Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Am J Gastroenterol ; 119(6): 1167-1176, 2024 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-38235740

RESUMEN

INTRODUCTION: There are limited data characterizing eating habits among pediatric patients with eosinophilic esophagitis (EoE). We compared eating behaviors in pediatric patients with EoE with healthy controls and assessed the degree of correlation with symptomatology, endoscopic and histologic findings, and esophageal distensibility. METHODS: We conducted a prospective, observational study where subjects consumed 4 food textures (puree, soft solid, chewable, and hard solid) and were scored for eating behaviors including number of chews per bite, sips of fluid per food, and consumption time. Symptomatic, endoscopic, histologic, and esophageal distensibility data were collected for case subjects. RESULTS: Twenty-seven case subjects and 25 healthy controls were enrolled in our study (mean age 11.0 years, 63.5% male). Compared with healthy controls, pediatric patients with EoE demonstrated more chews per bite with soft solid (13.6 vs 9.1, P = 0.031), chewable (14.7 vs 10.7, P = 0.047), and hard solid foods (19.0 vs 12.8, P = 0.037). Patients with EoE also demonstrated increased consumption time with soft solid (94.7 vs 58.3 seconds, P = 0.002), chewable (90.0 vs 65.1 seconds, P = 0.005), and hard solid foods (114.1 vs 76.4 seconds, P = 0.034) when compared with healthy controls. Subgroup analysis based on disease status showed no statistically significant differences in eating behaviors between active and inactive EoE. Total endoscopic reference score positively correlated with consumption time ( r = 0.53, P = 0.008) and number of chews ( r = 0.45, P = 0.027) for chewable foods and with number of chews ( r = 0.44, P = 0.043) for hard solid foods. Increased consumption time correlated with increased eosinophil count ( r = 0.42, P = 0.050) and decreased esophageal distensibility ( r = -0.82, P < 0.0001). DISCUSSION: Altered eating behaviors including increased chewing and increased consumption time can be seen in pediatric patients with EoE, can persist despite histologic remission, and may be driven by changes in esophageal distensibility.


Asunto(s)
Esofagitis Eosinofílica , Esófago , Conducta Alimentaria , Humanos , Esofagitis Eosinofílica/fisiopatología , Esofagitis Eosinofílica/patología , Masculino , Femenino , Estudios Prospectivos , Niño , Conducta Alimentaria/fisiología , Estudios de Casos y Controles , Esófago/patología , Esófago/fisiopatología , Adolescente , Esofagoscopía
2.
Ann Allergy Asthma Immunol ; 128(5): 589-593, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35085819

RESUMEN

BACKGROUND: Dupilumab has been approved to treat atopic dermatitis, asthma, and nasal polyps and is in active clinical trials for the treatment of eosinophilic esophagitis (EoE). Given its shared immunopathology, we hypothesized that EoE symptoms and inflammation would improve when dupilumab therapy was used for other allergic indications. OBJECTIVE: To measure the clinical and histologic response in EoE to dupilumab when treating other atopic diseases. METHODS: We completed a retrospective chart review of all patients at Children's Hospital of Philadelphia and Rady Children Hospital who were prescribed dupilumab for atopic dermatitis, asthma, or nasal polyps and had a concomitant clinical diagnosis of EoE. Demographic information along with histology, symptom scores, medications, and diet information were collected. Response to dupilumab was evaluated. RESULTS: A total of 45 patients were identified. Of which, 11 patients were prescribed dupilumab for asthma, 27 for atopic dermatitis, 3 for nasal polyps, and 4 for compassionate use for EoE. There was no follow-up data for 8 patients. Follow-up histology was available for 26 patients: 22 of 26 had less than 6 eosinophils per high power field after the initiation of dupilumab with significant improvement (pre: 52.9 + 35.1 to post: 4.5 + 10.9 eosinophils/high power field, P < .005). A total of 28 patients had improvement of symptoms, with 24 patients reporting complete resolution of symptoms after dupilumab initiation. Reductions in EoE treatment medications (swallowed steroids, proton pump inhibitors) or expansion of diet occurred in 29 patients treated with dupilumab. CONCLUSION: Dupilumab therapy initiated for atopic disease effectively induces symptomatic and histologic remission of esophageal disease and reduces the need for EoE-directed therapy in patients with concomitant EoE.


Asunto(s)
Asma , Dermatitis Atópica , Esofagitis Eosinofílica , Pólipos Nasales , Anticuerpos Monoclonales Humanizados , Asma/complicaciones , Asma/tratamiento farmacológico , Niño , Ensayos de Uso Compasivo , Dermatitis Atópica/complicaciones , Dermatitis Atópica/tratamiento farmacológico , Esofagitis Eosinofílica/diagnóstico , Humanos , Pólipos Nasales/complicaciones , Estudios Retrospectivos
3.
Ital J Pediatr ; 47(1): 230, 2021 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-34809686

RESUMEN

Eosinophilic esophagitis (EoE) is a chronic clinical-pathologic disease characterized by eosinophilic infiltration of the esophageal epithelium with esophageal dysfunction symptoms.EoE can occur at any age and has different clinical manifestations depending on the age onset.To date, esophago-gastroduodenal endoscopy (EGD) with biopsy is the gold-standard for EoE diagnosis.According to the recent consensus guidelines, proton pump inhibitors, corticosteroids and elimination diets could be a first-line therapy option. The aim of the treatment is clinical and histological remission for preventing long-lasting untreatable fibrosis.A multidisciplinary approach (allergist, gastroenterology, dietitian, and pathologist) is recommended for managing patients affected by EoE, given the complexity of its treatment.This review will provide a practical guide to assist pediatricians treating children with EoE.Moreover, it highlights the unmet needs in diagnosis and treatment that require urgent attention from the scientific community in the aim of improving the management of patients with EoE.


Asunto(s)
Esofagitis Eosinofílica , Algoritmos , Dieta , Dilatación , Exposición a Riesgos Ambientales , Esofagitis Eosinofílica/diagnóstico , Esofagitis Eosinofílica/etiología , Esofagitis Eosinofílica/terapia , Esofagoscopía , Predisposición Genética a la Enfermedad , Glucocorticoides/uso terapéutico , Humanos , Pediatría , Inhibidores de la Bomba de Protones/uso terapéutico , Factores de Riesgo
4.
Clin Transl Immunology ; 10(7): e1314, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34322233

RESUMEN

OBJECTIVE: There are no disease-modifying therapies for the treatment of eosinophilic oesophagitis (EoE), which is driven by non-IgE-mediated allergic inflammation. A recent clinical trial of milk epicutaneous immunotherapy (EPIT) has shown initial promise, with 47% of treated EoE patients tolerating milk without recurrence of disease. Mechanisms of EPIT in EoE have not been studied in humans. Here, we identify transcriptional changes in the peripheral CD4+ T-cell compartment during active EoE and following EPIT. METHODS: RNA isolation, sequencing and integrative data analysis were performed on peripheral CD4+ T cells isolated from 15 of 20 patients enrolled in a clinical trial of EPIT for EoE. Gene expression changes in peripheral CD4+ T cells were examined during diet therapy and following trial of milk antigen EPIT. RESULTS: We identify 244 differentially expressed genes in peripheral blood CD4+ cells of EoE patients consuming versus those eliminating milk, and 129 DEGs in CD4+ cells were isolated after EPIT versus after placebo (FDR ≤ 0.05). Gene set enrichment analysis identifies enrichment of hallmark interferon-α and interferon-γ response pathways in peripheral CD4+ T cells from EoE patients during active disease on a milk-containing diet. We demonstrate overlap of this gene signature with the altered gene expression signature seen in EoE patient biopsy tissue. EPIT therapy response is associated with significant enrichment in pathways related to T-cell receptor signalling (P = 1.16 × 10-14), antigen presentation and costimulation, and cytokine signalling (P = 1.11 × 10-16), as well as upregulation of genes associated with regulatory T-cell function. CONCLUSIONS: EoE is associated with distinct global transcriptional changes in CD4+ T cells, one feature of which is an IFN response signature. Clinically favorable response to EPIT is likely multifactorial but is associated with a distinct transcriptional profile in peripheral CD4+ cells supporting the hypothesis that EPIT alters peripheral CD4+ responses in EoE patients.

5.
J Allergy Clin Immunol ; 144(1): 171-182, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30578874

RESUMEN

BACKGROUND: Fibrosis and stricture are major comorbidities in patients with eosinophilic esophagitis (EoE). Lysyl oxidase (LOX), a collagen cross-linking enzyme, has not been investigated in the context of EoE. OBJECTIVE: We investigated regulation of epithelial LOX expression as a novel biomarker and functional effector of fibrostenotic disease conditions associated with EoE. METHODS: LOX expression was analyzed by using RNA-sequencing, PCR assays, and immunostaining in patients with EoE; cytokine-stimulated esophageal 3-dimensional organoids; and fibroblast-epithelial cell coculture, the latter coupled with fluorescence-activated cell sorting. RESULTS: Gene ontology and pathway analyses linked TNF-α and LOX expression in patients with EoE, which was validated in independent sets of patients with fibrostenotic conditions. TNF-α-mediated epithelial LOX upregulation was recapitulated in 3-dimensional organoids and coculture experiments. We find that fibroblast-derived TNF-α stimulates epithelial LOX expression through activation of nuclear factor κB and TGF-ß-mediated signaling. In patients receiver operating characteristic analyses suggested that LOX upregulation indicates disease complications and fibrostenotic conditions in patients with EoE. CONCLUSIONS: There is a novel positive feedback mechanism in epithelial LOX induction through fibroblast-derived TNF-α secretion. Esophageal epithelial LOX might have a role in the development of fibrosis with substantial translational implications.


Asunto(s)
Biomarcadores/metabolismo , Esofagitis Eosinofílica/genética , Células Epiteliales/fisiología , Esófago/patología , Fibroblastos/fisiología , Proteína-Lisina 6-Oxidasa/genética , Factor de Necrosis Tumoral alfa/metabolismo , Adolescente , Adulto , Anciano , Células Cultivadas , Niño , Preescolar , Técnicas de Cocultivo , Constricción Patológica , Esofagitis Eosinofílica/diagnóstico , Femenino , Fibrosis , Ontología de Genes , Humanos , Lactante , Masculino , Persona de Mediana Edad , Proteína-Lisina 6-Oxidasa/metabolismo , Regulación hacia Arriba , Adulto Joven
6.
Ann Allergy Asthma Immunol ; 121(6): 711-716, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30194971

RESUMEN

BACKGROUND: Previous reports suggest a higher prevalence of comorbid diseases in patients with eosinophilic esophagitis (EoE), although few have systematically quantified comorbidities in pediatric patients. OBJECTIVE: To define the rate of comorbid diagnoses in pediatric EoE patients compared with rates in those without EoE. METHODS: Retrospective cross-sectional review of electronic medical records for patients seen in a single large pediatric primary care network between January 2007 and December 2016 (n = 456,148). International Classification of Diseases, Ninth and Tenth Revision codes were used to determine prevalence rates for coexisting diagnoses. RESULTS: A total of 428 patients held a diagnosis for EoE. Significant differences in rate of comorbid diseases included allergic rhinoconjunctivitis (60.0% of EoE cohort vs 17.4% of non-EoE cohort, P < .0001); asthma (59.8% of EoE, 21.4% of non-EoE, P < .0001); atopic dermatitis (17.8% of EoE, 6.6% of non-EoE, P < .0001); adrenal insufficiency (2.6% of EoE, 0.4% of non-EoE, P < .0001); autism spectrum disorder (7.5% of EoE, 1.9% of non-EoE, P < .0001); celiac disease (5.6% of EoE, 0.9% of non-EoE, P < .0001); connective tissue diseases (1.4% of EoE, 0.1% of non-EoE, P < .0001); cystic fibrosis (0.9% of EoE, 0.05% of non-EoE, P < .0001); inflammatory bowel disease (0.7% of EoE, 0.2% of non-EoE, P = .03); type 1 diabetes mellitus (1.2% of EoE, 0.3% of non-EoE, P = .0069). CONCLUSION: Children with EoE have markedly higher rates of both atopic and non-atopic diseases compared with children without EoE. These associations have important implications for comprehensive EoE care and future research regarding associated disease mechanisms.


Asunto(s)
Comorbilidad , Esofagitis Eosinofílica/diagnóstico , Adolescente , Insuficiencia Suprarrenal/epidemiología , Adulto , Asma/epidemiología , Trastorno del Espectro Autista/epidemiología , Niño , Preescolar , Enfermedades del Tejido Conjuntivo/epidemiología , Estudios Transversales , Fibrosis Quística/epidemiología , Dermatitis Atópica/epidemiología , Diabetes Mellitus Tipo 1/epidemiología , Femenino , Humanos , Lactante , Enfermedades Inflamatorias del Intestino/epidemiología , Masculino , Prevalencia , Estudios Retrospectivos , Adulto Joven
8.
Ann Allergy Asthma Immunol ; 120(2): 177-183.e2, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29289462

RESUMEN

BACKGROUND: Eosinophilic esophagitis (EoE) is an allergic inflammatory disease that is triggered by food allergens and characterized by progressive esophageal dysfunction. Esophageal biopsy specimens are characterized by eosinophilia and expression of TH2 cytokines. OBJECTIVE: To ascertain whether TH2 cells can exist in the peripheral blood in patients with milk-induced EoE. METHODS: Peripheral blood mononuclear cells from 20 children with milk-induced EoE were collected during active EoE (EoE-A) while consuming milk and inactive EoE (EoE-I) while not consuming milk, and 8 healthy patients without EoE were used as controls. The samples were analyzed for T-cell phenotype, including intracellular cytokines before and after incubation with milk antigens and assessed by flow cytometry. RESULTS: We found a significant increase in CD4+ TH2 cells in the peripheral blood of patients with EoE-A compared with the controls. Furthermore, we observed a significant mean (SD) increase in the activation marker of CD154+ T cells (0.17% [0.047%]) in patients with EoE-A compared with control patients (0.034% [0.007%]) and EoE-I (0.025% [0.008]). These CD4+ T cells expressed significantly increase levels of TH2 cytokines (interleukins 4, 5, and 13) compared with the EoE-I and control groups. CD3+CD4+CD154+IL-5+ cells were significantly increased by milk antigens in both milk-induced EoE-A (0.050% [0.008%] to 0.079% [0.017%]) and EoE-I (0.0045% [0.002%] to 0.014% [0.008%]) compared with the controls (0.008% [0.003%] to 0.003% [0.001%]). CONCLUSION: Our findings indicate that in EoE peripheral T cells have specific activation to milk allergens.


Asunto(s)
Esofagitis Eosinofílica/inmunología , Eosinófilos/inmunología , Esófago/patología , Hipersensibilidad a la Leche/inmunología , Células Th2/inmunología , Adolescente , Alérgenos/inmunología , Animales , Biopsia , Bovinos , Niño , Preescolar , Citocinas/metabolismo , Femenino , Humanos , Inmunofenotipificación , Activación de Linfocitos , Masculino , Leche , Proteínas de la Leche/inmunología
10.
J Asthma Allergy ; 9: 13-25, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26889090

RESUMEN

Triticum aestivum (bread wheat) is the most widely grown crop worldwide. In genetically predisposed individuals, wheat can cause specific immune responses. A food allergy to wheat is characterized by T helper type 2 activation which can result in immunoglobulin E (IgE) and non-IgE mediated reactions. IgE mediated reactions are immediate, are characterized by the presence of wheat-specific IgE antibodies, and can be life-threatening. Non-IgE mediated reactions are characterized by chronic eosinophilic and lymphocytic infiltration of the gastrointestinal tract. IgE mediated responses to wheat can be related to wheat ingestion (food allergy) or wheat inhalation (respiratory allergy). A food allergy to wheat is more common in children and can be associated with a severe reaction such as anaphylaxis and wheat-dependent, exercise-induced anaphylaxis. An inhalation induced IgE mediated wheat allergy can cause baker's asthma or rhinitis, which are common occupational diseases in workers who have significant repetitive exposure to wheat flour, such as bakers. Non-IgE mediated food allergy reactions to wheat are mainly eosinophilic esophagitis (EoE) or eosinophilic gastritis (EG), which are both characterized by chronic eosinophilic inflammation. EG is a systemic disease, and is associated with severe inflammation that requires oral steroids to resolve. EoE is a less severe disease, which can lead to complications in feeding intolerance and fibrosis. In both EoE and EG, wheat allergy diagnosis is based on both an elimination diet preceded by a tissue biopsy obtained by esophagogastroduodenoscopy in order to show the effectiveness of the diet. Diagnosis of IgE mediated wheat allergy is based on the medical history, the detection of specific IgE to wheat, and oral food challenges. Currently, the main treatment of a wheat allergy is based on avoidance of wheat altogether. However, in the near future immunotherapy may represent a valid way to treat IgE mediated reactions to wheat.

11.
Clin Rev Allergy Immunol ; 50(2): 159-74, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26194940

RESUMEN

Eosinophilic esophagitis (EoE) is an emerging chronic atopic clinical-pathologic disease with an estimated prevalence of 1/1000 similar to the one of Crohn's diseases. Usually, EoE is firstly suspected due to symptoms that are caused by esophageal dysfunction and/or fibrosis. EoE diagnosis is confirmed if the esophageal biopsy shows at least 15 eosinophils per high power field (eos/hpf) as a peak value in one or more of at least four specimens obtained randomly from the esophagus. Most of the patients affected by EoE have other atopic diseases such as allergic rhinitis, asthma, IgE-mediated food allergies, and/or atopic dermatitis. The local inflammation is a T helper type 2 (Th2) flogosis, which most likely is driven by a mixed IgE and non-IgE-mediated reaction to food and/or environmental allergens. Recently published genetic studies showed also that EoE is associated with single nucleotide polymorphism (SNP) on genes which are important in atopic inflammation such as thymic stromal lymphopoietin (TSLP) located close to the Th2 cytokine cluster (IL-4, IL-5, IL-13) on chromosome 5q22. When the EoE diagnosis is made, it is imperative to control the local eosinophilic inflammation not only to give symptomatic relief to the patient but also to prevent complications such as esophageal stricture and food impaction. EoE is treated like many other atopic diseases with a combination of topical steroids and/or food antigen avoidance.


Asunto(s)
Esofagitis Eosinofílica/diagnóstico , Esofagitis Eosinofílica/etiología , Esofagitis Eosinofílica/terapia , Terapia Combinada , Manejo de la Enfermedad , Esofagitis Eosinofílica/epidemiología , Epigénesis Genética , Predisposición Genética a la Enfermedad , Humanos , Factores de Riesgo , Resultado del Tratamiento
12.
Expert Rev Gastroenterol Hepatol ; 9(12): 1501-10, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26470602

RESUMEN

Eosinophilic esophagitis (EoE) is a chronic allergen-mediated inflammatory disease of the esophagus. This inflammation leads to feeding difficulties, failure to thrive and vomiting in young children, and causes food impaction and esophageal stricture in adolescents and adults. In the 20 years since EoE was first described, we have gained a great deal of knowledge regarding the genetic predisposition of disease, the inflammatory milieu associated with EoE and the long-term complications of chronic inflammation. Herein, we summarize the important breakthroughs in the field including both in vitro and in vivo analysis. We discuss insights that we have gained from large-scale unbiased genetic analysis, a multitude of genetically and chemically altered mouse models of EoE and most importantly, the results of clinical trials of various pharmacologic agents. Understanding these successes and failures may be the key to developing more effective therapeutic strategies.


Asunto(s)
Esofagitis Eosinofílica/genética , Esofagitis Eosinofílica/inmunología , Esófago/patología , Células Th2/inmunología , Animales , Basófilos/inmunología , Calpaína/genética , Microambiente Celular , Citocinas/genética , Citocinas/metabolismo , Disbiosis/complicaciones , Esofagitis Eosinofílica/patología , Fibrosis , Predisposición Genética a la Enfermedad , Humanos , Inmunoglobulina E/metabolismo , Interleucina-13/metabolismo , Interleucina-5/metabolismo , Mastocitos/inmunología , Membrana Mucosa/fisiopatología , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Receptores de Citocinas/metabolismo , Proteínas Represoras/genética , Linfopoyetina del Estroma Tímico
13.
Curr Opin Allergy Clin Immunol ; 15(5): 417-25, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26258919

RESUMEN

PURPOSE OF REVIEW: Eosinophilic Esophagitis (EoE) is an emerging chronic atopic disease. Recent advances in understanding its genetic and molecular biology pathogenesis may lead to a better management of the disease RECENT FINDINGS: EoE is an atopic disease. Most of the patients affected by EoE have other atopic diseases such as allergic rhinitis, asthma, IgE-mediated food allergies and/or atopic dermatitis. The local inflammation is a T helper type 2 (Th2) flogosis, which most likely is driven by a mixed IgE and n-IgE-mediated reaction to food and/or environmental allergens. Epidemiological studies show that EoE is an atopic disease with a strong genetic component. Genetic studies have shown that EoE is associated with single nucleotide polymorphism on genes, which are released by the epithelium and important in atopic inflammation such as thymic stromal lymphopoietin located (TSLP) close to the Th2 cytokine cluster [interleukin (IL)-4, IL-5, IL-13] on chromosome 5q22, Calpain 14, EMSY, and Eotaxin3. When the EoE diagnosis is made, it is imperative to control the local eosinophilic inflammation not only to give symptomatic relief to the patient, but also to prevent complications such as esophageal stricture and food impaction. SUMMARY: EoE is treated like many other atopic diseases with a combination of topical steroids and/or food antigen avoidance. The new understanding of EoE may lead to more specific and definitive treatments of EoE.


Asunto(s)
Esofagitis Eosinofílica/terapia , Epitelio/metabolismo , Inmunoterapia , Células Th2/fisiología , Calpaína/genética , Citocinas/genética , Esofagitis Eosinofílica/genética , Predisposición Genética a la Enfermedad , Humanos , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Polimorfismo Genético , Proteínas Represoras/genética , Esteroides/uso terapéutico , Linfopoyetina del Estroma Tímico
14.
Ann Allergy Asthma Immunol ; 115(3): 224-228.e1, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26235409

RESUMEN

BACKGROUND: Evidence supports a possible link between eosinophilic esophagitis (EoE) and environmental aeroallergens, which can manifest as seasonal exacerbation of esophageal eosinophilia. Few studies have examined this link in pediatric patients with EoE. OBJECTIVE: To identify the proportion of patients with seasonal induced esophageal eosinophilia. METHODS: A retrospective chart review was conducted of all patients diagnosed with EoE at the authors' institution. Demographic data were collected by chart review. Seasonal variation or flare was defined as a change from fewer than to at least 15 eosinophils per high-power field and a minimum of a 2-fold increase in eosinophil count between 2 consecutive biopsy specimens in different seasons without dietary or medication modifications. RESULTS: Of the 1,180 patients with EoE, 160 (14%) were suspected of having aeroallergen-associated triggers by history. Of these 160 patients, 32 (20%) had biopsy examination-confirmed variation of EoE triggered by aeroallergens. Most of these patients were boys (84%), all had a history or examination consistent with allergic rhinitis, and most had a history of asthma (75%). Thirty-two subjects had obvious seasonal variation, 22 of whom also had known food-induced symptoms. CONCLUSION: Children with EoE and allergic rhinitis might have exacerbations in their esophageal eosinophilia during certain seasons depending on the specific aeroallergens to which they are sensitized. Identification of environmental allergens to sensitized patients is important and can guide therapy.


Asunto(s)
Progresión de la Enfermedad , Eosinofilia/inmunología , Esofagitis Eosinofílica/inmunología , Exposición por Inhalación , Rinitis Alérgica/inmunología , Alérgenos/inmunología , Asma/inmunología , Niño , Eosinófilos/citología , Eosinófilos/inmunología , Esófago/inmunología , Femenino , Humanos , Masculino , Estudios Retrospectivos , Estaciones del Año
15.
Curr Allergy Asthma Rep ; 15(9): 58, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26233430

RESUMEN

Eosinophilic gastrointestinal disease (EGID) can be classified as eosinophilic esophagitis (EoE) when the eosinophilia is limited to the esophagus or as eosinophilic gastritis (EG) if it is limited to the gastric tract, eosinophilic colitis (EC) if it is limited to the colon, and eosinophilic gastroenteritis (EGE) if the eosinophilia involves one or more parts of the gastrointestinal tract. EoE is by far the most common EGID. It is a well-defined chronic atopic disease due to a T helper type 2 (Th2) inflammation triggered often by food allergens. EoE diagnosis is done if an esophageal biopsy shows at least 15 eosinophils per high power field (eos/hpf). Globally accepted long-term therapies for EoE are the use of swallowed inhaled steroids or food antigen avoidance. The treatment of EoE is done not only to control symptoms but also to prevent complications such as esophageal stricture and food impaction. EGE cause non-specific gastrointestinal (GI) symptoms and are diagnosed if esophagogastroduodenoscopy (EGD)/colonoscopy show eosinophilia in one or more parts of the GI tract. They are rare diseases with an unclear pathogenesis, and they are poorly defined in terms of diagnostic criteria and treatment. Before initiating treatment of any EGE, it is imperative to conduct a differential diagnosis to exclude other causes of hypereosinophilia with GI localization. EGE are often poorly responsive to therapy and there is no commonly accepted long-term treatment. EG has many characteristics similar to EoE, including the fact that it is often due to a food allergen-driven Th2 inflammation; transcriptome analysis however shows that it is more a systemic disease and has a different gene signature than EoE. EC is a benign form of delayed food allergy in infant and is instead a difficult-to-treat severe inflammatory condition in older children and adults. EC in the latter groups can be a manifestation of drug allergy or autoimmune disease. Overall EGE, EC, and EG are rare and are a diagnosis of exclusion until more common causes of eosinophilia have been excluded.


Asunto(s)
Esofagitis Eosinofílica/inmunología , Gastroenteritis/inmunología , Alérgenos/inmunología , Animales , Esofagitis Eosinofílica/epidemiología , Esofagitis Eosinofílica/genética , Eosinófilos/inmunología , Hipersensibilidad a los Alimentos/inmunología , Gastroenteritis/epidemiología , Gastroenteritis/genética , Predisposición Genética a la Enfermedad , Humanos
16.
Nat Commun ; 5: 5593, 2014 Nov 19.
Artículo en Inglés | MEDLINE | ID: mdl-25407941

RESUMEN

Eosinophilic esophagitis (EoE) is an allergic disorder characterized by infiltration of the oesophagus with eosinophils. We had previously reported association of the TSLP/WDR36 locus with EoE. Here we report genome-wide significant associations at four additional loci; c11orf30 and STAT6, which have been previously associated with both atopic and autoimmune diseases, and two EoE-specific loci, ANKRD27 that regulates the trafficking of melanogenic enzymes to epidermal melanocytes and CAPN14, that encodes a calpain whose expression is highly enriched in the oesophagus. The identification of five EoE loci, not only expands our aetiological understanding of the disease but may also represent new therapeutic targets to treat the most debilitating aspect of EoE, oesophageal inflammation and remodelling.


Asunto(s)
Calpaína/genética , Esofagitis Eosinofílica/genética , Factores de Intercambio de Guanina Nucleótido/genética , Proteínas de Neoplasias/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Factor de Transcripción STAT6/genética , Adulto , Estudios de Casos y Controles , Femenino , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Masculino
17.
Expert Rev Clin Immunol ; 10(11): 1463-74, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25340427

RESUMEN

Thymic stromal lymphopoietin (TSLP) is an epithelial-derived cytokine similar to IL- 7, whose gene is located on chromosome 5q22.1 and it exerts its biological function through the TSLP-Receptor (TSLP-R). TSLP is expressed primarily by epithelial cells at barrier surfaces such as the skin, gut and lung in response to danger signals. Since it was cloned in 1994, there has been accumulating evidence that TSLP is crucial for the maturation of antigen presenting cells and hematopoietic cells. TSLP genetic variants and its dysregulated expression have been linked to atopic diseases such as atopic dermatitis, asthma, allergic rhinitis and eosinophilic esophagitis.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Citocinas/inmunología , Regulación de la Expresión Génica/inmunología , Células Madre Hematopoyéticas/inmunología , Hipersensibilidad/inmunología , Transducción de Señal/inmunología , Animales , Células Presentadoras de Antígenos/patología , Cromosomas Humanos Par 5/genética , Cromosomas Humanos Par 5/inmunología , Citocinas/genética , Regulación de la Expresión Génica/genética , Células Madre Hematopoyéticas/patología , Humanos , Hipersensibilidad/genética , Hipersensibilidad/terapia , Especificidad de Órganos , Receptores de Citocinas/genética , Receptores de Citocinas/inmunología , Transducción de Señal/genética , Linfopoyetina del Estroma Tímico
18.
Immunotherapy ; 6(3): 321-31, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24762076

RESUMEN

Eosinophilic esophagitis (EoE) is a clinical pathologic disease characterized by symptoms of esophageal dysfunction and eosinophilia of the esophagus. When the diagnosis is confirmed, it is important to treat the eosinophilic inflammation not only to control the presenting symptoms, but also to prevent acute and chronic complications. The pathogenesis of EoE is most likely a mixed IgE and non-IgE food-mediated reaction, where Th2 cytokines drive esophageal eosinophilia as in other atopic diseases. Hence, it is not surprising that therapy is based on inflammation control, with steroids (oral or topical) and/or food antigen avoidance. However, these treatment options are not specific, reduce the quality of life of patients and have significant side effects, therefore, there is an ongoing effort to design more specific immunotherapies. In this review, we review standard and immunotherapeutic options for EoE treatment, such as anti-IL-5, anti-TNFα, anti-IgE, anti-CRTH, oral allergy desensitization and environmental immunotherapy.


Asunto(s)
Esofagitis Eosinofílica/terapia , Inmunoterapia/métodos , Corticoesteroides/uso terapéutico , Alérgenos/efectos adversos , Alérgenos/uso terapéutico , Animales , Anticuerpos Antiidiotipos/uso terapéutico , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/uso terapéutico , Especificidad de Anticuerpos , Terapia Combinada , Citocinas/genética , Citocinas/inmunología , Citocinas/fisiología , Desensibilización Inmunológica , Diagnóstico Diferencial , Esofagitis Eosinofílica/diagnóstico , Esofagitis Eosinofílica/dietoterapia , Esofagitis Eosinofílica/tratamiento farmacológico , Esofagitis Eosinofílica/inmunología , Hipersensibilidad a los Alimentos/inmunología , Reflujo Gastroesofágico/complicaciones , Reflujo Gastroesofágico/diagnóstico , Humanos , Inmunoglobulina E/inmunología , Inmunosupresores/uso terapéutico , Interleucina-5/antagonistas & inhibidores , Interleucina-5/fisiología , Ratones , Receptores Inmunológicos/antagonistas & inhibidores , Receptores Inmunológicos/inmunología , Receptores de Prostaglandina/antagonistas & inhibidores , Receptores de Prostaglandina/inmunología , Células Th2/inmunología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Linfopoyetina del Estroma Tímico
19.
Immunity ; 39(6): 1158-70, 2013 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-24332033

RESUMEN

Extramedullary hematopoiesis (EMH) refers to the differentiation of hematopoietic stem cells (HSCs) into effector cells that occurs in compartments outside of the bone marrow. Previous studies linked pattern-recognition receptor (PRR)-expressing HSCs, EMH, and immune responses to microbial stimuli. However, whether EMH operates in broader immune contexts remains unknown. Here, we demonstrate a previously unrecognized role for thymic stromal lymphopoietin (TSLP) in promoting the population expansion of progenitor cells in the periphery and identify that TSLP-elicited progenitors differentiated into effector cells including macrophages, dendritic cells, and granulocytes and that these cells contributed to type 2 cytokine responses. The frequency of circulating progenitor cells was also increased in allergic patients with a gain-of-function polymorphism in TSLP, suggesting the TSLP-EMH pathway might operate in human disease. These data identify that TSLP-induced EMH contributes to the development of allergic inflammation and indicate that EMH is a conserved mechanism of innate immunity.


Asunto(s)
Citocinas/metabolismo , Hematopoyesis Extramedular/inmunología , Hipersensibilidad/inmunología , Inflamación , Bazo/inmunología , Animales , Citocinas/genética , Citocinas/inmunología , Modelos Animales de Enfermedad , Citometría de Flujo , Perfilación de la Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Polimorfismo Genético , Células Precursoras de Linfocitos B/citología , Bazo/citología , Triquinelosis/inmunología , Linfopoyetina del Estroma Tímico
20.
Pediatrics ; 117(2): e336-9, 2006 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-16452340

RESUMEN

A 16-year-old girl developed a cough, hypereosinophilia (absolute eosinophil count: 32000/mm3), hypergammaglobulinemia, and multiple noncavitary pulmonary nodules 1 month after having ingested an earthworm on a dare. Spirometry revealed moderate restriction and reduced gas diffusion. Parabronchial biopsy demonstrated eosinophilic organizing pneumonitis with multiple eosinophilic microabscesses, and Toxocara titers were elevated (>1:4096). Ophthalmologic examination ruled out ocular larva migrans. The patient received a 10-day course of albendazole (400 mg orally twice daily) and demonstrated significant clinical improvement with resolution of cough and pulmonary function abnormalities. Her white blood cell count and hypergammaglobulinemia normalized within 20 days, yet eosinophils (absolute eosinophil count: 1780/mm3) and Toxocara serologies (>1:4096) remained elevated 3 months after completing antihelminthic therapy. In this instance, the ingested earthworm served as the paratenic carrier of Toxocara larvae from the soil to the patient. This case highlights the clinical evolution of pulmonary visceral larva migrans infection caused by Toxocara spp. associated with a discrete ingestion in an adolescent patient. In addition, it provides a rare opportunity to define the incubation period of visceral larva migrans and emphasizes the importance of education regarding sources of Toxocara infection.


Asunto(s)
Vectores de Enfermedades , Larva Migrans Visceral/diagnóstico , Enfermedades Pulmonares Parasitarias/diagnóstico , Oligoquetos , Adolescente , Animales , Ingestión de Alimentos , Femenino , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA