Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 586
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 181(1): 19, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32243789

RESUMO

We asked three researchers how their personal connection to disease has affected them and what lessons it has taught them along the way.


Assuntos
Hiperplasia do Linfonodo Gigante/tratamento farmacológico , Doença Celíaca/tratamento farmacológico , Progéria/terapia , Desenvolvimento de Medicamentos , Reposicionamento de Medicamentos , Humanos
2.
Gastroenterology ; 167(1): 183-193, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38355059

RESUMO

As it appears that we are currently at the cusp of an era in which drugs that are new, re-purposed, or "supplements" will be introduced to the management of celiac disease, we need to reflect on whether the framework is set for celiac disease to be treated increasingly with pharmaceuticals as well as diet. This refers to reflecting on the rigor of current diagnostic practices; the limitations of the current standard of care, which is a gluten-free diet; and that we lack objective markers of disease severity. Investigating these issues will help us to identify gaps in technology and practices that could be critical for selecting patients with a well-defined need for an improved or alternative treatment. Both aspects, circumscribed limitations of the gluten-free diet and diagnostics helping to define celiac disease target groups, together with the guiding requirements by the responsible regulatory authorities, will contribute to defining the subgroups of patients with confirmed celiac disease eligible for distinct pharmacologic strategies. Because many patients with celiac disease are diagnosed in childhood, these aspects need to be differentially discussed for the pediatric setting. In this perspective, we aimed to describe these contextual issues and then looked ahead to the future. What might be the major challenges in celiac disease clinics in the coming years once drugs are an option alongside diet? And what will be the future objectives for researchers who further decipher the mucosal immunology of celiac disease? Speculating on the answers to these questions is as stimulating as it is fascinating to be part of this turning point.


Assuntos
Doença Celíaca , Dieta Livre de Glúten , Doença Celíaca/diagnóstico , Doença Celíaca/dietoterapia , Doença Celíaca/imunologia , Doença Celíaca/tratamento farmacológico , Humanos , Fármacos Gastrointestinais/uso terapêutico , Previsões , Aprovação de Drogas , Índice de Gravidade de Doença
3.
Gastroenterology ; 167(1): 90-103, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38604542

RESUMO

The only proven treatment for celiac disease is adherence to a strict, lifelong, gluten-free diet. However, complete dietary gluten avoidance is challenging and a substantial number of patients do not respond fully, clinically, or histologically, despite their best efforts. As celiac disease is common and its central pathophysiology is well elucidated, it has become attractive for drug development to address the limitations of dietary treatment. Most efforts address nonresponsive celiac disease, defined as continued symptoms and/or signs of disease activity despite a gluten-free diet, and the more severe forms of refractory celiac disease, types I and II. An increasing spectrum of therapeutic approaches target defined mechanisms in celiac disease pathogenesis and some have advanced to current phase 2 and 3 clinical studies. We discuss these approaches in terms of potential efficiency, practicability, safety, and need, as defined by patients, regulatory authorities, health care providers, and payors.


Assuntos
Doença Celíaca , Dieta Livre de Glúten , Doença Celíaca/dietoterapia , Doença Celíaca/imunologia , Doença Celíaca/tratamento farmacológico , Humanos , Resultado do Tratamento , Fármacos Gastrointestinais/uso terapêutico , Fármacos Gastrointestinais/efeitos adversos , Fármacos Gastrointestinais/administração & dosagem , Animais
4.
N Engl J Med ; 385(1): 35-45, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34192430

RESUMO

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.).


Assuntos
Doença Celíaca/tratamento farmacológico , Duodeno/patologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Imidazóis/administração & dosagem , Mucosa Intestinal/patologia , Piridinas/administração & dosagem , Transglutaminases/antagonistas & inibidores , Administração Oral , Adulto , Doença Celíaca/patologia , Relação Dose-Resposta a Droga , Método Duplo-Cego , Duodeno/imunologia , Feminino , Glutens/administração & dosagem , Glutens/efeitos adversos , Humanos , Imidazóis/efeitos adversos , Mucosa Intestinal/imunologia , Contagem de Linfócitos , Masculino , Pessoa de Meia-Idade , Estudo de Prova de Conceito , Proteína 2 Glutamina gama-Glutamiltransferase , Piridinas/efeitos adversos , Qualidade de Vida , Índice de Gravidade de Doença
5.
EMBO J ; 38(2)2019 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-30498130

RESUMO

Intestinal handling of dietary proteins usually prevents local inflammatory and immune responses and promotes oral tolerance. However, in ~ 1% of the world population, gluten proteins from wheat and related cereals trigger an HLA DQ2/8-restricted TH1 immune and antibody response leading to celiac disease. Prior epithelial stress and innate immune activation are essential for breaking oral tolerance to the gluten component gliadin. How gliadin subverts host intestinal mucosal defenses remains elusive. Here, we show that the α-gliadin-derived LGQQQPFPPQQPY peptide (P31-43) inhibits the function of cystic fibrosis transmembrane conductance regulator (CFTR), an anion channel pivotal for epithelial adaptation to cell-autonomous or environmental stress. P31-43 binds to, and reduces ATPase activity of, the nucleotide-binding domain-1 (NBD1) of CFTR, thus impairing CFTR function. This generates epithelial stress, tissue transglutaminase and inflammasome activation, NF-κB nuclear translocation and IL-15 production, that all can be prevented by potentiators of CFTR channel gating. The CFTR potentiator VX-770 attenuates gliadin-induced inflammation and promotes a tolerogenic response in gluten-sensitive mice and cells from celiac patients. Our results unveil a primordial role for CFTR as a central hub orchestrating gliadin activities and identify a novel therapeutic option for celiac disease.


Assuntos
Doença Celíaca/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Gliadina/farmacologia , Fragmentos de Peptídeos/farmacologia , Adolescente , Aminofenóis/administração & dosagem , Aminofenóis/farmacologia , Animais , Células CACO-2 , Doença Celíaca/tratamento farmacológico , Doença Celíaca/genética , Linhagem Celular , Criança , Regulador de Condutância Transmembrana em Fibrose Cística/química , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Humanos , Masculino , Camundongos , Ligação Proteica/efeitos dos fármacos , Conformação Proteica , Domínios Proteicos , Quinolonas/administração & dosagem , Quinolonas/farmacologia , Adulto Jovem
6.
Gastroenterology ; 163(6): 1510-1521.e6, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35931103

RESUMO

BACKGROUND & AIMS: Gluten ingestion in patients with celiac disease can lead to gastrointestinal symptoms and small intestinal mucosal injury. METHODS: This gluten challenge phase 2 trial was double blind and placebo controlled, and it assessed the efficacy and safety of a 1200-mg dose of IMGX003 in patients with celiac disease exposed to 2 g of gluten per day for 6 weeks. The change in the ratio of villus height to crypt depth was the primary endpoint. Secondary endpoints included density of intraepithelial lymphocytes and symptom severity. These endpoints were evaluated by analysis of covariance. Additional endpoints included serology and gluten-immunogenic peptides in urine. RESULTS: Fifty patients were randomized, and 43 patients completed the study (IMGX003, n = 21; placebo, n = 22). The mean change in the ratio of villus height to crypt depth (primary endpoint) for IMGX003 vs placebo was -0.04 vs -0.35 (P = .057). The mean change in the density of intraepithelial lymphocytes (secondary endpoint) for IMGX003 vs placebo was 9.8 vs 24.8 cells/mm epithelium (P = .018). The mean change (worsening) in symptom severity in relative units (secondary endpoint) for IMGX003 vs placebo was 0.22 vs 1.63 (abdominal pain, P = .231), 0.96 vs 3.29 (bloating, P = .204), and 0.02 vs 3.20 (tiredness, P = .113). The 3 × 2-week trend line significance values for these symptoms, respectively, were P = .014, .030, and .002. CONCLUSIONS: IMGX003 reduced gluten-induced intestinal mucosal damage and symptom severity. (ClinicalTrials.gov, Number: NCT03585478).


Assuntos
Doença Celíaca , Glutens , Humanos , Glutens/efeitos adversos , Doença Celíaca/diagnóstico , Doença Celíaca/tratamento farmacológico , Peptídeo Hidrolases , Mucosa Intestinal
7.
J Oncol Pharm Pract ; 29(3): 750-754, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-35918825

RESUMO

INTRODUCTION: Oral folic acid supplementation is essential for patients treated with pemetrexed, to prevent the risk of severe hematologic toxicity. In case of intestinal absorption disorder, no recommendations exist for intravenous folic acid supplementation. CASE REPORT: We describe a 74-year-old patient with multimetastatic non-small-cell lung adenocarcinoma, receiving first-line chemotherapy with carboplatin AUC5, pemetrexed 500 mg/m2 and pembrolizumab 200 mg intravenously every 3 weeks. The patient presented neglected celiac disease, resulting in malabsorption syndrome with iron and folic acid deficiency. The question was how to administer folic acid supplementation during the pemetrexed-based chemotherapy. MANAGEMENT AND OUTCOMES: Intravenous injection of 200 mg levoleucovorin on day 1 of cycle 1 of pemetrexed-based chemotherapy was administered and well tolerated. During the second cycle, the levoleucovorin perfusion was not renewed by omission. The patient was hospitalized for 7 days because of febrile aplasia. Piperacillin-tazobactam was started, and then switched to amoxicillin-clavulanate plus ciprofloxacin. After this episode of post-chemotherapy febrile aplasia, it was decided to systematically supplement the patient with intravenous levoleucovorin, with blood folate concentration monitoring at each cycle. At 16 months after start of treatment, the patient was in complete remission, indicating that the immune-chemotherapy was effective, with no further febrile neutropenia. DISCUSSION/CONCLUSION: This case report highlights intravenous levoleucovorin supplementation as an alternative to oral folic acid if needed during pemetrexed-antifolate-based chemotherapy.


Assuntos
Adenocarcinoma , Carcinoma Pulmonar de Células não Pequenas , Doença Celíaca , Neoplasias Pulmonares , Humanos , Idoso , Pemetrexede/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/patologia , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Injeções Intravenosas , Levoleucovorina , Doença Celíaca/tratamento farmacológico , Doença Celíaca/etiologia , Ácido Fólico/uso terapêutico , Suplementos Nutricionais , Adenocarcinoma/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico
8.
Int J Mol Sci ; 24(5)2023 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-36902226

RESUMO

Enzymatic modification of gliadin peptides by human transglutaminase 2 (TG2) is a key mechanism in the pathogenesis of celiac disease (CD) and represents a potential therapeutic target. Recently, we have identified the small oxidative molecule PX-12 as an effective inhibitor of TG2 in vitro. In this study, we further investigated the effect of PX-12 and the established active-site directed inhibitor ERW1041 on TG2 activity and epithelial transport of gliadin peptides. We analyzed TG2 activity using immobilized TG2, Caco-2 cell lysates, confluent Caco-2 cell monolayers and duodenal biopsies from CD patients. TG2-mediated cross-linking of pepsin-/trypsin-digested gliadin (PTG) and 5BP (5-biotinamidopentylamine) was quantified by colorimetry, fluorometry and confocal microscopy. Cell viability was tested with a resazurin-based fluorometric assay. Epithelial transport of promofluor-conjugated gliadin peptides P31-43 and P56-88 was analyzed by fluorometry and confocal microscopy. PX-12 reduced TG2-mediated cross-linking of PTG and was significantly more effective than ERW1041 (10 µM, 15 ± 3 vs. 48 ± 8%, p < 0.001). In addition, PX-12 inhibited TG2 in cell lysates obtained from Caco-2 cells more than ERW1041 (10 µM; 12 ± 7% vs. 45 ± 19%, p < 0.05). Both substances inhibited TG2 comparably in the intestinal lamina propria of duodenal biopsies (100 µM, 25 ± 13% vs. 22 ± 11%). However, PX-12 did not inhibit TG2 in confluent Caco-2 cells, whereas ERW1041 showed a dose-dependent effect. Similarly, epithelial transport of P56-88 was inhibited by ERW1041, but not by PX-12. Cell viability was not negatively affected by either substance at concentrations up to 100 µM. PX-12 did not reduce TG2 activity or gliadin peptide transport in confluent Caco-2 cells. This could be caused by rapid inactivation or degradation of the substance in the Caco-2 cell culture. Still, our in vitro data underline the potential of the oxidative inhibition of TG2. The fact that the TG2-specific inhibitor ERW1041 reduced the epithelial uptake of P56-88 in Caco-2 cells further strengthens the therapeutic potential of TG2 inhibitors in CD.


Assuntos
Doença Celíaca , Proteína 2 Glutamina gama-Glutamiltransferase , Humanos , Biópsia , Células CACO-2 , Doença Celíaca/tratamento farmacológico , Doença Celíaca/enzimologia , Gliadina/metabolismo , Mucosa Intestinal/metabolismo , Peptídeos/metabolismo , Proteína 2 Glutamina gama-Glutamiltransferase/antagonistas & inibidores , Transglutaminases/metabolismo , Intestinos/enzimologia
9.
Gastroenterology ; 161(1): 81-93.e3, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33741317

RESUMO

BACKGROUND AND AIMS: Celiac disease (CeD) is an immune-mediated disorder triggered by the ingestion of gluten. Despite adhering to a gluten-free diet (the only management option available to patients with CeD), many patients continue to experience symptoms and intestinal injury. Degradation of immunogenic fractions of gluten peptides in the stomach has been proposed as an approach to reduce toxicity of ingested gluten; however, no enzymes evaluated to date have demonstrated sufficient gluten degradation in complex meals. TAK-062 is a novel, computationally designed endopeptidase under development for the treatment of patients with CeD. METHODS: Pharmacokinetics, safety, and tolerability of TAK-062 100-900 mg were evaluated in a phase I dose escalation study in healthy participants and patients with CeD. Gluten degradation by TAK-062 was evaluated under simulated gastric conditions in vitro and in healthy participants in the phase I study, with and without pretreatment with a proton pump inhibitor. Residual gluten (collected through gastric aspiration in the phase I study) was quantified using R5 and G12 monoclonal antibody enzyme-linked immunosorbent assays. RESULTS: In vitro, TAK-062 degraded more than 99% of gluten (3 g and 9 g) within 10 minutes. In the phase I study, administration of TAK-062 was well tolerated and resulted in a median gluten degradation ranging from 97% to more than 99% in complex meals containing 1-6 g gluten at 20-65 minutes postdose. CONCLUSIONS: TAK-062 is well tolerated and rapidly and effectively degrades large amounts of gluten, supporting the development of this novel enzyme as an oral therapeutic for patients with CeD. (ClinicalTrials.gov: NCT03701555, https://clinicaltrials.gov/ct2/show/NCT03701555.).


Assuntos
Doença Celíaca/metabolismo , Endopeptidases/farmacocinética , Suco Gástrico/química , Glutens/metabolismo , Adulto , Doença Celíaca/tratamento farmacológico , Dieta Livre de Glúten , Endopeptidases/análise , Endopeptidases/farmacologia , Feminino , Gliadina/análise , Gliadina/metabolismo , Glutens/análise , Humanos , Masculino , Pessoa de Meia-Idade , Engenharia de Proteínas , Distribuição Aleatória
10.
J Pediatr Gastroenterol Nutr ; 75(5): 616-622, 2022 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-36305882

RESUMO

OBJECTIVES: Non-responsive celiac disease (NRCD) is defined as patients having persistent symptoms and enteropathy (Marsh 3 histology) suggestive of active celiac disease (CeD), after following a gluten-free diet (GFD) for at least 12 months. NRCD is suggested to affect 15% of children with CeD but data are limited and there is no research to date describing treatment of children with this condition. The aim of this study was to describe our center's approach to identifying and treating NRCD with budesonide and the Gluten Containing Elimination Diet (GCED). METHODS: We performed a retrospective, single center analysis over a 5-year period of patients with CD less than 18 years of age (inclusive) who underwent treatment for persistent symptoms and enteropathy despite following a GFD. RESULTS: We identified 22 patients with NRCD. Thirteen were treated with the GCED for 3 months with 46% achieving both histological and symptomatic resolution. Nine patients were treated with budesonide (6-9 mg), with 89% achieving both symptomatic and histologic resolution after a median 3-month treatment course. Further, 67% of patients who responded to the GCED and 100% of patients who responded to budesonide remained in remission for at least 6 months following treatment transition back to exclusive GFD. CONCLUSIONS: The GCED and budesonide can provide benefit for NRCD. Most patients with NRCD can return to a GFD after 3 months of treatment.


Assuntos
Doença Celíaca , Criança , Humanos , Doença Celíaca/tratamento farmacológico , Doença Celíaca/diagnóstico , Glutens/efeitos adversos , Estudos Retrospectivos , Budesonida/uso terapêutico , Dieta Livre de Glúten
11.
Pediatr Dev Pathol ; 25(4): 470-473, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35316607

RESUMO

Familial mediterranean fever (FMF) and Celiac Disease (CD) are both autoinflammatory and autoimmune diseases with common clinical features affecting the gastrointestinal system. Familial mediterranean fever may coexist with CD. Inflammatory cells and cytokines are up-regulated within the intestinal mucosa of patients with CD. In this perspective, lymphocyte infiltration to the lamina propria of the small intestinal mucosa is a critical pathological sign. In the present study, we aimed to find out whether the treatment of an FMF patient with colchicine affects the pathological signs of concomitant developed CD on the same patient. We retrospectively reviewed the medical records of 147 patients with FMF who were followed up in our center between 2015 and 2020. Of the 147 patients with FMF, 3 patients also had CD. In this report, we presented these 3 cases. The findings obtained in this study suggest that pathological findings may be misinterpreted in autoimmune diseases, such as CD, when patients administer anti-inflammatory treatments, such as colchicine.


Assuntos
Doenças Autoimunes , Doença Celíaca , Febre Familiar do Mediterrâneo , Doença Celíaca/diagnóstico , Doença Celíaca/tratamento farmacológico , Colchicina/uso terapêutico , Febre Familiar do Mediterrâneo/complicações , Febre Familiar do Mediterrâneo/diagnóstico , Febre Familiar do Mediterrâneo/tratamento farmacológico , Humanos , Estudos Retrospectivos
12.
Altern Ther Health Med ; 28(8): 46-49, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-35839104

RESUMO

Background: Celiac disease (CD) is an autoimmune disorder characterized by abnormal serological response (autoimmune anti-tissue-Transglutaminase antibodies) triggered by gluten ingestion in genetically predisposed individuals. The only available effective management for those patients is a strict gluten-free diet. Aim: To investigate the improvement of CD after completing a traditional herbal supplementation and alternative medicine treatment. Case description, treatment, and results: A 23-year-old female presented with a confirmed CD diagnosis (positive anti-tissue Transglutaminase IgA with infiltration of mixed inflammatory cell detected in small bowel biopsy). After 9 months of alternative treatment with traditional herbal supplementation (Taraxaf®, Ferrolina®, and Indomirol®), the clinical, laboratory, and endoscopy profile tests have shown an overall improvement with negative results for anti-tissue Transglutaminase IgA, and normal small bowel mucosal appearance. She was returned to an ordinary diet containing gluten. Conclusion: A traditional herbal supplementation with specific doses followed by a physician's instruction led to obvious improvement in this CD patient.


Assuntos
Doença Celíaca , Feminino , Humanos , Adulto Jovem , Adulto , Doença Celíaca/tratamento farmacológico , Doença Celíaca/diagnóstico , Glutens , Proteína 2 Glutamina gama-Glutamiltransferase , Imunoglobulina A , Imunidade , Suplementos Nutricionais
13.
J Am Pharm Assoc (2003) ; 62(2): 546-550, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34764035

RESUMO

BACKGROUND: Celiac disease is an autoimmune disorder that causes an intolerance to gluten. Owing to hidden sources, lack of clear labeling, and cross-contamination, it is not uncommon for individuals with celiac disease to inadvertently ingest gluten. A strict gluten-free diet is the only treatment. OBJECTIVES: The purpose of this study was to identify the frequency of activated charcoal use in celiac disease as a purported remedy despite the lack of literature on the safety and efficacy of the practice and to elucidate the manner in which using activated charcoal as a treatment for acute gluten ingestion is being communicated. METHODS: Using a descriptive study design, a Web-based survey was sent to members of the celiac disease community via social media pages for voluntary participants 18 years and older diagnosed with celiac disease. Participants were stratified into 3 major groups: (1) those who had heard of using activated charcoal as a remedy to counteract gluten ingestion and used it, (2) those who had heard of using activated charcoal but had not used it, and (3) those who had not heard of using activated charcoal. RESULTS: In subjects with celiac disease, 424 of 1613 respondents (26%) had heard of using activated charcoal as a potential remedy for gluten ingestion, and 12% had used it. The top source of information regarding any general information including remedies for maladies related to celiac disease was social media networks 72.3% (879/1613). Of those who had used activated charcoal, 122 participants (61.3%) reported subjectively feeling a reduced severity of symptoms. CONCLUSION: Pharmacists should be aware of the off-label use of activated charcoal for accidental gluten ingestion in celiac disease. Pharmacists should educate patients with celiac disease that there is not sufficient evidence to support the use of activated charcoal.


Assuntos
Doença Celíaca , Doença Celíaca/diagnóstico , Doença Celíaca/tratamento farmacológico , Carvão Vegetal/uso terapêutico , Dieta Livre de Glúten , Glutens , Humanos , Prevalência
14.
Am J Physiol Gastrointest Liver Physiol ; 320(6): G983-G989, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33881350

RESUMO

Larazotide acetate (LA) is a single-chain peptide of eight amino acids that acts as a tight junction regulator to restore intestinal barrier function. LA is currently being studied in phase III clinical trials and is orally administered to adult patients with celiac disease as an adjunct therapeutic to enhance intestinal barrier function that has been disrupted by gliadin-induced immune reactivity. Mechanistically, LA is thought to act as a zonulin antagonist to reduce zonulin-induced increases in barrier permeability and has been associated with the redistribution and rearrangement of tight junction proteins and actin filaments to restore intestinal barrier function. More recently, LA has been linked to inhibition of myosin light chain kinase, which likely reduces tension on actin filaments, thereby facilitating tight junction closure. Small (rodent) and large (porcine) animal studies have been conducted that demonstrate the importance of LA as a tight junction regulatory peptide in conditions other than celiac disease, including collagen-induced arthritis in mice and intestinal ischemic injury in pigs.


Assuntos
Doença Celíaca/tratamento farmacológico , Oligopeptídeos/farmacologia , Junções Íntimas/efeitos dos fármacos , Animais , Doença Celíaca/metabolismo , Humanos , Oligopeptídeos/uso terapêutico , Permeabilidade , Proteínas de Junções Íntimas/metabolismo , Junções Íntimas/metabolismo
15.
Gastroenterology ; 158(6): 1667-1681.e12, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32032584

RESUMO

BACKGROUND & AIMS: Celiac disease could be treated, and potentially cured, by restoring T-cell tolerance to gliadin. We investigated the safety and efficacy of negatively charged 500-nm poly(lactide-co-glycolide) nanoparticles encapsulating gliadin protein (TIMP-GLIA) in 3 mouse models of celiac disease. Uptake of these nanoparticles by antigen-presenting cells was shown to induce immune tolerance in other animal models of autoimmune disease. METHODS: We performed studies with C57BL/6; RAG1-/- (C57BL/6); and HLA-DQ8, huCD4 transgenic Ab0 NOD mice. Mice were given 1 or 2 tail-vein injections of TIMP-GLIA or control nanoparticles. Some mice were given intradermal injections of gliadin in complete Freund's adjuvant (immunization) or of soluble gliadin or ovalbumin (ear challenge). RAG-/- mice were given intraperitoneal injections of CD4+CD62L-CD44hi T cells from gliadin-immunized C57BL/6 mice and were fed with an AIN-76A-based diet containing wheat gluten (oral challenge) or without gluten. Spleen or lymph node cells were analyzed in proliferation and cytokine secretion assays or by flow cytometry, RNA sequencing, or real-time quantitative polymerase chain reaction. Serum samples were analyzed by gliadin antibody enzyme-linked immunosorbent assay, and intestinal tissues were analyzed by histology. Human peripheral blood mononuclear cells, or immature dendritic cells derived from human peripheral blood mononuclear cells, were cultured in medium containing TIMP-GLIA, anti-CD3 antibody, or lipopolysaccharide (controls) and analyzed in proliferation and cytokine secretion assays or by flow cytometry. Whole blood or plasma from healthy volunteers was incubated with TIMP-GLIA, and hemolysis, platelet activation and aggregation, and complement activation or coagulation were analyzed. RESULTS: TIMP-GLIA did not increase markers of maturation on cultured human dendritic cells or induce activation of T cells from patients with active or treated celiac disease. In the delayed-type hypersensitivity (model 1), the HLA-DQ8 transgenic (model 2), and the gliadin memory T-cell enteropathy (model 3) models of celiac disease, intravenous injections of TIMP-GLIA significantly decreased gliadin-specific T-cell proliferation (in models 1 and 2), inflammatory cytokine secretion (in models 1, 2, and 3), circulating gliadin-specific IgG/IgG2c (in models 1 and 2), ear swelling (in model 1), gluten-dependent enteropathy (in model 3), and body weight loss (in model 3). In model 1, the effects were shown to be dose dependent. Splenocytes from HLA-DQ8 transgenic mice given TIMP-GLIA nanoparticles, but not control nanoparticles, had increased levels of FOXP3 and gene expression signatures associated with tolerance induction. CONCLUSIONS: In mice with gliadin sensitivity, injection of TIMP-GLIA nanoparticles induced unresponsiveness to gliadin and reduced markers of inflammation and enteropathy. This strategy might be developed for the treatment of celiac disease.


Assuntos
Doença Celíaca/tratamento farmacológico , Gliadina/administração & dosagem , Tolerância Imunológica/efeitos dos fármacos , Nanopartículas/administração & dosagem , Administração Intravenosa , Animais , Linfócitos T CD4-Positivos , Doença Celíaca/sangue , Doença Celíaca/imunologia , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Feminino , Gliadina/imunologia , Gliadina/toxicidade , Glutens/administração & dosagem , Glutens/imunologia , Antígenos HLA-DQ/genética , Antígenos HLA-DQ/imunologia , Humanos , Mucosa Intestinal , Leucócitos Mononucleares , Camundongos , Camundongos Transgênicos , Nanopartículas/química , Nanopartículas/toxicidade , Poliglactina 910/química , Cultura Primária de Células , Testes de Toxicidade Aguda
16.
Mol Pharm ; 18(8): 3099-3107, 2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34228470

RESUMO

Celiac disease is a chronic inflammatory condition characterized by activation of the immune system in response to deamidation of gluten peptides brought about by tissue transglutaminase-2 (TG2). Overexpression of interleukin-15 (IL-15) in the intestinal epithelium and the lamina propria leads to the dysregulation of the immune system, leading to epithelial damage. The goal of this study was to develop an RNA interference therapeutic strategy for celiac disease using a combination of TG2 and IL-15 gene silencing in the inflamed intestine. TG2 and IL-15 silencing siRNA sequences, along with scrambled control, were encapsulated in a nanoparticle-in-microsphere oral system (NiMOS) and administered in a poly(I:C) mouse model of celiac disease. Single TG2 and IL-15 siRNA therapy and the combination showed effective gene silencing in vivo. Additionally, it was found that IL-15 gene silencing alone and combination in the NiMOS significantly reduced other proinflammatory cytokines. The tissue histopathology data also confirmed a reduction in immune cell infiltration and restoration of the mucosal architecture and barrier function in the intestine upon treatment. Overall, the results of this study show evidence that celiac disease can be potentially treated with an oral microsphere formulation using a combination of TG2 and IL-15 RNA interference therapeutic strategies.


Assuntos
Doença Celíaca/tratamento farmacológico , Doença Celíaca/genética , Gastroenterite/tratamento farmacológico , Gastroenterite/genética , Interleucina-15/genética , Microesferas , Sistemas de Liberação de Fármacos por Nanopartículas/química , Nanopartículas/química , Proteína 2 Glutamina gama-Glutamiltransferase/genética , Interferência de RNA , Administração Oral , Animais , Doença Celíaca/induzido quimicamente , Modelos Animais de Doenças , Composição de Medicamentos/métodos , Gastroenterite/induzido quimicamente , Interleucina-15/administração & dosagem , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Poli I-C/efeitos adversos , Proteína 2 Glutamina gama-Glutamiltransferase/administração & dosagem , RNA Interferente Pequeno/administração & dosagem , RNA Interferente Pequeno/genética , Resultado do Tratamento
17.
Dig Dis Sci ; 66(6): 1989-1997, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32654085

RESUMO

BACKGROUND: Non-responsive celiac disease (NRCD) has many aetiologies, including gluten exposure. Budesonide may be used for refractory celiac disease (RCD) and celiac crisis. AIMS: We reviewed the effectiveness of budesonide to induce clinical and histologic response in NRCD with villous atrophy (VA). METHODS: Case series of adult cases with NRCD and VA prescribed budesonide at two celiac centers. Clinical variables and mucosal recovery (i.e., normal villous architecture within 1 year of treatment) were evaluated. RESULTS: Forty-two cases [77% female, median age 45.0 (IQR 28.3-60.0) years] were included. Most common symptoms were diarrhea (64%) and abdominal pain (62%). Budesonide was initiated at 9 mg (83%) for a median duration of 16.0 weeks (IQR 6.8-25.0 weeks). In total, 57% exhibited a clinical response, positively associated with diarrhea (adjusted OR 6.08 95% CI 1.04-35.47) and negatively with fatigue (adjusted OR 0.18 95% CI 0.03-0.98). Clinical response was higher among those with dietitian counseling prior to budesonide (n = 29, 70 vs. 23%, p < 0.01). Mucosal recovery was observed in 11/24 with follow-up duodenal biopsies. There was no association between clinical response and mucosal recovery, and 79% of clinical responders had a symptomatic relapse. RCD (48%) and chronic gluten exposure (24%) were the main suspected aetiologies of NRCD. Most individuals without a clinical response subsequently received an IBS-related diagnosis. CONCLUSIONS: Budesonide may be effective to induce clinical response in NRCD presenting with diarrhea and VA, but clinical recurrence and lack of mucosal recovery are frequent after tapering. Other diagnoses, including coexisting IBS, may be considered in non-responders to budesonide therapy.


Assuntos
Anti-Inflamatórios/administração & dosagem , Budesonida/administração & dosagem , Doença Celíaca/diagnóstico , Doença Celíaca/tratamento farmacológico , Gerenciamento Clínico , Adulto , Anti-Inflamatórios/metabolismo , Budesonida/metabolismo , Doença Celíaca/metabolismo , Estudos de Coortes , Dieta Livre de Glúten/métodos , Dieta Livre de Glúten/tendências , Relação Dose-Resposta a Droga , Esquema de Medicação , Feminino , Seguimentos , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
18.
Int J Mol Sci ; 22(13)2021 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-34209932

RESUMO

Enzymatic transamidation of gliadins by microbial transglutaminase (mTG) inhibits interferon-γ (IFN-γ) secretion by intestinal T cell lines in patients with celiac disease (CD). To gain insight into the cellular mechanisms underlying the down-regulatory effects of transamidation, we tested a single recombinant α-gliadin (r-gliadin) harbouring two immunodominant peptides, p13 (aa. 120-139) and p23 (aa. 220-239), in HLA-DQ8 transgenic mice, a model of gluten sensitivity. Mice were intranasally immunised with r-gliadin or r-gliadin transamidated by mTG (K-r-gliadin) along with cholera toxin, and the response of mesenteric lymph node cells was analysed by cytokine multiplex assay. An in vitro challenge with r-gliadin was characterised by secretion of specific cytokines featuring both innate immunity and the Th1/Th2/Th17 pattern of the adaptive response. Notably, transamidation specifically down-regulated the Th1 response. Structural studies performed on K-r-gliadin confirmed that specific glutamine residues in p13 and p23, previously found to be deamidated by tissue transglutaminase, were also transamidated by mTG. In silico analysis, simulating p13 and p23 peptide binding to HLA-DQ8 showed that these glutamines, in the form of glutamate, could interact by means of salt bridges with peculiar amino acids of the alpha chain of HLA-DQ8, suggesting that their transamidation may influence the HLA-restricted recognition of these peptides. Thus, the structural findings provided a rationale to explain the down-regulation of the r-gliadin-specific Th1 response following transamidation.


Assuntos
Doença Celíaca/tratamento farmacológico , Toxina da Cólera/administração & dosagem , Citocinas/metabolismo , Gliadina/administração & dosagem , Antígenos HLA-DQ/genética , Transglutaminases/metabolismo , Administração Intranasal , Animais , Doença Celíaca/genética , Doença Celíaca/imunologia , Toxina da Cólera/imunologia , Citocinas/efeitos dos fármacos , Modelos Animais de Doenças , Regulação para Baixo , Regulação da Expressão Gênica , Gliadina/química , Gliadina/genética , Gliadina/imunologia , Antígenos HLA-DQ/metabolismo , Imunização , Epitopos Imunodominantes/imunologia , Camundongos , Camundongos Transgênicos , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/química , Proteínas Recombinantes/imunologia , Células Th1/imunologia , Células Th17/imunologia , Células Th2/imunologia
20.
Anal Biochem ; 592: 113584, 2020 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-31953047

RESUMO

Celiac disease (CD) is a chronic immune-mediated disease in which gluten ingestion leads to damage of the small intestinal mucosa in genetically susceptible individuals. The enteropathy is mainly induced by the production of IFN-γ from intestinal CD4+T cells that recognise gliadin peptides following deamidation by tissue transglutaminase. The only available therapy is a strict, lifelong gluten-free diet (GFD). This diet is strongly demanding for patients, which justifies the search for alternative strategies. The enzyme approach is one promising strategy to address this issue. In particular, transamidation of wheat gliadin by microbial transglutaminase (mTG) was fully effective at inhibiting gliadin-specific IFN-γ secretion in intestinal T cells from CD patients. Furthermore, transamidated gliadin induced higher levels of the anti-inflammatory IL-10 than native gliadin in different in vitro models. These data suggest that a more balanced immune response could be induced by mTG-treated gliadin in the small intestine of celiac patients. Furthermore, the highlighted biological property of mTG-treated gliadin could be exploited to induce tolerance to native gliadin in at-risk individuals.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Doença Celíaca/tratamento farmacológico , Gliadina/metabolismo , Mucosa Intestinal/efeitos dos fármacos , Transglutaminases , Triticum , Bactérias/enzimologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/patologia , Farinha , Humanos , Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Transglutaminases/farmacologia , Transglutaminases/uso terapêutico , Triticum/efeitos dos fármacos , Triticum/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA