV integrin and competition for interleukin-2. Finally, single-cell analyses suggest that interactions between ILC3s and RORγt+ Treg cells are impaired in inflammatory bowel disease. Our results define a paradigm whereby ILC3s select for antigen-specific RORγt+ Treg cells, and against T helper 17 cells, to establish immune tolerance to the microbiota and intestinal health.


Asunto(s)
Tolerancia Inmunológica , Intestinos , Linfocitos , Microbiota , Linfocitos T Reguladores , Animales , Inmunidad Innata , Integrina alfaV/metabolismo , Interleucina-2/inmunología , Intestinos/inmunología , Intestinos/microbiología , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Linfocitos/inmunología , Microbiota/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Análisis de la Célula Individual , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Factores de Transcripción/metabolismo , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/patología
9.
Nature ; 600(7890): 707-712, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34853467

RESUMEN

Pro-inflammatory T cells in the central nervous system (CNS) are causally associated with multiple demyelinating and neurodegenerative diseases1-6, but the pathways that control these responses remain unclear. Here we define a population of inflammatory group 3 innate lymphoid cells (ILC3s) that infiltrate the CNS in a mouse model of multiple sclerosis. These ILC3s are derived from the circulation, localize in proximity to infiltrating T cells in the CNS, function as antigen-presenting cells that restimulate myelin-specific T cells, and are increased in individuals with multiple sclerosis. Notably, antigen presentation by inflammatory ILC3s is required to promote T cell responses in the CNS and the development of multiple-sclerosis-like disease in mouse models. By contrast, conventional and tissue-resident ILC3s in the periphery do not appear to contribute to disease induction, but instead limit autoimmune T cell responses and prevent multiple-sclerosis-like disease when experimentally targeted to present myelin antigen. Collectively, our data define a population of inflammatory ILC3s that is essential for directly promoting T-cell-dependent neuroinflammation in the CNS and reveal the potential of harnessing peripheral tissue-resident ILC3s for the prevention of autoimmune disease.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Esclerosis Múltiple , Animales , Células Presentadoras de Antígenos , Antígenos/metabolismo , Inmunidad Innata , Linfocitos , Ratones , Enfermedades Neuroinflamatorias , Esclerosis/metabolismo
10.
Nature ; 568(7752): 405-409, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30944470

RESUMEN

Interleukin (IL)-2 is a pleiotropic cytokine that is necessary to prevent chronic inflammation in the gastrointestinal tract1-4. The protective effects of IL-2 involve the generation, maintenance and function of regulatory T (Treg) cells4-8, and the use of low doses of IL-2 has emerged as a potential therapeutic strategy for patients with inflammatory bowel disease9. However, the cellular and molecular pathways that control the production of IL-2 in the context of intestinal health are undefined. Here we show, in a mouse model, that IL-2 is acutely required to maintain Treg cells and immunological homeostasis throughout the gastrointestinal tract. Notably, lineage-specific deletion of IL-2 in T cells did not reduce Treg cells in the small intestine. Unbiased analyses revealed that, in the small intestine, group-3 innate lymphoid cells (ILC3s) are the dominant cellular source of IL-2, which is induced selectively by IL-1ß. Macrophages in the small intestine produce IL-1ß, and activation of this pathway involves MYD88- and NOD2-dependent sensing of the microbiota. Our loss-of-function studies show that ILC3-derived IL-2 is essential for maintaining Treg cells, immunological homeostasis and oral tolerance to dietary antigens in the small intestine. Furthermore, production of IL-2 by ILC3s was significantly reduced in the small intestine of patients with Crohn's disease, and this correlated with lower frequencies of Treg cells. Our results reveal a previously unappreciated pathway in which a microbiota- and IL-1ß-dependent axis promotes the production of IL-2 by ILC3s to orchestrate immune regulation in the intestine.


Asunto(s)
Inmunidad Innata/inmunología , Interleucina-2/inmunología , Intestinos/citología , Intestinos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Antígenos/administración & dosificación , Antígenos/inmunología , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Femenino , Microbioma Gastrointestinal/inmunología , Homeostasis/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Interleucina-2/deficiencia , Interleucina-2/metabolismo , Intestino Delgado/citología , Intestino Delgado/inmunología , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Proteína Adaptadora de Señalización NOD2/deficiencia , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/metabolismo , Linfocitos T Reguladores/clasificación , Linfocitos T Reguladores/metabolismo
11.
Immunity ; 42(1): 145-58, 2015 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-25577440

RESUMEN

Kupffer cells, the phagocytes of fetal origin that line the liver sinusoids, are key contributors of host defense against enteroinvasive bacteria. Here, we found that infection by Listeria monocytogenes induced the early necroptotic death of Kupffer cells, which was followed by monocyte recruitment and an anti-bacterial type 1 inflammatory response. Kupffer cell death also triggered a type 2 response that involved the hepatocyte-derived alarmin interleukin-33 (IL-33) and basophil-derived interleukin-4 (IL-4). This led to the alternative activation of the monocyte-derived macrophages recruited to the liver, which thereby replaced ablated Kupffer cells and restored liver homeostasis. Kupffer cell death is therefore a key signal orchestrating type 1 microbicidal inflammation and type-2-mediated liver repair upon infection. This indicates that beyond the classical dichotomy of type 1 and type 2 responses, these responses can develop sequentially in the context of a bacterial infection and act interdependently, orchestrating liver immune responses and return to homeostasis, respectively.


Asunto(s)
Macrófagos del Hígado/fisiología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Hígado/patología , Monocitos/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Vía Alternativa del Complemento , Homeostasis , Inflamación/microbiología , Interleucina-33 , Interleucina-4/metabolismo , Interleucinas/metabolismo , Macrófagos del Hígado/microbiología , Hígado/microbiología , Ratones , Ratones Endogámicos , Monocitos/microbiología , Necrosis , Fagocitosis , Cicatrización de Heridas
13.
Nat Immunol ; 12(4): 320-6, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21336274

RESUMEN

Lymphoid cells that express the nuclear hormone receptor RORγt are involved in containment of the large intestinal microbiota and defense against pathogens through the production of interleukin 17 (IL-17) and IL-22. They include adaptive IL-17-producing helper T cells (T(H)17 cells), as well as innate lymphoid cells (ILCs) such as lymphoid tissue-inducer (LTi) cells and IL-22-producing NKp46+ cells. Here we show that in contrast to T(H)17 cells, both types of RORγt+ ILCs constitutively produced most of the intestinal IL-22 and that the symbiotic microbiota repressed this function through epithelial expression of IL-25. This function was greater in the absence of adaptive immunity and was fully restored and required after epithelial damage, which demonstrates a central role for RORγt+ ILCs in intestinal homeostasis. Our data identify a finely tuned equilibrium among intestinal symbionts, adaptive immunity and RORγt+ ILCs.


Asunto(s)
Intestinos/inmunología , Tejido Linfoide/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Transducción de Señal/inmunología , Inmunidad Adaptativa/inmunología , Animales , Antígenos Ly/genética , Antígenos Ly/metabolismo , Femenino , Citometría de Flujo , Homeostasis/inmunología , Humanos , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Mucosa Intestinal/metabolismo , Intestinos/microbiología , Tejido Linfoide/citología , Tejido Linfoide/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Receptor 1 Gatillante de la Citotoxidad Natural/genética , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Simbiosis/inmunología , Factores de Tiempo , Interleucina-22
14.
Immunity ; 41(3): 366-374, 2014 Sep 18.
Artículo en Inglés | MEDLINE | ID: mdl-25238094

RESUMEN

Innate lymphoid cells (ILCs) were first described as playing important roles in the development of lymphoid tissues and more recently in the initiation of inflammation at barrier surfaces in response to infection or tissue damage. It has now become apparent that ILCs play more complex roles throughout the duration of immune responses, participating in the transition from innate to adaptive immunity and contributing to chronic inflammation. The proximity of ILCs to epithelial surfaces and their constitutive strategic positioning in other tissues throughout the body ensures that, in spite of their rarity, ILCs are able to regulate immune homeostasis effectively. Dysregulation of ILC function might result in chronic pathologies such as allergies, autoimmunity, and inflammation. A new role for ILCs in the maintenance of metabolic homeostasis has started to emerge, underlining their importance in fundamental physiological processes beyond infection and immunity.


Asunto(s)
Epitelio/inmunología , Inflamación/inmunología , Células Asesinas Naturales/inmunología , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Diferenciación Celular/inmunología , Citocinas/metabolismo , Humanos , Inmunidad Innata , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Ratones , Uniones Estrechas/inmunología
15.
Immunity ; 40(4): 608-20, 2014 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-24745335

RESUMEN

Segmented filamentous bacterium (SFB) is a symbiont that drives postnatal maturation of gut adaptive immune responses. In contrast to nonpathogenic E. coli, SFB stimulated vigorous development of Peyer's patches germinal centers but paradoxically induced only a low frequency of specific immunoglobulin A (IgA)-secreting cells with delayed accumulation of somatic mutations. Moreover, blocking Peyer's patch development abolished IgA responses to E. coli, but not to SFB. Indeed, SFB stimulated the postnatal development of isolated lymphoid follicles and tertiary lymphoid tissue, which substituted for Peyer's patches as inductive sites for intestinal IgA and SFB-specific T helper 17 (Th17) cell responses. Strikingly, in mice depleted of gut organized lymphoid tissue, SFB still induced a substantial but nonspecific intestinal Th17 cell response. These results demonstrate that SFB has the remarkable capacity to induce and stimulate multiple types of intestinal lymphoid tissues that cooperate to generate potent IgA and Th17 cell responses displaying only limited target specificity.


Asunto(s)
Infecciones por Clostridium/inmunología , Clostridium/inmunología , Infecciones por Escherichia coli/inmunología , Escherichia coli/inmunología , Inmunoglobulina A/metabolismo , Intestinos/inmunología , Células Plasmáticas/inmunología , Células Th17/inmunología , Animales , Antígenos Bacterianos/inmunología , Comunicación Celular , Diferenciación Celular , Interacciones Huésped-Patógeno , Tejido Linfoide/inmunología , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ganglios Linfáticos Agregados/inmunología
16.
Part Fibre Toxicol ; 20(1): 45, 2023 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-37996842

RESUMEN

BACKGROUND: Perinatal exposure to titanium dioxide (TiO2), as a foodborne particle, may influence the intestinal barrier function and the susceptibility to develop inflammatory bowel disease (IBD) later in life. Here, we investigate the impact of perinatal foodborne TiO2 exposure on the intestinal mucosal function and the susceptibility to develop IBD-associated colitis. Pregnant and lactating mother mice were exposed to TiO2 until pups weaning and the gut microbiota and intestinal barrier function of their offspring was assessed at day 30 post-birth (weaning) and at adult age (50 days). Epigenetic marks was studied by DNA methylation profile measuring the level of 5-methyl-2'-deoxycytosine (5-Me-dC) in DNA from colic epithelial cells. The susceptibility to develop IBD has been monitored using dextran-sulfate sodium (DSS)-induced colitis model. Germ-free mice were used to define whether microbial transfer influence the mucosal homeostasis and subsequent exacerbation of DSS-induced colitis. RESULTS: In pregnant and lactating mice, foodborne TiO2 was able to translocate across the host barriers including gut, placenta and mammary gland to reach embryos and pups, respectively. This passage modified the chemical element composition of foetus, and spleen and liver of mothers and their offspring. We showed that perinatal exposure to TiO2 early in life alters the gut microbiota composition, increases the intestinal epithelial permeability and enhances the colonic cytokines and myosin light chain kinase expression. Moreover, perinatal exposure to TiO2 also modifies the abilities of intestinal stem cells to survive, grow and generate a functional epithelium. Maternal TiO2 exposure increases the susceptibility of offspring mice to develop severe DSS-induced colitis later in life. Finally, transfer of TiO2-induced microbiota dysbiosis to pregnant germ-free mice affects the homeostasis of the intestinal mucosal barrier early in life and confers an increased susceptibility to develop colitis in adult offspring. CONCLUSIONS: Our findings indicate that foodborne TiO2 consumption during the perinatal period has negative long-lasting consequences on the development of the intestinal mucosal barrier toward higher colitis susceptibility. This demonstrates to which extent environmental factors influence the microbial-host interplay and impact the long-term mucosal homeostasis.


Asunto(s)
Colitis , Enfermedades Inflamatorias del Intestino , Embarazo , Femenino , Animales , Ratones , Disbiosis/inducido químicamente , Lactancia , Colitis/inducido químicamente , Colitis/genética , Colitis/metabolismo , Enfermedades Inflamatorias del Intestino/metabolismo , Ratones Endogámicos C57BL , Modelos Animales de Enfermedad
18.
Nature ; 535(7612): 440-443, 2016 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-27409807

RESUMEN

Group 3 innate lymphoid cells (ILC3) are major regulators of inflammation and infection at mucosal barriers. ILC3 development is thought to be programmed, but how ILC3 perceive, integrate and respond to local environmental signals remains unclear. Here we show that ILC3 in mice sense their environment and control gut defence as part of a glial­ILC3­epithelial cell unit orchestrated by neurotrophic factors. We found that enteric ILC3 express the neuroregulatory receptor RET. ILC3-autonomous Ret ablation led to decreased innate interleukin-22 (IL-22), impaired epithelial reactivity, dysbiosis and increased susceptibility to bowel inflammation and infection. Neurotrophic factors directly controlled innate Il22 downstream of the p38 MAPK/ERK-AKT cascade and STAT3 activation. Notably, ILC3 were adjacent to neurotrophic-factor-expressing glial cells that exhibited stellate-shaped projections into ILC3 aggregates. Glial cells sensed microenvironmental cues in a MYD88-dependent manner to control neurotrophic factors and innate IL-22. Accordingly, glial-intrinsic Myd88 deletion led to impaired production of ILC3-derived IL-22 and a pronounced propensity towards gut inflammation and infection. Our work sheds light on a novel multi-tissue defence unit, revealing that glial cells are central hubs of neuron and innate immune regulation by neurotrophic factor signals.


Asunto(s)
Inmunidad Innata , Intestinos/inmunología , Linfocitos/inmunología , Neuroglía/metabolismo , Neurotransmisores/metabolismo , Animales , Microambiente Celular/inmunología , Células Epiteliales/citología , Células Epiteliales/inmunología , Femenino , Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa , Inflamación/inmunología , Inflamación/metabolismo , Interleucinas/biosíntesis , Interleucinas/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Intestinos/citología , Linfocitos/citología , Linfocitos/metabolismo , Sistema de Señalización de MAP Quinasas , Masculino , Ratones , Factor 88 de Diferenciación Mieloide/deficiencia , Factor 88 de Diferenciación Mieloide/metabolismo , Neuroglía/inmunología , Neurotransmisores/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Proto-Oncogénicas c-ret/deficiencia , Proteínas Proto-Oncogénicas c-ret/metabolismo , Factor de Transcripción STAT3/metabolismo , Interleucina-22
19.
Semin Immunol ; 36: 56-57, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29258798

RESUMEN

In order to survive and reproduce, living organisms must be robust, tolerate injuries and undergo repair. Robustness in living organisms compares to robustness in human inventions, such as buildings and machines, which have to withstand occasional damage to avoid critical dysfunctions. However, the nature of robustness is fundamentally different in biology and in engineering. In living organisms, robustness is provided by homeostatic mechanisms, whereas in buildings and machines, it is provided by the redundancy of key elements. In this short essay, I discuss the nature of robustness in living organisms, and argue that redundancy, while important in engineering, is rare in biology.


Asunto(s)
Adaptación Fisiológica , Ingeniería , Homeostasis , Vida , Robótica , Animales , Evolución Biológica , Humanos , Cicatrización de Heridas
20.
Eur J Immunol ; 50(6): 779-782, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32330290

RESUMEN

In the past 15 years, it became clear that the symbiotic microbiota has an important impact on the development and regulation of the immune system. Consequently, it is incorrect to interpret a phenotype solely as a direct result of the genotype, without considering the impact of the microbiota. In fact, ignorance of the effects exerted by the microbiota may account for a large part of the "replication issues" found in many studies. In this issue of the European Journal of Immunology, Beller et al. [Eur. J. Immunol. 2020. 50: 783-794] provide data suggesting that eosinophils are not required to maintain IgA-producing plasma cells in the intestine, contrary to earlier reports. This paper shows that mice lacking eosinophils develop an altered intestinal microbiota, which poorly induces IgA. Normal levels of IgA were obtained in mice lacking eosinophils when these were colonized by microbiota from the WT mice. Therefore, the use of littermate controls carrying the same microbiota, in experiments comparing WT and mutant mice, is necessary to control the potential role of the microbiota. Nevertheless, caution should always be exercised in the interpretation of the results: changes in the microbiota may result from mutations in the host, and thereby, indirectly convey the effect of genotypes on phenotypes.


Asunto(s)
Eosinófilos/inmunología , Microbioma Gastrointestinal/inmunología , Inmunoglobulina A/inmunología , Células Plasmáticas/inmunología , Animales , Microbioma Gastrointestinal/genética , Humanos , Inmunoglobulina A/genética , Ratones , Ratones Mutantes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA