Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 751
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nature ; 599(7883): 125-130, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34671159

RESUMEN

Tissue maintenance and repair depend on the integrated activity of multiple cell types1. Whereas the contributions of epithelial2,3, immune4,5 and stromal cells6,7 in intestinal tissue integrity are well understood, the role of intrinsic neuroglia networks remains largely unknown. Here we uncover important roles of enteric glial cells (EGCs) in intestinal homeostasis, immunity and tissue repair. We demonstrate that infection of mice with Heligmosomoides polygyrus leads to enteric gliosis and the upregulation of an interferon gamma (IFNγ) gene signature. IFNγ-dependent gene modules were also induced in EGCs from patients with inflammatory bowel disease8. Single-cell transcriptomics analysis of the tunica muscularis showed that glia-specific abrogation of IFNγ signalling leads to tissue-wide activation of pro-inflammatory transcriptional programs. Furthermore, disruption of the IFNγ-EGC signalling axis enhanced the inflammatory and granulomatous response of the tunica muscularis to helminths. Mechanistically, we show that the upregulation of Cxcl10 is an early immediate response of EGCs to IFNγ signalling and provide evidence that this chemokine and the downstream amplification of IFNγ signalling in the tunica muscularis are required for a measured inflammatory response to helminths and resolution of the granulomatous pathology. Our study demonstrates that IFNγ signalling in enteric glia is central to intestinal homeostasis and reveals critical roles of the IFNγ-EGC-CXCL10 axis in immune response and tissue repair after infectious challenge.


Asunto(s)
Homeostasis , Intestinos/inmunología , Intestinos/fisiología , Neuroglía/inmunología , Neuroglía/fisiología , Regeneración , Adventicia/inmunología , Adventicia/parasitología , Animales , Quimiocina CXCL10/inmunología , Duodeno/inmunología , Duodeno/parasitología , Duodeno/patología , Duodeno/fisiología , Femenino , Gliosis , Homeostasis/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Interferón gamma/inmunología , Intestinos/parasitología , Intestinos/patología , Masculino , Ratones , Nematospiroides dubius/inmunología , Nematospiroides dubius/patogenicidad , Transducción de Señal/inmunología , Infecciones por Strongylida/inmunología , Infecciones por Strongylida/parasitología , Infecciones por Strongylida/patología
2.
Gastroenterology ; 167(1): 104-115, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38286391

RESUMEN

In its conventional form, celiac disease (CeD) is characterized by both positive serology and flat villi in the duodenum, and is well known by gastroenterologists and general practitioners. The aim of this review was to shed light on 2 neglected and not yet well-defined celiac phenotypes, that is, seronegative and ultrashort CeD. Seronegative CeD can be suspected in the presence of flat villi, positive HLA-DQ2 and/or HLA-DQ8, and the absence of CeD antibodies. After ruling out other seronegative enteropathies, the diagnosis can be confirmed by both clinical and histologic improvements after 1 year of a gluten-free diet. Ultrashort CeD is characterized by the finding of flat villi in the duodenal bulb in the absence of mucosal damage in the distal duodenum and with serologic positivity. Data on the prevalence, clinical manifestations, histologic lesions, genetic features, and outcome of seronegative and ultrashort CeD are inconclusive due to the few studies available and the small number of patients diagnosed. Some additional diagnostic tools have been developed recently, such as assessing intestinal transglutaminase 2 deposits, flow cytometry technique, microRNA detection, or proteomic analysis, and they seem to be useful in the identification of complex cases. Further cooperative studies are highly desirable to improve the knowledge of these 2 still-obscure variants of CeD.


Asunto(s)
Enfermedad Celíaca , Dieta Sin Gluten , Duodeno , Antígenos HLA-DQ , Enfermedad Celíaca/diagnóstico , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/sangre , Humanos , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/sangre , Antígenos HLA-DQ/inmunología , Duodeno/patología , Duodeno/inmunología , Fenotipo , Transglutaminasas/inmunología , Mucosa Intestinal/patología , Mucosa Intestinal/inmunología , Proteína Glutamina Gamma Glutamiltransferasa 2 , Biopsia , Proteínas de Unión al GTP/inmunología , Biomarcadores/sangre , Autoanticuerpos/sangre , Pruebas Serológicas , Valor Predictivo de las Pruebas
3.
Nature ; 569(7754): 126-130, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30988509

RESUMEN

The intestinal immune system has the challenging task of tolerating foreign nutrients and the commensal microbiome, while excluding or eliminating ingested pathogens. Failure of this balance leads to conditions such as inflammatory bowel diseases, food allergies and invasive gastrointestinal infections1. Multiple immune mechanisms are therefore in place to maintain tissue integrity, including balanced generation of effector T (TH) cells and FOXP3+ regulatory T (pTreg) cells, which mediate resistance to pathogens and regulate excessive immune activation, respectively1-4. The gut-draining lymph nodes (gLNs) are key sites for orchestrating adaptive immunity to luminal perturbations5-7. However, it is unclear how they simultaneously support tolerogenic and inflammatory reactions. Here we show that gLNs are immunologically specific to the functional gut segment that they drain. Stromal and dendritic cell gene signatures and polarization of T cells against the same luminal antigen differ between gLNs, with the proximal small intestine-draining gLNs preferentially giving rise to tolerogenic responses and the distal gLNs to pro-inflammatory T cell responses. This segregation permitted the targeting of distal gLNs for vaccination and the maintenance of duodenal pTreg cell induction during colonic infection. Conversely, the compartmentalized dichotomy was perturbed by surgical removal of select distal gLNs and duodenal infection, with effects on both lymphoid organ and tissue immune responses. Our findings reveal that the conflict between tolerogenic and inflammatory intestinal responses is in part resolved by discrete gLN drainage, and encourage antigen targeting to specific gut segments for therapeutic immune modulation.


Asunto(s)
Duodeno/inmunología , Ganglios Linfáticos/inmunología , Linfocitos T/inmunología , Animales , Antígenos CD4/metabolismo , Diferenciación Celular , Movimiento Celular , Polaridad Celular , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Duodeno/citología , Duodeno/microbiología , Femenino , Ganglios Linfáticos/citología , Ganglios Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Boca/inmunología , Boca/microbiología , Ratas , Ratas Wistar , Células del Estroma/inmunología , Células del Estroma/microbiología , Linfocitos T/citología , Linfocitos T/microbiología
4.
Clin Immunol ; 263: 110202, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38575045

RESUMEN

Celiac disease (CD) is an immune-driven disease characterized by tissue damage in the small intestine of genetically-susceptible individuals. We evaluated here a crucial immune regulatory pathway involving TYRO3, AXL, and MERTK (TAM) receptors and their ligands PROS1 and GAS6 in duodenal biopsies of controls and CD patients. We found increased GAS6 expression associated with downregulation of PROS1 and variable TAM receptors levels in duodenum tissue of CD patients. Interestingly, CD3+ lymphocytes, CD68+, CD11c+ myeloid and epithelial cells, showed differential expressions of TAM components comparing CD vs controls. Principal component analysis revealed a clear segregation of two groups of CD patients based on TAM components and IFN signaling. In vitro validation demonstrated that monocytes, T lymphocytes and epithelial cells upregulated TAM components in response to IFN stimulation. Our findings highlight a dysregulated TAM axis in CD related to IFN signaling and contribute to a deeper understanding of the pathophysiology of CD.


Asunto(s)
Tirosina Quinasa del Receptor Axl , Enfermedad Celíaca , Duodeno , Péptidos y Proteínas de Señalización Intercelular , Mucosa Intestinal , Proteína S , Proteínas Tirosina Quinasas Receptoras , Tirosina Quinasa c-Mer , Humanos , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/genética , Proteínas Tirosina Quinasas Receptoras/metabolismo , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/inmunología , Masculino , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Femenino , Péptidos y Proteínas de Señalización Intercelular/genética , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Adulto , Duodeno/metabolismo , Duodeno/inmunología , Duodeno/patología , Tirosina Quinasa c-Mer/genética , Tirosina Quinasa c-Mer/metabolismo , Proteína S/metabolismo , Proteína S/genética , Persona de Mediana Edad , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Adulto Joven , Transducción de Señal , Adolescente , Interferones/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo
5.
N Engl J Med ; 385(1): 35-45, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34192430

RESUMEN

BACKGROUND: In celiac disease, small intestinal transglutaminase 2 causes deamidation of glutamine residues in gluten peptides, which enhances stimulation of T cells and leads to mucosal injury. Inhibition of transglutaminase 2 is a potential treatment for celiac disease. METHODS: In a proof-of-concept trial, we assessed the efficacy and safety of a 6-week treatment with ZED1227, a selective oral transglutaminase 2 inhibitor, at three dose levels as compared with placebo, in adults with well-controlled celiac disease who underwent a daily gluten challenge. The primary end point was the attenuation of gluten-induced mucosal damage, as measured by the ratio of villus height to crypt depth. Secondary end points included intraepithelial lymphocyte density, the Celiac Symptom Index score, and the Celiac Disease Questionnaire score (for assessment of health-related quality of life). RESULTS: Of the 41 patients assigned to the 10-mg ZED1227 group, the 41 assigned to the 50-mg group, the 41 assigned to the 100-mg group, and the 40 assigned to the placebo group, 35, 39, 38, and 30 patients, respectively, had adequate duodenal-biopsy samples for the assessment of the primary end point. Treatment with ZED1227 at all three dose levels attenuated gluten-induced duodenal mucosal injury. The estimated difference from placebo in the change in the mean ratio of villus height to crypt depth from baseline to week 6 was 0.44 (95% confidence interval [CI], 0.15 to 0.73) in the 10-mg group (P = 0.001), 0.49 (95% CI, 0.20 to 0.77) in the 50-mg group (P<0.001), and 0.48 (95% CI, 0.20 to 0.77) in the 100-mg group (P<0.001). The estimated differences from placebo in the change in intraepithelial lymphocyte density were -2.7 cells per 100 epithelial cells (95% CI, -7.6 to 2.2) in the 10-mg group, -4.2 cells per 100 epithelial cells (95% CI, -8.9 to 0.6) in the 50-mg group, and -9.6 cells per 100 epithelial cells (95% CI, -14.4 to -4.8) in the 100-mg group. Use of the 100-mg dose may have improved symptom and quality-of-life scores. The most common adverse events, the incidences of which were similar across all groups, were headache, nausea, diarrhea, vomiting, and abdominal pain. Rash developed in 3 of 40 patients (8%) in the 100-mg group. CONCLUSIONS: In this preliminary trial, treatment with ZED1227 attenuated gluten-induced duodenal mucosal damage in patients with celiac disease. (Funded by Dr. Falk Pharma; CEC-3 EudraCT number, 2017-002241-30.).


Asunto(s)
Enfermedad Celíaca/tratamiento farmacológico , Duodeno/patología , Proteínas de Unión al GTP/antagonistas & inhibidores , Imidazoles/administración & dosificación , Mucosa Intestinal/patología , Piridinas/administración & dosificación , Transglutaminasas/antagonistas & inhibidores , Administración Oral , Adulto , Enfermedad Celíaca/patología , Relación Dosis-Respuesta a Droga , Método Doble Ciego , Duodeno/inmunología , Femenino , Glútenes/administración & dosificación , Glútenes/efectos adversos , Humanos , Imidazoles/efectos adversos , Mucosa Intestinal/inmunología , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Prueba de Estudio Conceptual , Proteína Glutamina Gamma Glutamiltransferasa 2 , Piridinas/efectos adversos , Calidad de Vida , Índice de Severidad de la Enfermedad
6.
J Autoimmun ; 146: 103241, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38754235

RESUMEN

Many antibody responses induced by infection, vaccination or autoimmunity show signs of convergence across individuals with epitope-dependent selection of particular variable region gene segments and complementarity determining region 3 properties. However, not much is known about the relationship between antigen-specific effector cells and antigen-specific precursors present in the naïve B-cell repertoire. Here, we sought to address this relationship in the context of celiac disease, where there is a stereotyped autoantibody response against the enzyme transglutaminase 2 (TG2). By generating TG2-specific monoclonal antibodies from both duodenal plasma cells and circulating naïve B cells, we demonstrate a discord between the naïve TG2-specific repertoire and the cells that are selected for autoantibody production. Hence, the naïve repertoire does not fully reflect the epitope preference and gene usage observed for memory B cells and plasma cells. Instead, distinct naïve B cells that target particular TG2 epitopes appear to be selectively activated at the expense of TG2-binding B cells targeting other epitopes.


Asunto(s)
Autoanticuerpos , Linfocitos B , Enfermedad Celíaca , Epítopos de Linfocito B , Proteínas de Unión al GTP , Activación de Linfocitos , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas , Enfermedad Celíaca/inmunología , Humanos , Autoanticuerpos/inmunología , Transglutaminasas/inmunología , Epítopos de Linfocito B/inmunología , Proteínas de Unión al GTP/inmunología , Activación de Linfocitos/inmunología , Linfocitos B/inmunología , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo , Femenino , Anticuerpos Monoclonales/inmunología , Epítopos/inmunología , Masculino , Adulto , Duodeno/inmunología , Duodeno/patología
7.
Immunity ; 43(4): 629-31, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26488813

RESUMEN

In this issue of Immunity, Haghikia and colleagues (2015) demonstrate that dietary fatty acids, by modulating gut microbes and their metabolism, regulate mucosal immune cells to impact systemic immunity. Using this mechanism, dietary and bacteria-derived medium-chain and long-chain fatty acids exacerbate, whereas short-chain fatty acids ameliorate, autoimmunity in the brain.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Sistema Nervioso Central/inmunología , Grasas de la Dieta/farmacología , Duodeno/inmunología , Encefalomielitis Autoinmune Experimental/etiología , Ácidos Grasos/farmacología , Linfopoyesis/efectos de los fármacos , Subgrupos de Linfocitos T/efectos de los fármacos , Animales
8.
Immunity ; 43(4): 788-802, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26410628

RESUMEN

Experimental IgE-mediated food allergy depends on intestinal anaphylaxis driven by interleukin-9 (IL-9). However, the primary cellular source of IL-9 and the mechanisms underlying the susceptibility to food-induced intestinal anaphylaxis remain unclear. Herein, we have reported the identification of multifunctional IL-9-producing mucosal mast cells (MMC9s) that can secrete prodigious amounts of IL-9 and IL-13 in response to IL-33, and mast cell protease-1 (MCPt-1) in response to antigen and IgE complex crosslinking, respectively. Repeated intragastric antigen challenge induced MMC9 development that required T cells, IL-4, and STAT6 transcription factor, but not IL-9 signals. Mice ablated of MMC9 induction failed to develop intestinal mastocytosis, which resulted in decreased food allergy symptoms that could be restored by adoptively transferred MMC9s. Finally, atopic patients that developed food allergy displayed increased intestinal expression of Il9- and MC-specific transcripts. Thus, the induction of MMC9s is a pivotal step to acquire the susceptibility to IgE-mediated food allergy.


Asunto(s)
Hipersensibilidad a los Alimentos/inmunología , Inmunoglobulina E/inmunología , Interleucina-9/metabolismo , Mucosa Intestinal/inmunología , Mastocitos/inmunología , Mastocitosis/inmunología , Traslado Adoptivo , Anafilaxia/etiología , Anafilaxia/inmunología , Animales , Secuencia de Bases , Células de la Médula Ósea/citología , Linaje de la Célula , Quimasas/biosíntesis , Quimasas/genética , Diarrea/etiología , Diarrea/inmunología , Susceptibilidad a Enfermedades , Duodeno/inmunología , Duodeno/patología , Hipersensibilidad a los Alimentos/etiología , Hipersensibilidad a los Alimentos/patología , Humanos , Hipersensibilidad Inmediata/complicaciones , Interleucina-9/biosíntesis , Interleucina-9/genética , Interleucinas/biosíntesis , Interleucinas/metabolismo , Interleucinas/fisiología , Mastocitos/metabolismo , Mastocitos/trasplante , Mastocitosis/patología , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología , Ovalbúmina/toxicidad , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Factor de Transcripción STAT6/fisiología , Especificidad de la Especie , Linfocitos T/inmunología
9.
Immunity ; 43(4): 817-29, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26488817

RESUMEN

Growing empirical evidence suggests that nutrition and bacterial metabolites might impact the systemic immune response in the context of disease and autoimmunity. We report that long-chain fatty acids (LCFAs) enhanced differentiation and proliferation of T helper 1 (Th1) and/or Th17 cells and impaired their intestinal sequestration via p38-MAPK pathway. Alternatively, dietary short-chain FAs (SCFAs) expanded gut T regulatory (Treg) cells by suppression of the JNK1 and p38 pathway. We used experimental autoimmune encephalomyelitis (EAE) as a model of T cell-mediated autoimmunity to show that LCFAs consistently decreased SCFAs in the gut and exacerbated disease by expanding pathogenic Th1 and/or Th17 cell populations in the small intestine. Treatment with SCFAs ameliorated EAE and reduced axonal damage via long-lasting imprinting on lamina-propria-derived Treg cells. These data demonstrate a direct dietary impact on intestinal-specific, and subsequently central nervous system-specific, Th cell responses in autoimmunity, and thus might have therapeutic implications for autoimmune diseases such as multiple sclerosis.


Asunto(s)
Autoinmunidad/efectos de los fármacos , Sistema Nervioso Central/inmunología , Grasas de la Dieta/farmacología , Duodeno/inmunología , Encefalomielitis Autoinmune Experimental/etiología , Ácidos Grasos/farmacología , Linfopoyesis/efectos de los fármacos , Subgrupos de Linfocitos T/efectos de los fármacos , Animales , Grasas de la Dieta/toxicidad , Duodeno/metabolismo , Duodeno/microbiología , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/metabolismo , Ácidos Grasos/química , Ácidos Grasos/toxicidad , Trasplante de Microbiota Fecal , Microbioma Gastrointestinal/fisiología , Regulación de la Expresión Génica/inmunología , Ácidos Láuricos/toxicidad , Receptores X del Hígado , Sistema de Señalización de MAP Quinasas , Ratones , Peso Molecular , Receptores Nucleares Huérfanos/biosíntesis , Receptores Nucleares Huérfanos/genética , Receptores Acoplados a Proteínas G/biosíntesis , Receptores Acoplados a Proteínas G/genética , Bazo/inmunología , Bazo/patología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/inmunología , Células TH1/inmunología , Células Th17/inmunología , Transcriptoma
10.
Gastroenterology ; 160(5): 1521-1531.e9, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33346007

RESUMEN

BACKGROUND & AIMS: Despite the growing recognition of duodenal alterations in the pathophysiology of functional dyspepsia (FD), the effect and mechanism of proton pump inhibitors (PPIs) or first-line therapy remain unclear. We studied duodenal and systemic alterations in relation to PPI therapy in patients with FD and healthy volunteers (HVs). METHODS: We performed a prospective interventional study assessing symptoms (Patient Assessment of Gastrointestinal Symptom Severity Index), duodenal alterations, and systemic factors in patients with FD ("FD-starters") and HVs before and after PPI therapy (pantoprazole 40 mg once daily for 4 weeks). Duodenal mucosal eosinophils, mast cells and permeability were quantified. Luminal pH and bile salts were determined in duodenal aspirates. Procedures were also performed in PPI-refractory patients with FD ("FD-stoppers") before and 8 weeks after PPI withdrawal. Between- and within-group changes from baseline and associations with duodenal or systemic factors were analyzed using linear mixed models. RESULTS: The study was completed by 30 HV, 27 FD-starters, and 18 FD-stoppers. Symptoms and duodenal eosinophils, mast cells (all, P < .0001), and paracellular passage (P = .02) were significantly higher in FD-starters vs HVs and reduced with PPI therapy. Symptoms and duodenal immune cells also decreased in FD-stoppers off PPIs. In contrast, immune cells and permeability increased in HVs on PPIs. Dyspeptic symptoms correlated with eosinophils before and during PPI therapy, and increased eosinophils and permeability in HVs on PPIs were associated with changes in bile salts. CONCLUSIONS: We provide the first prospective evidence for eosinophil-reducing effects as a therapeutic mechanism of PPIs in FD, with differential effects in HVs pointing to a role of luminal changes. ClinicalTrials.gov, Number: NCT03545243.


Asunto(s)
Enfermedades Duodenales/tratamiento farmacológico , Duodeno/efectos de los fármacos , Dispepsia/tratamiento farmacológico , Eosinofilia/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Mucosa Intestinal/efectos de los fármacos , Mastocitos/efectos de los fármacos , Pantoprazol/uso terapéutico , Inhibidores de la Bomba de Protones/uso terapéutico , Adulto , Bélgica , Ácidos y Sales Biliares/metabolismo , Estudios de Casos y Controles , Enfermedades Duodenales/diagnóstico , Enfermedades Duodenales/inmunología , Enfermedades Duodenales/metabolismo , Duodeno/inmunología , Duodeno/metabolismo , Dispepsia/diagnóstico , Dispepsia/inmunología , Dispepsia/metabolismo , Eosinofilia/diagnóstico , Eosinofilia/inmunología , Eosinofilia/metabolismo , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Enfermedades Inflamatorias del Intestino/diagnóstico , Enfermedades Inflamatorias del Intestino/inmunología , Enfermedades Inflamatorias del Intestino/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Masculino , Mastocitos/inmunología , Mastocitos/metabolismo , Pantoprazol/efectos adversos , Permeabilidad , Estudios Prospectivos , Inhibidores de la Bomba de Protones/efectos adversos , Factores de Tiempo , Resultado del Tratamiento
11.
Clin Exp Allergy ; 52(2): 324-333, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34570395

RESUMEN

BACKGROUND: Deaths attributed to Coronavirus Disease 2019 (COVID-19) are mainly due to severe hypoxemic respiratory failure. Although the inflammatory storm has been considered the main pathogenesis of severe COVID-19, hypersensitivity may be another important mechanism involved in severe cases, which have a perfect response to corticosteroids (CS). METHOD: We detected the serum level of anti-SARS-CoV-2-spike S1 protein-specific IgE (SP-IgE) and anti-SARS-CoV-2 nucleocapsid protein-specific IgE (NP-IgE) in COVID-19. Correlation of levels of specific IgE and clinical severity were analysed. Pulmonary function test and bronchial provocation test were conducted in early convalescence of COVID-19. We also obtained histological samples via endoscopy to detect the evidence of mast cell activation. RESULT: The levels of serum SP-IgE and NP-IgE were significantly higher in severe cases, and were correlated with the total lung severity scores (TLSS) and the PaO2 /FiO2 ratio. Nucleocapsid protein could be detected in both airway and intestinal tissues, which was stained positive together with activated mast cells, binded with IgE. Airway hyperresponsiveness (AHR) exists in the early convalescence of COVID-19. After the application of CS in severe COVID-19, SP-IgE and NP-IgE decreased, but maintained at a high level. CONCLUSION: Hypersensitivity may be involved in severe COVID-19.


Asunto(s)
Bronquios/inmunología , COVID-19/inmunología , Proteínas de la Nucleocápside de Coronavirus/inmunología , Duodeno/inmunología , Hipersensibilidad/inmunología , Inmunoglobulina E/inmunología , Mastocitos/inmunología , Glicoproteína de la Espiga del Coronavirus/inmunología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Bronquios/metabolismo , Bronquios/patología , COVID-19/metabolismo , COVID-19/patología , COVID-19/fisiopatología , Estudios de Casos y Controles , Proteínas de la Nucleocápside de Coronavirus/metabolismo , Duodeno/metabolismo , Duodeno/patología , Femenino , Humanos , Hipersensibilidad/metabolismo , Hipersensibilidad/patología , Hipersensibilidad/fisiopatología , Pulmón/fisiopatología , Masculino , Mastocitos/metabolismo , Mastocitos/patología , Persona de Mediana Edad , Membrana Mucosa/inmunología , Membrana Mucosa/metabolismo , Membrana Mucosa/patología , Fosfoproteínas/inmunología , Fosfoproteínas/metabolismo , Recuperación de la Función , Hipersensibilidad Respiratoria/fisiopatología , Estudios Retrospectivos , SARS-CoV-2 , Índice de Severidad de la Enfermedad , Glicoproteína de la Espiga del Coronavirus/metabolismo , Adulto Joven
12.
BMC Microbiol ; 22(1): 8, 2022 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-34983374

RESUMEN

BACKGROUND: Type 1 diabetes (T1D) is an autoimmune disease that is increasing in prevalence worldwide. One of the contributing factors to the pathogenesis of T1D is the composition of the intestinal microbiota, as has been demonstrated. in T1D patients, with some studies demonstrating a deficiency in their levels of Prevotella. We have isolated a strain of Prevotella histicola from a duodenal biopsy that has anti-inflammatory properties, and in addition, alters the development of autoimmune diseases in mouse models. Therefore, our hypothesis is that the oral administration of P. histicola might delay the development of T1D in the non-obese diabetic (NOD) mice. To assess this, we used the following materials and methods. Female NOD mice (ages 5-8 weeks) were administered every other day P. histicola that was cultured in-house. Blood glucose levels were measured every other week. Mice were sacrificed at various time points for histopathological analysis of the pancreas. Modulation of immune response by the commensal was tested by analyzing regulatory T-cells and NKp46+ cells using flow cytometry and intestinal cytokine mRNA transcript levels using quantitative RT-PCR. For microbial composition, 16 s rRNA gene analysis was conducted on stool samples collected at various time points. RESULTS: Administration of P. histicola in NOD mice delayed the onset of T1D. Beta diversity in the fecal microbiomes demonstrated that the microbial composition of the mice administered P. histicola was different from those that were not treated. Treatment with P. histicola led to a significant increase in regulatory T cells with a concomitant decrease in NKp46+ cells in the pancreatic lymph nodes as compared to the untreated group after 5 weeks of treatment. CONCLUSIONS: These observations suggest that P. histicola treatment delayed onset of diabetes by increasing the levels of regulatory T cells in the pancreatic lymph nodes. This preliminary work supports the rationale that enteral exposure to a non pathogenic commensal P. histicola be tested as a future therapy for T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/dietoterapia , Microbioma Gastrointestinal/fisiología , Prevotella/fisiología , Probióticos/administración & dosificación , Animales , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Citocinas/genética , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/microbiología , Duodeno/inmunología , Duodeno/microbiología , Heces/microbiología , Femenino , Humanos , Ratones , Ratones Endogámicos NOD , Páncreas/inmunología , Páncreas/patología
13.
Proc Natl Acad Sci U S A ; 116(30): 15134-15139, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31285344

RESUMEN

B cells play important roles in autoimmune diseases through autoantibody production, cytokine secretion, or antigen presentation to T cells. In most cases, the contribution of B cells as antigen-presenting cells is not well understood. We have studied the autoantibody response against the enzyme transglutaminase 2 (TG2) in celiac disease patients by generating recombinant antibodies from single gut plasma cells reactive with discrete antigen domains and by undertaking proteomic analysis of anti-TG2 serum antibodies. The majority of the cells recognized epitopes in the N-terminal domain of TG2. Antibodies recognizing C-terminal epitopes interfered with TG2 cross-linking activity, and B cells specific for C-terminal epitopes were inefficient at taking up TG2-gluten complexes for presentation to gluten-specific T cells. The bias toward N-terminal epitopes hence reflects efficient T-B collaboration. Production of antibodies against N-terminal epitopes coincided with clinical onset of disease, suggesting that TG2-reactive B cells with certain epitope specificities could be the main antigen-presenting cells for pathogenic, gluten-specific T cells. The link between B cell epitopes, antigen presentation, and disease onset provides insight into the pathogenic mechanisms of a T cell-mediated autoimmune condition.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos B/inmunología , Enfermedad Celíaca/inmunología , Epítopos de Linfocito B/inmunología , Proteínas de Unión al GTP/inmunología , Linfocitos T/inmunología , Transglutaminasas/inmunología , Edad de Inicio , Células Presentadoras de Antígenos/patología , Autoanticuerpos/biosíntesis , Autoanticuerpos/genética , Autoantígenos/genética , Autoantígenos/inmunología , Linfocitos B/patología , Enfermedad Celíaca/genética , Enfermedad Celíaca/patología , Duodeno/inmunología , Duodeno/patología , Epítopos de Linfocito B/química , Epítopos de Linfocito B/genética , Proteínas de Unión al GTP/química , Proteínas de Unión al GTP/genética , Glútenes/química , Glútenes/inmunología , Humanos , Sueros Inmunes/química , Cadenas Ligeras de Inmunoglobulina/biosíntesis , Cadenas Ligeras de Inmunoglobulina/genética , Modelos Moleculares , Unión Proteica , Conformación Proteica , Proteína Glutamina Gamma Glutamiltransferasa 2 , Linfocitos T/patología , Transglutaminasas/química , Transglutaminasas/genética
14.
Int J Mol Sci ; 22(3)2021 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525473

RESUMEN

Celiac disease (CD) is an autoimmune enteropathy arising in genetically predisposed subjects exposed to gluten, which activates both innate and adaptive immunity. Although the pathogenesis is common to all patients, the clinical spectrum is quite variable, and differences could be explained by gene expression variations. Among the factors able to affect gene expression, there are lncRNAs. We evaluated the expression profile of 87 lncRNAs in CD vs. healthy control (HC) intestinal biopsies by RT-qPCR array. Nuclear enriched abundant transcript 1 (NEAT1) and taurine upregulated gene 1 (TUG1) were detected as downregulated in CD patients at diagnosis, but their expression increased in biopsies of patients on a gluten-free diet (GFD) exposed to gluten. The increase in NEAT1 expression after gluten exposure was mediated by IL-15 and STAT3 activation and binding to the NEAT1 promoter, as demonstrated by gel shift assay. NEAT1 is localized in the nucleus and can regulate gene expression by sequestering transcription factors, and it has been implicated in immune regulation and control of cell proliferation. The demonstration of its regulation by gluten thus also supports the role of lncRNAs in CD and prompts further research on these RNAs as gene expression regulators.


Asunto(s)
Enfermedad Celíaca/genética , Regulación hacia Abajo , Duodeno/química , Gliadina/efectos adversos , ARN Largo no Codificante/genética , Adulto , Estudios de Casos y Controles , Enfermedad Celíaca/inmunología , Proliferación Celular , Células Cultivadas , Niño , Regulación hacia Abajo/efectos de los fármacos , Duodeno/inmunología , Femenino , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Interleucina-15/genética , Mucosa Intestinal/química , Mucosa Intestinal/inmunología , Masculino , Factor de Transcripción STAT3/genética
15.
Bull Exp Biol Med ; 171(3): 352-356, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34297293

RESUMEN

To better explore the pathophysiology of FA and its therapy, we aimed to establish a simple and practicable FA model with Freund's adjuvant and introduce an easy and reliable laboratory evaluation method for assessment of inflammation in intestinal segments at different anatomical locations. BALB/c mice were sensitized with ovalbumin combined with Freund's adjuvant. Complete Freund's adjuvant was chosen for the first sensitization and two weeks later incomplete Freund's adjuvant was used for a second sensitization. Two weeks later, the sensitized mice were challenged with 50 mg ovalbumin every other day. After the 6 challenge, all mice were assessed for systemic anaphylaxis, and then sacrificed for sample collection. All sensitized mice showed anaphylactic symptoms and markedly increased levels of serum ovalbumin-specific IgE and IgG1. The activity of mast cell protease-1 (mMCPT-1) was significantly increased in the serum and interstitial fluid of the duodenum, jejunum, ileum, and colon. A successful FA model was established, of which inflammation occurred in the duodenum, jejunum, ileum, and colon. This model provides a reliable and simple tool for analysis of the mechanism of FA and methods of immunotherapy. Moreover, combined detection of ovalbumin-specific antibody and local mMCPT-1 levels could potentially be used as the major indicator for assessment of food allergy.


Asunto(s)
Anafilaxia/inmunología , Quimasas/genética , Hipersensibilidad al Huevo/inmunología , Adyuvante de Freund/administración & dosificación , Inmunoglobulina E/sangre , Inmunoglobulina G/sangre , Ovalbúmina/administración & dosificación , Anafilaxia/inducido químicamente , Anafilaxia/genética , Anafilaxia/patología , Animales , Biomarcadores/metabolismo , Quimasas/inmunología , Colon/inmunología , Colon/patología , Duodeno/inmunología , Duodeno/patología , Hipersensibilidad al Huevo/genética , Hipersensibilidad al Huevo/patología , Líquido Extracelular/química , Líquido Extracelular/inmunología , Femenino , Expresión Génica , Íleon/inmunología , Íleon/patología , Yeyuno/inmunología , Yeyuno/patología , Ratones , Ratones Endogámicos BALB C , Ovalbúmina/inmunología
16.
Gut ; 69(3): 591-600, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31784469

RESUMEN

Emerging data increasingly point towards the duodenum as a key region underlying the pathophysiology of functional dyspepsia (FD), one of the most prevalent functional GI disorders. The duodenum plays a major role in the control and coordination of gastroduodenal function. Impaired duodenal mucosal integrity and low-grade inflammation have been associated with altered neuronal signalling and systemic immune activation, and these alterations may ultimately lead to dyspeptic symptoms. Likely luminal candidates inducing the duodenal barrier defect include acid, bile, the microbiota and food antigens although no causal association with symptoms has been convincingly demonstrated. Recognition of duodenal pathology in FD will hopefully lead to the discovery of new biomarkers and therapeutic targets, allowing biologically targeted rather than symptom-based therapy. In this review, we summarise the recent advances in the diagnosis and treatment of FD with a focus on the duodenum.


Asunto(s)
Duodeno/fisiopatología , Dispepsia/tratamiento farmacológico , Dispepsia/etiología , Antibacterianos/uso terapéutico , Ácidos y Sales Biliares/metabolismo , Encéfalo/fisiopatología , Duodeno/inmunología , Duodeno/metabolismo , Duodeno/microbiología , Disbiosis/tratamiento farmacológico , Disbiosis/fisiopatología , Dispepsia/diagnóstico , Dispepsia/fisiopatología , Vaciamiento Gástrico , Humanos , Neurotransmisores/uso terapéutico , Probióticos/uso terapéutico , Inhibidores de la Bomba de Protones/uso terapéutico
17.
Infect Immun ; 88(9)2020 08 19.
Artículo en Inglés | MEDLINE | ID: mdl-32631917

RESUMEN

Symptomatic and asymptomatic infection with the diarrheal pathogen enteroaggregative Escherichia coli (EAEC) is associated with growth faltering in children in developing settings. The mechanism of this association is unknown, emphasizing a need for better understanding of the interactions between EAEC and the human gastrointestinal mucosa. In this study, we investigated the role of the aggregative adherence fimbriae II (AAF/II) in EAEC adherence and pathogenesis using human colonoids and duodenal enteroids. We found that a null mutant in aafA, the major subunit of AAF/II, adhered significantly less than wild-type (WT) EAEC strain 042, and adherence was restored in a complemented strain. Immunofluorescence confocal microscopy of differentiated colonoids, which produce an intact mucus layer comprised of the secreted mucin MUC2, revealed bacteria at the epithelial surface and within the MUC2 layer. The WT strain adhered to the epithelial surface, whereas the aafA deletion strain remained within the MUC2 layer, suggesting that the presence or absence of AAF/II determines both the abundance and location of EAEC adherence. In order to determine the consequences of EAEC adherence on epithelial barrier integrity, colonoid monolayers were exposed to EAEC constructs expressing or lacking aafA Colonoids infected with WT EAEC had significantly decreased epithelial resistance, an effect that required AAF/II, suggesting that binding of EAEC to the epithelium is necessary to impair barrier function. In summary, we show that production of AAF/II is critical for adherence and barrier disruption in human colonoids, suggesting a role for this virulence factor in EAEC colonization of the gastrointestinal mucosa.


Asunto(s)
Adhesinas de Escherichia coli/inmunología , Células Epiteliales/microbiología , Escherichia coli/inmunología , Fimbrias Bacterianas/inmunología , Interacciones Microbiota-Huesped/inmunología , Organoides/microbiología , Adhesinas de Escherichia coli/genética , Adhesión Bacteriana , Colon/inmunología , Colon/metabolismo , Colon/microbiología , Recuento de Colonia Microbiana , Duodeno/inmunología , Duodeno/metabolismo , Duodeno/microbiología , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Escherichia coli/genética , Escherichia coli/patogenicidad , Infecciones por Escherichia coli/inmunología , Infecciones por Escherichia coli/microbiología , Infecciones por Escherichia coli/patología , Fimbrias Bacterianas/genética , Eliminación de Gen , Regulación de la Expresión Génica , Prueba de Complementación Genética , Interacciones Microbiota-Huesped/genética , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucina 2/genética , Mucina 2/inmunología , Organoides/inmunología , Organoides/metabolismo , Transducción de Señal
18.
Am J Physiol Gastrointest Liver Physiol ; 318(5): G966-G979, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32308038

RESUMEN

Iron overload induces intestinal-permeability defect (gut leakage), and gut translocation of organismal molecules might enhance systemic inflammation and sepsis severity in patients with thalassemia (Thal). Hence, iron administration in Hbbth3/+ mice, heterozygous ß-globin-deficient Thal mice, was explored. Oral iron administration induced more severe secondary hemochromatosis and gut leakage in Thal mice compared with wild-type (WT) mice. Gut leakage was determined by 1) FITC-dextran assay, 2) spontaneous serum elevation of endotoxin (LPS) and (1→3)-ß-d-glucan (BG), molecular structures of gut-organisms, and 3) reduction of tight-junction molecules with increased enterocyte apoptosis (activated caspase-3) by immunofluorescent staining. Iron overload also enhanced serum cytokines and increased Bacteroides spp. (gram-negative bacteria) in feces as analyzed by microbiome analysis. LPS injection in iron-overloaded Thal mice produced higher mortality and prominent cytokine responses. Additionally, stimulation with LPS plus iron in macrophage from Thal mice induced higher cytokines production with lower ß-globin gene expression compared with WT. Furthermore, possible gut leakage as determined by elevated LPS or BG (>60 pg/mL) in serum without systemic infection was demonstrated in 18 out of 41 patients with ß-thalassemia major. Finally, enhanced LPS-induced cytokine responses of mononuclear cells from these patients compared with cells from healthy volunteers were demonstrated. In conclusion, oral iron administration in Thal mice induced more severe gut leakage and increased fecal gram-negative bacteria, resulting in higher levels of endotoxemia and serum inflammatory cytokines compared with WT. Preexisting hyperinflammatory cytokines in iron-overloaded Thal enhanced susceptibility toward infection.NEW & NOTEWORTHY Although the impact of iron accumulation in several organs of patients with thalassemia is well known, the adverse effect of iron accumulation in gut is not frequently mentioned. Here, we demonstrated iron-induced gut-permeability defect, impact of organismal molecules from gut translocation of, and macrophage functional defect upon the increased sepsis susceptibility in thalassemia mice.


Asunto(s)
Citocinas/metabolismo , Duodeno/metabolismo , Microbioma Gastrointestinal , Hemocromatosis/metabolismo , Mediadores de Inflamación/metabolismo , Hierro/metabolismo , Macrófagos/metabolismo , Sepsis/metabolismo , Talasemia beta/metabolismo , Adulto , Animales , Estudios de Casos y Controles , Células Cultivadas , Modelos Animales de Enfermedad , Duodeno/inmunología , Duodeno/microbiología , Femenino , Sacarato de Óxido Férrico , Hemocromatosis/inducido químicamente , Hemocromatosis/inmunología , Hemocromatosis/microbiología , Heterocigoto , Humanos , Lipopolisacáridos , Macrófagos/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Permeabilidad , Sepsis/inducido químicamente , Sepsis/inmunología , Sepsis/microbiología , Adulto Joven , Globinas beta/genética , Talasemia beta/genética , Talasemia beta/inmunología , Talasemia beta/microbiología
19.
Gastroenterology ; 156(5): 1428-1439.e10, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30593798

RESUMEN

BACKGROUND & AIMS: Development of celiac disease is believed to involve the transglutaminase-dependent response of CD4+ T cells toward deamidated gluten peptides in the intestinal mucosa of individuals with specific HLA-DQ haplotypes. We investigated the antigen presentation process during this mucosal immune response. METHODS: We generated monoclonal antibodies (mAbs) specific for the peptide-MHC (pMHC) complex of HLA-DQ2.5 and the immunodominant gluten epitope DQ2.5-glia-α1a using phage display. We used these mAbs to assess gluten peptide presentation and phenotypes of presenting cells by flow cytometry and enzyme-linked immune absorbent spot (ELISPOT) in freshly prepared single-cell suspensions from intestinal biopsies from 40 patients with celiac disease (35 untreated and 5 on a gluten-free diet) as well as 18 subjects with confirmed noninflamed gut mucosa (controls, 12 presumed healthy, 5 undergoing pancreatoduodenectomy, and 1 with potential celiac disease). RESULTS: Using the mAbs, we detected MHC complexes on cells from intestinal biopsies from patients with celiac disease who consume gluten, but not from patients on gluten-free diets. We found B cells and plasma cells to be the most abundant cells that present DQ2.5-glia-α1a in the inflamed mucosa. We identified a subset of plasma cells that expresses B-cell receptors (BCR) specific for gluten peptides or the autoantigen transglutaminase 2 (TG2). Expression of MHC class II (MHCII) was not restricted to these specific plasma cells in patients with celiac disease but was observed in an average 30% of gut plasma cells from patients and controls. CONCLUSIONS: A population of plasma cells from intestinal biopsies of patients with celiac disease express MHCII; this is the most abundant cell type presenting the immunodominant gluten peptide DQ2.5-glia-α1a in the tissues from these patients. These results indicate that plasma cells in the gut can function as antigen-presenting cells and might promote and maintain intestinal inflammation in patients with celiac disease or other inflammatory disorders.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Enfermedad Celíaca/inmunología , Duodeno/inmunología , Glútenes/inmunología , Antígenos HLA-DQ/inmunología , Inmunidad Mucosa , Epítopos Inmunodominantes , Mucosa Intestinal/inmunología , Fragmentos de Péptidos/inmunología , Células Plasmáticas/inmunología , Animales , Células Presentadoras de Antígenos/metabolismo , Estudios de Casos y Controles , Enfermedad Celíaca/diagnóstico , Enfermedad Celíaca/dietoterapia , Enfermedad Celíaca/metabolismo , Línea Celular , Dieta Sin Gluten , Duodeno/metabolismo , Duodeno/patología , Proteínas de Unión al GTP/inmunología , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Fenotipo , Células Plasmáticas/metabolismo , Proteína Glutamina Gamma Glutamiltransferasa 2 , Transglutaminasas/inmunología
20.
Int J Obes (Lond) ; 44(2): 340-352, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31554917

RESUMEN

OBJECTIVE: The intestinal immune response could play an important role in obesity-related comorbidities. We aim to study the profile of duodenal cytokines and chemokines in patients with morbid obesity (MO), its relation with insulin resistance (IR) and the intake of metformin, and with the evolution of MO after sleeve gastrectomy (SG). RESEARCH DESIGN AND METHODS: Duodenal levels of 24 cytokines and 9 chemokines were analyzed in 14 nonobese and in 54 MO who underwent SG: with lower IR (MO-lower-IR), with higher IR (MO-higher-IR), and with type 2 diabetes treated with metformin (MO-metf-T2DM). RESULTS: MO-lower-IR had higher levels of cytokines related to Th1, Th2, Th9, Th17, Th22, M1 macrophages, and chemokines involved in the recruitment of macrophages and T-lymphocytes (p < 0.05), and total (CD68 expression) and M1 macrophages (ITGAX expression) (p < 0.05) when compared with nonobese patients, but with a decrease in M2 macrophages (MRC1 expression) (p < 0.05). In MO-higher-IR, these chemokines and cytokines decreased and were similar to those found in nonobese patients. In MO-metf-T2DM, only IL-4 (Th2) and IL-22 (Th22) increased their levels with regard to MO-higher-IR (p < 0.05). In MO-higher-IR and MO-metf-T2DM, there was a decrease of CD68 expression (p < 0.05) while ITGAX and MRC1 were similar with regard to MO-lower-IR. We found an association between CXCL8, TNFß and IL-2 with the evolution of body mass index (BMI) after SG (p < 0.05). CONCLUSIONS: There is an association between a higher IR and a lower duodenal immune response in MO, with a slight increase in those patients with metformin treatment. Intestinal immune response could be involved in the evolution of BMI after SG.


Asunto(s)
Duodeno , Resistencia a la Insulina , Obesidad Mórbida , Adulto , Citocinas/análisis , Diabetes Mellitus Tipo 2/complicaciones , Diabetes Mellitus Tipo 2/tratamiento farmacológico , Duodeno/química , Duodeno/citología , Duodeno/inmunología , Femenino , Humanos , Hipoglucemiantes/uso terapéutico , Masculino , Metformina/uso terapéutico , Persona de Mediana Edad , Obesidad Mórbida/complicaciones , Obesidad Mórbida/epidemiología , Obesidad Mórbida/inmunología , Obesidad Mórbida/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA