Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Gastroenterology ; 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38670280

RESUMEN

Celiac disease (CeD) is a gluten-induced enteropathy that develops in genetically susceptible individuals upon consumption of cereal gluten proteins. It is a unique and complex immune disorder to study, as the driving antigen is known and the tissue targeted by the immune reaction can be interrogated. This review integrates findings gained from genetic, biochemical, and immunologic studies, which together have revealed mechanisms of gluten peptide modification and HLA binding, thereby enabling a maladapted anti-gluten immune response. Observations in human samples combined with experimental mouse models have revealed that the gluten-induced immune response involves CD4+ T cells, cytotoxic CD8+ T cells, and B cells; their cross-talks are critical for the tissue-damaging response. The emergence of high-throughput technologies is increasing our understanding of the phenotype, location, and presumably function of the gluten-specific cells, which are all required to identify novel therapeutic targets and strategies for CeD.

3.
Curr Protoc ; 2(8): e515, 2022 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-35994521

RESUMEN

The design and use of mouse models that reproduce key features of human diseases are critical to advance our understanding of the pathogenesis of autoimmune diseases and to test new therapeutic strategies. Celiac disease is a unique organ-specific autoimmune-like disorder occurring in genetically susceptible individuals carrying HLA-DQ2 or HLA-DQ8 molecules who consume gluten. The key histological characteristic of the disease in humans is the destruction of the lining of the small intestine, a feature that has been difficult to reproduce in immunocompetent animal models. This unit describes the DQ8-Dd -villin-IL-15 transgenic mouse model of CeD, which was engineered based on the knowledge acquired from studying CeD patients' intestinal samples, and which represents the first animal model that develops villous atrophy in an HLA- and gluten-dependent manner without administration of any adjuvant. We provide detailed protocols for inducing and monitoring intestinal tissue damage, evaluating the cytotoxic properties of intraepithelial lymphocytes that mediate enterocyte lysis, and assessing the activation of the enzyme transglutaminase 2, which contributes to the generation of highly immunogenic gluten peptides. Detailed protocols to prepare pepsin-trypsin digested gliadin (PT-gliadin) or chymotrypsin-digested gliadin (CT-gliadin), which allow antibody detection against native or deamidated gluten peptides, are also provided in this unit. © 2022 The Authors. Current Protocols published by Wiley Periodicals LLC. Basic Protocol 1: Induction of celiac-like disease in DQ8-Dd -villin-IL-15tg mice Basic Protocol 2: Histological assessment of villous atrophy Support Protocol 1: Morphometric assessment of villous/crypt ratio Support Protocol 2: Evaluation of epithelial cells renewal Support Protocol 3: Evaluation of the density of intraepithelial lymphocytes Basic Protocol 3: Analysis of cytotoxic intraepithelial lymphocytes Basic Protocol 4: Transglutaminase 2 activation and measurement of antibodies against native and deamidated gluten peptides Support Protocol 4: Preparation of CT-gliadin Support Protocol 5: Preparation of PT-gliadin.


Asunto(s)
Enfermedad Celíaca , Animales , Atrofia , Enfermedad Celíaca/diagnóstico , Modelos Animales de Enfermedad , Gliadina , Glútenes , Humanos , Ratones , Ratones Transgénicos
4.
Cell Rep ; 36(4): 109456, 2021 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-34320346

RESUMEN

Interleukin-17 (IL-17)-producing γδ (γδ17) T cells are innate-like lymphocytes that contribute to protective anti-microbial responses but are also implicated in pathogenic inflammation at barrier sites. Understanding tissue-specific signals that regulate this subset is important to boost host defense mechanisms, but also to mitigate immunopathology. Here, we demonstrate that prostaglandin E2 (PGE2), a cyclooxygenase-dependent member of the eicosanoid family, directly enhances cytokine production by circulating and tissue-specific γδ17 T cells in vitro. Gain- and loss-of-function in vivo approaches further reveal that although provision of PGE2 amplifies psoriasiform inflammation, ablation of host mPGES1-dependent PGE2 synthesis is dispensable for cutaneous γδ17 T cell activation. By contrast, loss of endogenous PGE2 production or depletion of the gut microbiota compromises intestinal γδ17 T cell responses and increases disease severity during experimental colitis. Together, our results demonstrate how a lipid mediator can synergize with tissue-specific signals to enhance innate lymphocyte production of IL-17 during barrier inflammation.


Asunto(s)
Dinoprostona/farmacología , Inflamación/metabolismo , Interleucina-17/biosíntesis , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Piel/patología , Animales , Colitis/inducido químicamente , Sulfato de Dextran , Femenino , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Masculino , Ratones Endogámicos C57BL , Prostaglandina-E Sintasas/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/efectos de los fármacos
5.
Front Immunol ; 12: 674313, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34149709

RESUMEN

Several environmental, genetic, and immune factors create a "perfect storm" for the development of coeliac disease: the antigen gluten, the strong association of coeliac disease with HLA, the deamidation of gluten peptides by the enzyme transglutaminase 2 (TG2) generating peptides that bind strongly to the predisposing HLA-DQ2 or HLA-DQ8 molecules, and the ensuing unrestrained T cell response. T cell immunity is at the center of the disease contributing to the inflammatory process through the loss of tolerance to gluten and the differentiation of HLA-DQ2 or HLA-DQ8-restricted anti-gluten inflammatory CD4+ T cells secreting pro-inflammatory cytokines and to the killing of intestinal epithelial cells by cytotoxic intraepithelial CD8+ lymphocytes. However, recent studies emphasize that the individual contribution of each of these cell subsets is not sufficient and that interactions between these different populations of T cells and the simultaneous activation of innate and adaptive immune pathways in distinct gut compartments are required to promote disease immunopathology. In this review, we will discuss how tissue destruction in the context of coeliac disease results from the complex interactions between gluten, HLA molecules, TG2, and multiple innate and adaptive immune components.


Asunto(s)
Inmunidad Adaptativa/inmunología , Enfermedad Celíaca/inmunología , Glútenes/inmunología , Antígenos HLA/inmunología , Inmunidad Innata/inmunología , Animales , Enfermedad Celíaca/patología , Proteínas de Unión al GTP/inmunología , Humanos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas/inmunología
8.
Nature ; 578(7796): 600-604, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-32051586

RESUMEN

Coeliac disease is a complex, polygenic inflammatory enteropathy caused by exposure to dietary gluten that occurs in a subset of genetically susceptible individuals who express either the HLA-DQ8 or HLA-DQ2 haplotypes1,2. The need to develop non-dietary treatments is now widely recognized3, but no pathophysiologically relevant gluten- and HLA-dependent preclinical model exists. Furthermore, although studies in humans have led to major advances in our understanding of the pathogenesis of coeliac disease4, the respective roles of disease-predisposing HLA molecules, and of adaptive and innate immunity in the development of tissue damage, have not been directly demonstrated. Here we describe a mouse model that reproduces the overexpression of interleukin-15 (IL-15) in the gut epithelium and lamina propria that is characteristic of active coeliac disease, expresses the predisposing HLA-DQ8 molecule, and develops villous atrophy after ingestion of gluten. Overexpression of IL-15 in both the epithelium and the lamina propria is required for the development of villous atrophy, which demonstrates the location-dependent central role of IL-15 in the pathogenesis of coeliac disease. In addition, CD4+ T cells and HLA-DQ8 have a crucial role in the licensing of cytotoxic T cells to mediate intestinal epithelial cell lysis. We also demonstrate a role for the cytokine interferon-γ (IFNγ) and the enzyme transglutaminase 2 (TG2) in tissue destruction. By reflecting the complex interaction between gluten, genetics and IL-15-driven tissue inflammation, this mouse model provides the opportunity to both increase our understanding of coeliac disease, and develop new therapeutic strategies.


Asunto(s)
Enfermedad Celíaca/inmunología , Enfermedad Celíaca/patología , Glútenes/inmunología , Antígenos HLA-DQ/inmunología , Interleucina-15/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Femenino , Antígenos HLA-DQ/genética , Humanos , Interferón gamma/inmunología , Interleucina-15/genética , Masculino , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo
9.
Science ; 356(6333): 44-50, 2017 04 07.
Artículo en Inglés | MEDLINE | ID: mdl-28386004

RESUMEN

Viral infections have been proposed to elicit pathological processes leading to the initiation of T helper 1 (TH1) immunity against dietary gluten and celiac disease (CeD). To test this hypothesis and gain insights into mechanisms underlying virus-induced loss of tolerance to dietary antigens, we developed a viral infection model that makes use of two reovirus strains that infect the intestine but differ in their immunopathological outcomes. Reovirus is an avirulent pathogen that elicits protective immunity, but we discovered that it can nonetheless disrupt intestinal immune homeostasis at inductive and effector sites of oral tolerance by suppressing peripheral regulatory T cell (pTreg) conversion and promoting TH1 immunity to dietary antigen. Initiation of TH1 immunity to dietary antigen was dependent on interferon regulatory factor 1 and dissociated from suppression of pTreg conversion, which was mediated by type-1 interferon. Last, our study in humans supports a role for infection with reovirus, a seemingly innocuous virus, in triggering the development of CeD.


Asunto(s)
Antígenos/inmunología , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/virología , Glútenes/inmunología , Inflamación/virología , Infecciones por Reoviridae/complicaciones , Infecciones por Reoviridae/inmunología , Células TH1/inmunología , Animales , Dieta/efectos adversos , Modelos Animales de Enfermedad , Ingeniería Genética , Humanos , Tolerancia Inmunológica , Inflamación/inmunología , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/inmunología , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Intestinos/inmunología , Intestinos/patología , Intestinos/virología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor de Interferón alfa y beta/genética , Reoviridae/genética
10.
Nat Rev Immunol ; 15(12): 771-83, 2015 12.
Artículo en Inglés | MEDLINE | ID: mdl-26567920

RESUMEN

In this Opinion article, we discuss the function of tissues as a crucial checkpoint for the regulation of effector T cell responses, and the notion that interleukin-15 (IL-15) functions as a danger molecule that communicates to the immune system that the tissue is under attack and poises it to mediate tissue destruction. More specifically, we propose that expression of IL-15 in tissues promotes T helper 1 cell-mediated immunity and provides co-stimulatory signals to effector cytotoxic T cells to exert their effector functions and drive tissue destruction. Therefore, we think that IL-15 contributes to tissue protection by promoting the elimination of infected cells but that when its expression is chronically dysregulated, it can promote the development of complex T cell-mediated disorders associated with tissue destruction, such as coeliac disease and type 1 diabetes.


Asunto(s)
Inmunidad Celular/inmunología , Interleucina-15/inmunología , Transducción de Señal/inmunología , Linfocitos T/inmunología , Infecciones Bacterianas/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/metabolismo , Humanos , Interleucina-15/metabolismo , Subunidad alfa del Receptor de Interleucina-15/inmunología , Subunidad alfa del Receptor de Interleucina-15/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Modelos Inmunológicos , Unión Proteica/inmunología , Linfocitos T/metabolismo , Células TH1/inmunología , Células TH1/metabolismo
11.
J Exp Med ; 212(10): 1487-95, 2015 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-26304964

RESUMEN

Eicosanoids are inflammatory mediators that play a key but incompletely understood role in linking the innate and adaptive immune systems. Here, we show that cytotoxic effector T cells (CTLs) are capable of both producing and responding to cysteinyl leukotrienes (CystLTs), allowing for the killing of target cells in a T cell receptor-independent manner. This process is dependent on the natural killer receptor NKG2D and exposure to IL-15, a cytokine induced in distressed tissues. IL-15 and NKG2D signaling drives the up-regulation of key enzymes implicated in the synthesis of CystLTs, as well as the expression of CystLT receptors, suggesting a positive feedback loop. Finally, although the CystLT pathway has been previously linked to various allergic disorders, we provide unexpected evidence for its involvement in the pathogenesis of celiac disease (CD), a T helper 1 cell-mediated enteropathy induced by gluten. These findings provide new insights into the cytolytic signaling pathway of NKG2D and the pathogenesis of organ-specific immune disorders. Furthermore, they suggest that the blockade of CystLT receptors may represent a potent therapeutic target for CD or potentially other autoimmune disorders in which NKG2D has been implicated.


Asunto(s)
Enfermedad Celíaca/inmunología , Cisteína/metabolismo , Interleucina-15/inmunología , Leucotrienos/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Linfocitos T Citotóxicos/inmunología , Adulto , Araquidonato 5-Lipooxigenasa/genética , Araquidonato 5-Lipooxigenasa/metabolismo , Estudios de Casos y Controles , Enfermedad Celíaca/fisiopatología , Células Cultivadas , Cisteína/inmunología , Femenino , Humanos , Interleucina-15/metabolismo , Leucotrienos/inmunología , Masculino , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Receptores de Leucotrienos/genética , Receptores de Leucotrienos/metabolismo , Células TH1/inmunología , Células TH1/metabolismo , Regulación hacia Arriba
12.
Gastroenterology ; 149(3): 681-91.e10, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26001928

RESUMEN

BACKGROUND & AIMS: The mechanisms of tissue destruction during progression of celiac disease are poorly defined. It is not clear how tissue stress and adaptive immunity contribute to the activation of intraepithelial cytotoxic T cells and the development of villous atrophy. We analyzed epithelial cells and intraepithelial cytotoxic T cells in family members of patients with celiac disease, who were without any signs of adaptive antigluten immunity, and in potential celiac disease patients, who have antibodies against tissue transglutaminase 2 in the absence of villous atrophy. METHODS: We collected blood and intestinal biopsy specimens from 268 patients at tertiary medical centers in the United States and Italy from 2004 to 2012. All subjects had normal small intestinal histology. Study groups included healthy individuals with no family history of celiac disease or antibodies against tissue transglutaminase 2 (controls), healthy family members of patients with celiac disease, and potential celiac disease patients. Intraepithelial cytotoxic T cells were isolated and levels of inhibitory and activating natural killer (NK) cells were measured by flow cytometry. Levels of heat shock protein (HSP) and interleukin 15 were measured by immunohistochemistry, and ultrastructural alterations in intestinal epithelial cells (IECs) were assessed by electron microscopy. RESULTS: IECs from subjects with a family history of celiac disease, but not from subjects who already had immunity to gluten, expressed higher levels of HS27, HSP70, and interleukin-15 than controls; their IECs also had ultrastructural alterations. Intraepithelial cytotoxic T cells from relatives of patients with celiac disease expressed higher levels of activating NK receptors than cells from controls, although at lower levels than patients with active celiac disease, and without loss of inhibitory receptors for NK cells. Intraepithelial cytotoxic T cells from potential celiac disease patients failed to up-regulate activating NK receptors. CONCLUSIONS: A significant subset of healthy family members of patients with celiac disease with normal intestinal architecture had epithelial alterations, detectable by immunohistochemistry and electron microscopy. The adaptive immune response to gluten appears to act in synergy with epithelial stress to allow intraepithelial cytotoxic T cells to kill epithelial cells and induce villous atrophy in patients with active celiac disease.


Asunto(s)
Inmunidad Adaptativa , Enfermedad Celíaca/inmunología , Comunicación Celular , Células Epiteliales/inmunología , Mucosa Intestinal/inmunología , Intestino Delgado/inmunología , Estrés Fisiológico , Linfocitos T Citotóxicos/inmunología , Autoanticuerpos/sangre , Estudios de Casos y Controles , Enfermedad Celíaca/sangre , Enfermedad Celíaca/patología , Células Epiteliales/metabolismo , Células Epiteliales/ultraestructura , Proteínas de Unión al GTP/inmunología , Proteínas de Choque Térmico HSP27/inmunología , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas HSP70 de Choque Térmico/inmunología , Proteínas HSP70 de Choque Térmico/metabolismo , Proteínas de Choque Térmico , Humanos , Interleucina-15/inmunología , Interleucina-15/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/ultraestructura , Intestino Delgado/metabolismo , Intestino Delgado/ultraestructura , Italia , Chaperonas Moleculares , Fenotipo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Factores de Riesgo , Transducción de Señal , Linfocitos T Citotóxicos/metabolismo , Linfocitos T Citotóxicos/ultraestructura , Transglutaminasas/inmunología , Estados Unidos
13.
Immunol Rev ; 260(1): 221-34, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24942692

RESUMEN

Interleukin-15 (IL-15) exerts many biological functions essential for the maintenance and function of multiple cell types. Although its expression is tightly regulated, IL-15 upregulation has been reported in many organ-specific autoimmune disorders. In celiac disease, an intestinal inflammatory disorder driven by gluten exposure, the upregulation of IL-15 expression in the intestinal mucosa has become a hallmark of the disease. Interestingly, because it is overexpressed both in the gut epithelium and in the lamina propria, IL-15 acts on distinct cell types and impacts distinct immune components and pathways to disrupt intestinal immune homeostasis. In this article, we review our current knowledge of the multifaceted roles of IL-15 with regard to the main immunological processes involved in the pathogenesis of celiac disease.


Asunto(s)
Enfermedad Celíaca/inmunología , Enfermedad Celíaca/metabolismo , Interleucina-15/metabolismo , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Enfermedad Celíaca/genética , Regulación de la Expresión Génica , Homeostasis , Humanos , Tolerancia Inmunológica , Interleucina-15/genética , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Transducción de Señal , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
14.
PLoS One ; 8(9): e76292, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24086722

RESUMEN

NK cells are large granular lymphocytes that form a critical component of the innate immune system, whose functions include the killing of cells expressing stress-induced molecules. It is increasingly accepted that despite being considered prototypical effector cells, NK cells require signals to reach their full cytotoxic potential. We previously showed that IL-15 is capable of arming CD8 effector T cells to kill independently of their TCR via NKG2D in a cPLA2-dependent process. As NK cells also express NKG2D, we wanted to investigate whether this pathway functioned in an analogous manner and if resting NK cells could be primed to the effector phase by IL-15. Furthermore, to establish relevance to human disease we studied a possible role for this pathway in the pathogenesis of psoriatic arthritis, since there are aspects of this disease that suggest a potential effector role for the innate immune system. We found that PsA patients had upregulated IL-15 and MIC in their affected synovial tissues, and that this unique inflammatory environment enabled NK cell activation and killing via NKG2D and cPLA2. Moreover, we were able to reproduce the phenotype of joint NK cells from blood NK cells by incubating them with IL-15. Altogether, these findings suggest a destructive role for NK cells when activated by environmental stress signals during the pathogenesis of PsA and demonstrate that IL-15 is capable of priming resting NK cells in tissues to the effector phase.


Asunto(s)
Artritis Psoriásica/inmunología , Regulación de la Expresión Génica/inmunología , Interleucina-15/inmunología , Células Asesinas Naturales/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Fosfolipasas A2 Citosólicas/metabolismo , Transducción de Señal/inmunología , Artritis Psoriásica/metabolismo , Citocinas/genética , Citocinas/metabolismo , Citometría de Flujo , Humanos , Inmunohistoquímica , Interleucina-15/metabolismo , Células Asesinas Naturales/metabolismo , ARN Interferente Pequeño/genética , Transfección
15.
Immunity ; 37(5): 917-29, 2012 Nov 16.
Artículo en Inglés | MEDLINE | ID: mdl-23142782

RESUMEN

The bone marrow (BM) has been identified as a possible organ for T cell priming, yet the fundamental mechanisms of a polyclonal immune response in the BM remain unknown. We found that after intradermal injection of modified vaccinia Ankara virus, unexpected sources of newly primed polyclonal virus-specific CD8(+), but not CD4(+), T cells were localized in the BM and the draining lymph nodes (dLNs) prior to blood circulation. We identified neutrophils as the virus-carrier cells from the dermis to the BM. In both neutrophil-depleted and Ccr1(-/-) mice, virus-specific BM CD8(+) responses were lost. Myeloid antigen-presenting cells were required for BM CD8(+) T cell priming. A systems biology analysis of dLN and BM virus-specific CD8(+) T cells revealed distinct transcriptional and multifunctional profiles for cells primed in each organ. We provide direct evidence for how antigen is transported to the BM, providing a source of virus-specific memory CD8(+) T cells.


Asunto(s)
Antígenos/inmunología , Médula Ósea/inmunología , Linfocitos T CD8-positivos/inmunología , Dermis/inmunología , Memoria Inmunológica/inmunología , Neutrófilos/inmunología , Animales , Células Presentadoras de Antígenos/inmunología , Linfocitos T CD4-Positivos/inmunología , Femenino , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Células Mieloides/inmunología , Receptores CCR1/inmunología
16.
Semin Immunopathol ; 34(4): 551-66, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22660791

RESUMEN

Celiac disease is a T cell-mediated immune disorder induced by dietary gluten that is characterized by the development of an inflammatory anti-gluten CD4 T cell response, anti-gluten antibodies, and autoantibodies against tissue transglutaminase 2 and the activation of intraepithelial lymphocytes (IELs) leading to the destruction of the intestinal epithelium. Intraepithelial lymphocytes represent a heterogeneous population of T cells composed mainly of cytotoxic CD8 T cells residing within the epithelial layer, whose main role is to maintain the integrity of the epithelium by eliminating infected cells and promoting epithelial repair. Dysregulated activation of IELs is a hallmark of CD and is critically involved in epithelial cell destruction and the subsequent development of villous atrophy. In this review, we compare and contrast the phenotype and function of human and mouse small intestinal IELs under physiological conditions. Furthermore, we discuss how conditions of epithelial distress associated with overexpression of IL-15 and non-classical MHC class I molecules induce cytotoxic IELs to become licensed killer cells that upregulate activating NKG2D and CD94/NKG2C natural killer receptors, acquiring lymphokine killer activity. Pathways leading to dysregulated IEL activation could eventually be targeted to prevent villous atrophy and treat patients who respond poorly to gluten-free diet.


Asunto(s)
Enfermedad Celíaca/inmunología , Mucosa Intestinal/inmunología , Linfocitos/inmunología , Animales , Enfermedad Celíaca/metabolismo , Homeostasis/inmunología , Humanos , Mucosa Intestinal/metabolismo , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Linfocitos/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2
17.
Annu Rev Immunol ; 29: 493-525, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21219178

RESUMEN

Celiac disease (CD) is a gluten-sensitive enteropathy that develops in genetically susceptible individuals by exposure to cereal gluten proteins. This review integrates insights from immunological studies with results of recent genetic genome-wide association studies into a disease model. Genetic data, among others, suggest that viral infections are implicated and that natural killer effector pathways are important in the pathogenesis of CD, but most prominently these data converge with existing immunological findings that CD is primarily a T cell-mediated immune disorder in which CD4(+) T cells that recognize gluten peptides in the context of major histocompatibility class II molecules play a central role. Comparison of genetic pathways as well as genetic susceptibility loci between CD and other autoimmune and inflammatory disorders reveals that CD bears stronger resemblance to T cell-mediated organ-specific autoimmune than to inflammatory diseases. Finally, we present evidence suggesting that the high prevalence of CD in modern societies may be the by-product of past selection for increased immune responses to combat infections in populations in which agriculture and cereals were introduced early on in the post-Neolithic period.


Asunto(s)
Enfermedad Celíaca/genética , Enfermedad Celíaca/inmunología , Animales , Enfermedad Celíaca/epidemiología , Enfermedad Celíaca/fisiopatología , Predisposición Genética a la Enfermedad , Glútenes/inmunología , Humanos , Factores de Riesgo
18.
J Immunol ; 184(4): 2038-47, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20083674

RESUMEN

Early immune response to the largely used Mycobacterium bovis bacillus Calmette-Guérin (BCG) intradermal vaccine remains ill defined. Three days after BCG inoculation into the mouse ear, in addition to neutrophils infiltrating skin, we observed CD11b(+)Ly-6C(int)Ly-6G(-) myeloid cells. Neutrophil depletion markedly enhanced their recruitment. These cells differed from inflammatory monocytes and required MyD88-dependent BCG-specific signals to invade skin, whereas neutrophil influx was MyD88 independent. Upon BCG phagocytosis, CD11b(+)Ly-6C(int)Ly-6G(-) cells produced NO, which required the IL-1 receptor. Despite NO production, they were unable to kill BCG or the nonpathogenic Mycobacterium smegmatis. However, they markedly impaired T cell priming in the draining lymph node. Their elimination by all-trans retinoid acid treatment increased the number of IFN-gamma-producing CD4 T cells. Thus, BCG vaccination recruits innate myeloid-derived suppressor cells, akin to mouse tumor-infiltrating cells. These propathogenic cells dampen the early T cell response and might facilitate BCG persistence.


Asunto(s)
Vacuna BCG/administración & dosificación , Vacuna BCG/inmunología , Movimiento Celular/inmunología , Activación de Linfocitos/inmunología , Células Mieloides/inmunología , Óxido Nítrico/biosíntesis , Receptores de Interleucina-1/fisiología , Subgrupos de Linfocitos T/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Femenino , Inmunidad Innata , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/citología , Neutrófilos/inmunología , Neutrófilos/patología , Óxido Nítrico/fisiología , Receptores de Interleucina-1/antagonistas & inhibidores , Subgrupos de Linfocitos T/patología
19.
PLoS One ; 4(12): e8159, 2009 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-19997562

RESUMEN

BACKGROUND: Obtaining a certain multi-functionality of cellular immunity for the control of infectious diseases is a burning question in immunology and in vaccine design. Early events, including antigen shuttling to secondary lymphoid organs and recruitment of innate immune cells for adaptive immune response, determine host responsiveness to antigens. However, the sequence of these events and their impact on the quality of the immune response remain to be elucidated. Here, we chose to study Modified Vaccinia virus Ankara (MVA) which is now replacing live Smallpox vaccines and is proposed as an attenuated vector for vaccination strategies against infectious diseases. METHODOLOGY/PRINCIPAL FINDINGS: We analyzed in vivo mechanisms triggered following intradermal (i.d.) and intramuscular (i.m.) Modified Vaccinia virus Ankara (MVA) administration. We demonstrated significant differences in the antigen shuttling to lymphoid organs by macrophages (MPhis), myeloid dendritic cells (DCs), and neutrophils (PMNs). MVA i.d. administration resulted in better antigen distribution and more sustained antigen-presenting cells (APCs) recruitment into draining lymph nodes than with i.m. administration. These APCs, which comprise both DCs and MPhis, were differentially involved in T cell priming and shaped remarkably the quality of cytokine-producing virus-specific T cells according to the entry route of MVA. CONCLUSIONS/SIGNIFICANCE: This study improves our understanding of the mechanisms of antigen delivery and their consequences on the quality of immune responses and provides new insights for vaccine development.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Antígenos/inmunología , Inmunidad/inmunología , Animales , Células Presentadoras de Antígenos/citología , Antígenos/administración & dosificación , Reactividad Cruzada/inmunología , Femenino , Inmunización , Inyecciones Intradérmicas , Inyecciones Intramusculares , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología , Células Mieloides/inmunología , Fenotipo , Linfocitos T/citología , Linfocitos T/inmunología , Distribución Tisular/inmunología , Virus Vaccinia/inmunología
20.
J Invest Dermatol ; 129(5): 1156-64, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19052565

RESUMEN

Particle-based drug delivery systems target active compounds to the hair follicle and may result in a better penetration and higher efficiency of compound uptake by skin resident cells. As previously proposed, such delivery systems could be important tools for vaccine delivery. In this study, we investigated the penetration of solid fluorescent 40 or 200 nm polystyrene nanoparticles (NPs) as well as virus particles in murine skin to further investigate the efficacy of transcutaneously (TC) applied particulate vaccine delivery route. We demonstrated that 40 and 200 nm NPs and modified vaccinia Ankara (MVA) expressing the green-fluorescent protein penetrated deeply into hair follicles and were internalized by perifollicular antigen-presenting cells (APCs). Fibered-based confocal microscopy analyses allowed visualizing in vivo particle penetration along the follicular duct, diffusion into the surrounding tissue, uptake by APCs and transport to the draining lymph nodes. The application of small particles, such as ovalbumin coding DNA or MVA, induced both humoral and cellular immune responses. Furthermore, TC applied MVA induced protection against vaccinia virus challenge. Our results strengthen the concept of TC targeting of cutaneous APCs by hair follicles and will contribute to the development of advanced vaccination protocols using NPs or viral vectors.


Asunto(s)
Células Presentadoras de Antígenos/metabolismo , Folículo Piloso/metabolismo , Nanoestructuras/administración & dosificación , Vacunas/administración & dosificación , Vaccinia/metabolismo , Administración Tópica , Animales , Anticuerpos Antivirales/metabolismo , Células Presentadoras de Antígenos/citología , Transporte Biológico/fisiología , Modelos Animales de Enfermedad , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Folículo Piloso/citología , Células de Langerhans/citología , Células de Langerhans/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/metabolismo , Ratones , Ratones Endogámicos C57BL , Nanotecnología/métodos , Poliestirenos/metabolismo , Vaccinia/inmunología , Virosis/inmunología , Virosis/prevención & control
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...