Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 174
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 56(8): 1862-1875.e9, 2023 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-37478853

RESUMO

Loss of oral tolerance (LOT) to gluten, driven by dendritic cell (DC) priming of gluten-specific T helper 1 (Th1) cell immune responses, is a hallmark of celiac disease (CeD) and can be triggered by enteric viral infections. Whether certain commensals can moderate virus-mediated LOT remains elusive. Here, using a mouse model of virus-mediated LOT, we discovered that the gut-colonizing protist Tritrichomonas (T.) arnold promotes oral tolerance and protects against reovirus- and murine norovirus-mediated LOT, independent of the microbiota. Protection was not attributable to antiviral host responses or T. arnold-mediated innate type 2 immunity. Mechanistically, T. arnold directly restrained the proinflammatory program in dietary antigen-presenting DCs, subsequently limiting Th1 and promoting regulatory T cell responses. Finally, analysis of fecal microbiomes showed that T. arnold-related Parabasalid strains are underrepresented in human CeD patients. Altogether, these findings will motivate further exploration of oral-tolerance-promoting protists in CeD and other immune-mediated food sensitivities.


Assuntos
Antígenos , Imunidade Inata , Animais , Camundongos , Humanos , Dieta , Glutens , Células Dendríticas , Tolerância Imunológica
2.
Nat Immunol ; 19(7): 685-695, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29925984

RESUMO

Risk factors for most autoimmune diseases are multifactorial genetic variants modified by environmental risk factors. Type 1 diabetes and celiac disease share high-risk HLA haplotypes, and the prevalence of both diseases has increased in many regions during the past half century. Unknown environmental factors are suspected to have increased the disease penetrance. Celiac disease depends on immune responses to dietary gluten, whereas the environmental risk factors for type 1 diabetes are not yet clear. Here, we consider the shared heritable genetic factors and review evidence of the dietary and microbial exposures, particularly in early life, that might influence the pathogenesis of one or both diseases. A deeper mechanistic understanding of the environmental factors responsible for increased risk of these diseases should provide opportunities to manipulate exposure in children carrying defined risk markers and thus prevent and attenuate disease, as well as to identify new therapeutic strategies for patients.


Assuntos
Doença Celíaca/genética , Doença Celíaca/imunologia , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Animais , Autoantígenos/imunologia , Doença Celíaca/microbiologia , Diabetes Mellitus Tipo 1/epidemiologia , Diabetes Mellitus Tipo 1/microbiologia , Dieta , Microbioma Gastrointestinal , Antígenos HLA/genética , Humanos , Lactente , Infecções/complicações , Fatores de Risco
3.
Gastroenterology ; 166(1): 88-102, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-37704112

RESUMO

BACKGROUND & AIMS: There is a need to develop safe and effective pharmacologic options for the treatment of celiac disease (CeD); however, consensus on the appropriate design and configuration of randomized controlled trials (RCTs) in this population is lacking. METHODS: A 2-round modified Research and Development/University of California Los Angeles Appropriateness Method study was conducted. Eighteen gastroenterologists (adult and pediatric) and gastrointestinal pathologists voted on statements pertaining to the configuration of CeD RCTs, inclusion and exclusion criteria, gluten challenge, and trial outcomes. Two RCT designs were considered, representing the following distinct clinical scenarios for which pharmacotherapy may be used: trials incorporating a gluten challenge to simulate exposure; and trials evaluating reversal of histologic changes, despite attempted adherence to a gluten-free diet. Each statement was rated as appropriate, uncertain, or inappropriate, using a 9-point Likert scale. RESULTS: For trials evaluating prevention of relapse after gluten challenge, participants adherent to a gluten-free diet for 12 months or more with normal or near-normal-sized villi should be enrolled. Gluten challenge should be FODMAPS (fermentable oligosaccharides, disaccharides, monosaccharides, and polyols) free, and efficacy evaluated using histology with a secondary patient-reported outcome measure. For trials evaluating reversal of villus atrophy, the panel voted it appropriate to enroll participants with a baseline villus height to crypt depth ratio ≤2 and measure efficacy using a primary histologic end point. Guidance for measuring histologic, endoscopic, and patient-reported outcomes in adult and pediatric patients with CeD are provided, along with recommendations regarding the merits and limitations of different end points. CONCLUSIONS: We developed standardized recommendations for clinical trial design, eligibility criteria, outcome measures, gluten challenge, and disease evaluations for RCTs in patients with CeD.


Assuntos
Doença Celíaca , Adulto , Humanos , Criança , Doença Celíaca/patologia , Recidiva Local de Neoplasia , Ensaios Clínicos Controlados Aleatórios como Assunto , Glutens/efeitos adversos , Dieta Livre de Glúten
4.
Am J Physiol Gastrointest Liver Physiol ; 326(6): G687-G696, 2024 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-38591144

RESUMO

Tryptophan is an essential amino acid transformed by host and gut microbial enzymes into metabolites that regulate mucosal homeostasis through aryl hydrocarbon receptor (AhR) activation. Alteration of tryptophan metabolism has been associated with chronic inflammation; however, whether tryptophan supplementation affects the metabolite repertoire and AhR activation under physiological conditions in humans is unknown. We performed a randomized, double blind, placebo-controlled, crossover study in 20 healthy volunteers. Subjects on a low tryptophan background diet were randomly assigned to a 3-wk l-tryptophan supplementation (3 g/day) or placebo, and after a 2-wk washout switched to opposite interventions. We assessed gastrointestinal and psychological symptoms by validated questionnaires, AhR activation by cell reporter assay, tryptophan metabolites by liquid chromatography and high-resolution mass spectrometry, cytokine production in isolated monocytes by ELISA, and microbiota profile by 16S rRNA Illumina technique. Oral tryptophan supplementation was well tolerated, with no changes in gastrointestinal or psychological scores. Compared with placebo, tryptophan increased AhR activation capacity by duodenal contents, but not by feces. This was paralleled by higher urinary and plasma kynurenine metabolites and indoles. Tryptophan had a modest impact on fecal microbiome profiles and no significant effect on cytokine production. At the doses used in this study, oral tryptophan supplementation in humans induces microbial indole and host kynurenine metabolic pathways in the small intestine, known to be immunomodulatory. The results should prompt tryptophan intervention strategies in inflammatory conditions of the small intestine where the AhR pathway is impaired.NEW & NOTEWORTHY We demonstrate that in healthy subjects, orally administered tryptophan activates microbial indole and host kynurenine pathways in the small intestine, the primary metabolic site for dietary components, and the richest source of immune cells along the gut. This study provides novel insights in how to optimally activate immunomodulatory AhR pathways and indole metabolism in the small intestine, serving as basis for future therapeutic trials using l-tryptophan supplementation in chronic inflammatory conditions affecting the small intestine.


Assuntos
Estudos Cross-Over , Duodeno , Voluntários Saudáveis , Receptores de Hidrocarboneto Arílico , Triptofano , Humanos , Triptofano/metabolismo , Triptofano/administração & dosagem , Receptores de Hidrocarboneto Arílico/metabolismo , Masculino , Adulto , Feminino , Duodeno/metabolismo , Duodeno/efeitos dos fármacos , Método Duplo-Cego , Suplementos Nutricionais , Microbioma Gastrointestinal/efeitos dos fármacos , Adulto Jovem , Administração Oral , Cinurenina/metabolismo , Citocinas/metabolismo , Fezes/microbiologia , Fezes/química , Indóis/farmacologia , Indóis/administração & dosagem , Fatores de Transcrição Hélice-Alça-Hélice Básicos
5.
Nature ; 557(7706): 580-584, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29769727

RESUMO

Somatic mutations in tet methylcytosine dioxygenase 2 (TET2), which encodes an epigenetic modifier enzyme, drive the development of haematopoietic malignancies1-7. In both humans and mice, TET2 deficiency leads to increased self-renewal of haematopoietic stem cells with a net developmental bias towards the myeloid lineage1,4,8,9. However, pre-leukaemic myeloproliferation (PMP) occurs in only a fraction of Tet2-/- mice8,9 and humans with TET2 mutations1,3,5-7, suggesting that extrinsic non-cell-autonomous factors are required for disease onset. Here we show that bacterial translocation and increased interleukin-6 production, resulting from dysfunction of the small-intestinal barrier, are critical for the development of PMP in mice that lack Tet2 expression in haematopoietic cells. Furthermore, in symptom-free Tet2-/- mice, PMP can be induced by disrupting intestinal barrier integrity, or in response to systemic bacterial stimuli such as the toll-like receptor 2 agonist. PMP was reversed by antibiotic treatment and failed to develop in germ-free Tet2-/- mice, which illustrates the importance of microbial signals in the development of this condition. Our findings demonstrate the requirement for microbial-dependent inflammation in the development of PMP and provide a mechanistic basis for the variation in PMP penetrance observed in Tet2-/- mice. This study will prompt new lines of investigation that may profoundly affect the prevention and management of haematopoietic malignancies.


Assuntos
Doenças Assintomáticas , Fenômenos Fisiológicos Bacterianos , Proliferação de Células , Proteínas de Ligação a DNA/deficiência , Leucemia/microbiologia , Leucemia/patologia , Proteínas Proto-Oncogênicas/deficiência , Animais , Infecções Bacterianas/imunologia , Infecções Bacterianas/microbiologia , Fenômenos Fisiológicos Bacterianos/imunologia , Proteínas de Ligação a DNA/genética , Dioxigenases , Feminino , Vida Livre de Germes , Inflamação/microbiologia , Interleucina-6/imunologia , Mucosa Intestinal/metabolismo , Lactobacillus/química , Lactobacillus/citologia , Lactobacillus/imunologia , Masculino , Camundongos , Penetrância , Permeabilidade , Proteínas Proto-Oncogênicas/genética , Receptor 2 Toll-Like/agonistas
6.
Am J Physiol Gastrointest Liver Physiol ; 324(4): G305-G321, 2023 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-36749921

RESUMO

Macrophages are essential for homeostatic maintenance of the anti-inflammatory and tolerogenic intestinal environment, yet monocyte-derived macrophages can promote local inflammation. Proinflammatory macrophage accumulation within the intestines may contribute to the development of systemic chronic inflammation and immunometabolic dysfunction in obesity. Using a model of high-fat diet-induced obesity in C57BL/6J female mice, we assessed intestinal paracellular permeability by in vivo and ex vivo assays and quantitated intestinal macrophages in ileum and colon tissues by multicolor flow cytometry after short (6 wk), intermediate (12 wk), and prolonged (18 wk) diet allocation. We characterized monocyte-derived CD4-TIM4- and CD4+TIM4- macrophages, as well as tissue-resident CD4+TIM4+ macrophages. Diet-induced obesity had tissue- and time-dependent effects on intestinal permeability, as well as monocyte and macrophage numbers, surface marker phenotype, and intracellular production of the cytokines IL-10 and tumor necrosis factor (TNF). We found that obese mice had increased paracellular permeability, in particular within the ileum, but this did not elicit recruitment of monocytes nor a local proinflammatory response by monocyte-derived or tissue-resident macrophages in either the ileum or colon. Proliferation of monocyte-derived and tissue-resident macrophages was also unchanged. Wild-type and TNF-/- littermate mice had similar intestinal permeability and macrophage population characteristics in response to diet-induced obesity. These data are unique from reported effects of diet-induced obesity on macrophages in metabolic tissues, as well as outcomes of acute inflammation within the intestines. These experiments also collectively indicate that TNF does not mediate effects of diet-induced obesity on paracellular permeability or intestinal monocyte-derived and tissue-resident intestinal macrophages in young female mice.NEW & NOTEWORTHY We found that diet-induced obesity in female mice has tissue- and time-dependent effects on intestinal paracellular permeability as well as monocyte-derived and tissue-resident macrophage numbers, surface marker phenotype, and intracellular production of the cytokines IL-10 and TNF. These changes were not mediated by TNF.


Assuntos
Interleucina-10 , Monócitos , Feminino , Animais , Camundongos , Monócitos/metabolismo , Interleucina-10/metabolismo , Camundongos Endogâmicos C57BL , Intestinos/patologia , Obesidade/metabolismo , Macrófagos/metabolismo , Inflamação/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Citocinas/metabolismo , Dieta Hiperlipídica , Permeabilidade
7.
Lancet ; 399(10344): 2413-2426, 2022 06 25.
Artigo em Inglês | MEDLINE | ID: mdl-35691302

RESUMO

Coeliac disease is an autoimmune disorder that primarily affects the small intestine, and is caused by the ingestion of gluten in genetically susceptible individuals. Prevalence in the general population ranges from 0·5% to 2%, with an average of about 1%. The development of the coeliac enteropathy depends on a complex immune response to gluten proteins, including both adaptive and innate mechanisms. Clinical presentation of coeliac disease is highly variable and includes classical and non-classical gastrointestinal symptoms, extraintestinal manifestations, and subclinical cases. The disease is associated with a risk of complications, such as osteoporosis and intestinal lymphoma. Diagnosis of coeliac disease requires a positive serology (IgA anti-transglutaminase 2 and anti-endomysial antibodies) and villous atrophy on small-intestinal biopsy. Treatment involves a gluten-free diet; however, owing to the high psychosocial burden of such a diet, research into alternative pharmacological treatments is currently very active.


Assuntos
Doença Celíaca , Doença Celíaca/diagnóstico , Doença Celíaca/epidemiologia , Doença Celíaca/terapia , Dieta Livre de Glúten , Glutens/efeitos adversos , Glutens/metabolismo , Humanos , Mucosa Intestinal/patologia , Intestino Delgado/patologia
8.
Gastroenterology ; 163(5): 1351-1363.e15, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-35810781

RESUMO

BACKGROUND & AIMS: Genes and gluten are necessary but insufficient to cause celiac disease (CeD). Altered gut microbiota has been implicated as an additional risk factor. Variability in sampling site may confound interpretation and mechanistic insight, as CeD primarily affects the small intestine. Thus, we characterized CeD microbiota along the duodenum and in feces and verified functional impact in gnotobiotic mice. METHODS: We used 16S rRNA gene sequencing (Illumina) and predicted gene function (PICRUSt2) in duodenal biopsies (D1, D2 and D3), aspirates, and stool from patients with active CeD and controls. CeD alleles were determined in consented participants. A subset of duodenal samples stratified according to similar CeD risk genotypes (controls DQ2-/- or DQ2+/- and CeD DQ2+/-) were used for further analysis and to colonize germ-free mice for gluten metabolism studies. RESULTS: Microbiota composition and predicted function in CeD was largely determined by intestinal location. In the duodenum, but not stool, there was higher abundance of Escherichia coli (D1), Prevotella salivae (D2), and Neisseria (D3) in CeD vs controls. Predicted bacterial protease and peptidase genes were altered in CeD and impaired gluten degradation was detected only in mice colonized with CeD microbiota. CONCLUSIONS: Our results showed luminal and mucosal microbial niches along the gut in CeD. We identified novel microbial proteolytic pathways involved in gluten detoxification that are impaired in CeD but not in controls carrying DQ2, suggesting an association with active duodenal inflammation. Sampling site should be considered a confounding factor in microbiome studies in CeD.


Assuntos
Doença Celíaca , Microbioma Gastrointestinal , Camundongos , Animais , Doença Celíaca/complicações , RNA Ribossômico 16S/genética , Glutens/metabolismo , Peptídeo Hidrolases
9.
PLoS Pathog ; 17(8): e1009719, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34352037

RESUMO

Reducing food intake is a common host response to infection, yet it remains unclear whether fasting is detrimental or beneficial to an infected host. Despite the gastrointestinal tract being the primary site of nutrient uptake and a common route for infection, studies have yet to examine how fasting alters the host's response to an enteric infection. To test this, mice were fasted before and during oral infection with the invasive bacterium Salmonella enterica serovar Typhimurium. Fasting dramatically interrupted infection and subsequent gastroenteritis by suppressing Salmonella's SPI-1 virulence program, preventing invasion of the gut epithelium. Virulence suppression depended on the gut microbiota, as Salmonella's invasion of the epithelium proceeded in fasting gnotobiotic mice. Despite Salmonella's restored virulence within the intestines of gnotobiotic mice, fasting downregulated pro-inflammatory signaling, greatly reducing intestinal pathology. Our study highlights how food intake controls the complex relationship between host, pathogen and gut microbiota during an enteric infection.


Assuntos
Bactérias/crescimento & desenvolvimento , Jejum , Gastroenterite/prevenção & controle , Inflamação/prevenção & controle , Intestinos/imunologia , NF-kappa B/antagonistas & inibidores , Salmonelose Animal/imunologia , Salmonella typhimurium/fisiologia , Animais , Bactérias/imunologia , Bactérias/metabolismo , Feminino , Gastroenterite/imunologia , Gastroenterite/microbiologia , Microbioma Gastrointestinal , Inflamação/imunologia , Inflamação/microbiologia , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Salmonelose Animal/complicações , Salmonelose Animal/microbiologia , Salmonelose Animal/patologia
10.
Gut ; 71(1): 68-76, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-33526437

RESUMO

OBJECTIVES: Coeliac disease (CD) is a complex autoimmune disorder that develops in genetically susceptible individuals. Dietary gluten triggers an immune response for which the only available treatment so far is a strict, lifelong gluten free diet. Human leucocyte antigen (HLA) genes and several non-HLA regions have been associated with the genetic susceptibility to CD, but their role in the pathogenesis of the disease is still essentially unknown, making it complicated to develop much needed non-dietary treatments. Here, we describe the functional involvement of a CD-associated single-nucleotide polymorphism (SNP) located in the 5'UTR of XPO1 in the inflammatory environment characteristic of the coeliac intestinal epithelium. DESIGN: The function of the CD-associated SNP was investigated using an intestinal cell line heterozygous for the SNP, N6-methyladenosine (m6A)-related knock-out and HLA-DQ2 mice, and human samples from patients with CD. RESULTS: Individuals harbouring the risk allele had higher m6A methylation in the 5'UTR of XPO1 RNA, rendering greater XPO1 protein amounts that led to downstream nuclear factor kappa B (NFkB) activity and subsequent inflammation. Furthermore, gluten exposure increased overall m6A methylation in humans as well as in in vitro and in vivo models. CONCLUSION: We identify a novel m6A-XPO1-NFkB pathway that is activated in CD patients. The findings will prompt the development of new therapeutic approaches directed at m6A proteins and XPO1, a target under evaluation for the treatment of intestinal disorders.


Assuntos
Doença Celíaca/genética , Carioferinas/genética , Polimorfismo de Nucleotídeo Único , RNA/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Adenosina/análogos & derivados , Adenosina/genética , Animais , Linhagem Celular , Modelos Animais de Doenças , Células Epiteliais/patologia , Antígenos HLA-DQ/genética , Humanos , Mucosa Intestinal/patologia , Metilação , Camundongos Knockout , NF-kappa B/metabolismo , Proteína Exportina 1
11.
Gut ; 2022 Dec 13.
Artigo em Inglês | MEDLINE | ID: mdl-36591617

RESUMO

OBJECTIVE: Dietary therapies for irritable bowel syndrome (IBS) have received increasing interest but predicting which patients will benefit remains a challenge due to a lack of mechanistic insight. We recently found evidence of a role for the microbiota in dietary modulation of pain signalling in a humanised mouse model of IBS. This randomised cross-over study aimed to test the hypothesis that pain relief following reduced consumption of fermentable carbohydrates is the result of changes in luminal neuroactive metabolites. DESIGN: IBS (Rome IV) participants underwent four trial periods: two non-intervention periods, followed by a diet low (LFD) and high in fermentable carbohydrates for 3 weeks each. At the end of each period, participants completed questionnaires and provided stool. The effects of faecal supernatants (FS) collected before (IBS FS) and after a LFD (LFD FS) on nociceptive afferent neurons were assessed in mice using patch-clamp and ex vivo colonic afferent nerve recording techniques. RESULTS: Total IBS symptom severity score and abdominal pain were reduced by the LFD (N=25; p<0.01). Excitability of neurons was increased in response to IBS FS, but this effect was reduced (p<0.01) with LFD FS from pain-responders. IBS FS from pain-responders increased mechanosensitivity of nociceptive afferent nerve axons (p<0.001), an effect lost following LFD FS administration (p=NS) or when IBS FS was administered in the presence of antagonists of histamine receptors or protease inhibitors. CONCLUSIONS: In a subset of IBS patients with improvement in abdominal pain following a LFD, there is a decrease in pronociceptive signalling from FS, suggesting that changes in luminal mediators may contribute to symptom response.

12.
Gastroenterology ; 161(5): 1395-1411.e4, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34416277

RESUMO

Celiac disease (CeD) is a frequent immune-mediated disease that affects not only the small intestine but also many extraintestinal sites. The role of gluten proteins as dietary triggers, HLA-DQ2 or -DQ8 as major necessary genetic predisposition, and tissue transglutaminase (TG2) as mechanistically involved autoantigen, are unique features of CeD. Recent research implicates many cofactors working in synergism with these key triggers, including the intestinal microbiota and their metabolites, nongluten dietary triggers, intestinal barrier defects, novel immune cell phenotypes, and mediators and cytokines. In addition, apart from HLA-DQ2 and -DQ8, multiple and complex predisposing genetic factors and interactions have been defined, most of which overlap with predispositions in other, usually autoimmune, diseases that are linked to CeD. The resultant better understanding of CeD pathogenesis, and its manifold manifestations has already paved the way for novel therapeutic approaches beyond the lifelong strict gluten-free diet, which poses a burden to patients and often does not lead to complete mucosal healing. Thus, supported by improved mouse models for CeD and in vitro organoid cultures, several targeted therapies are in phase 2-3 clinical studies, such as highly effective gluten-degrading oral enzymes, inhibition of TG2, cytokine therapies, induction of tolerance to gluten ingestion, along with adjunctive and preventive approaches using beneficial probiotics and micronutrients. These developments are supported by novel noninvasive markers of CeD severity and activity that may be used as companion diagnostics, allow easy-to perform and reliable monitoring of patients, and finally support personalized therapy for CeD.


Assuntos
Bactérias/imunologia , Doença Celíaca/terapia , Microbioma Gastrointestinal , Glutens/imunologia , Fenômenos Imunogenéticos , Testes Imunológicos , Intestinos/imunologia , Animais , Bactérias/patogenicidade , Doença Celíaca/genética , Doença Celíaca/imunologia , Doença Celíaca/microbiologia , Modelos Animais de Doenças , Predisposição Genética para Doença , Interações Hospedeiro-Patógeno , Humanos , Intestinos/microbiologia , Intestinos/patologia , Fenótipo , Valor Preditivo dos Testes , Prognóstico , Fatores de Risco
13.
Gastroenterology ; 160(5): 1532-1545, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33310084

RESUMO

BACKGROUND & AIMS: Altered gut microbiota composition and function have been associated with inflammatory bowel diseases, including ulcerative colitis (UC), but the causality and mechanisms remain unknown. METHODS: We applied 16S ribosomal RNA gene sequencing, shotgun metagenomic sequencing, in vitro functional assays, and gnotobiotic colonizations to define the microbial composition and function in fecal samples obtained from a cohort of healthy individuals at risk for inflammatory bowel diseases (pre-UC) who later developed UC (post-UC) and matched healthy control individuals (HCs). RESULTS: Microbiota composition of post-UC samples was different from HC and pre-UC samples; however, functional analysis showed increased fecal proteolytic and elastase activity before UC onset. Metagenomics identified more than 22,000 gene families that were significantly different between HC, pre-UC, and post-UC samples. Of these, 237 related to proteases and peptidases, suggesting a bacterial component to the pre-UC proteolytic signature. Elastase activity inversely correlated with the relative abundance of Adlercreutzia and other potentially beneficial taxa and directly correlated with known proteolytic taxa, such as Bacteroides vulgatus. High elastase activity was confirmed in Bacteroides isolates from fecal samples. The bacterial contribution and functional significance of the proteolytic signature were investigated in germ-free adult mice and in dams colonized with HC, pre-UC, or post-UC microbiota. Mice colonized with or born from pre-UC-colonized dams developed higher fecal proteolytic activity and an inflammatory immune tone compared with HC-colonized mice. CONCLUSIONS: We have identified increased fecal proteolytic activity that precedes the clinical diagnosis of UC and associates with gut microbiota changes. This proteolytic signature may constitute a noninvasive biomarker of inflammation to monitor at-risk populations that can be targeted therapeutically with antiproteases.


Assuntos
Bactérias/enzimologia , Proteínas de Bactérias/metabolismo , Colite Ulcerativa/microbiologia , Fezes/microbiologia , Microbioma Gastrointestinal , Peptídeo Hidrolases/metabolismo , Adolescente , Adulto , Animais , Bactérias/efeitos dos fármacos , Bactérias/genética , Proteínas de Bactérias/genética , Biomarcadores/metabolismo , Estudos de Casos e Controles , Criança , Colite Ulcerativa/diagnóstico , Colite Ulcerativa/tratamento farmacológico , Modelos Animais de Doenças , Transplante de Microbiota Fecal , Feminino , Microbioma Gastrointestinal/efeitos dos fármacos , Vida Livre de Germes , Humanos , Masculino , Metagenoma , Metagenômica , Camundongos Endogâmicos C57BL , Peptídeo Hidrolases/genética , Valor Preditivo dos Testes , Estudos Prospectivos , Inibidores de Proteases/uso terapêutico , Proteólise , Reprodutibilidade dos Testes , Ribotipagem , Adulto Jovem
14.
Dermatol Ther ; 35(11): e15844, 2022 11.
Artigo em Inglês | MEDLINE | ID: mdl-36124339

RESUMO

Apremilast is an oral small molecule approved for the treatment of psoriasis, psoriatic arthritis and oral ulcers associated with Behçet's disease. This research was conducted to describe the characteristics of patients who received treatment with apremilast for a skin disorder, either before, during, or after a biological treatment, with the aim of analyze the reasons that lead to start this drug in real clinical practice or suspend it for another. A total of 41 patients were enrolled: nine (22.0%) had received biological treatment prior to apremilast, seven (17.0%) both before and after apremilast and 25 (61.0%) after apremilast. One patient received concomitant treatment with adalimumab and apremilast. Most patients (85.4%) received apremilast as treatment for psoriasis. Reasons for starting apremilast were lack of efficacy with previous treatments (85.4%) and adverse effects or contraindication to previous treatments (14.6%), without statistically significant differences between patients who had received a previous biologic and those who had not. Drug survival was not influenced by previous biological treatment, but we found an increased risk of drug discontinuation in patients with chronic kidney disease (log-rank p = 0.028). The main reason of apremilast withdrawal was lack of adequate disease control (60.0%), most of whom required treatment with biologics. Therefore, despite the extensive development of new therapies for psoriasis and other dermatological conditions, apremilast is a widely used drug even in patients who are candidates for biologic treatment. Its initiation is more frequent due to poor disease control than because of other therapies contraindications.


Assuntos
Artrite Psoriásica , Produtos Biológicos , Psoríase , Humanos , Anti-Inflamatórios não Esteroides/efeitos adversos , Talidomida/efeitos adversos , Artrite Psoriásica/tratamento farmacológico , Psoríase/tratamento farmacológico , Psoríase/induzido quimicamente , Produtos Biológicos/efeitos adversos
15.
Clin Gastroenterol Hepatol ; 19(3): 484-491.e1, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32217152

RESUMO

BACKGROUND & AIMS: It is not clear how often patients who are on gluten-free diets (GFDs) for treatment of celiac disease still are exposed to gluten. We studied levels of gluten immunogenic peptides (GIP) in fecal and urine samples, collected over 4 weeks, from patients with celiac disease on a long-term GFD. METHODS: We performed a prospective study of 53 adults with celiac disease who had been on a GFD for more than 2 years (median duration, 8 y; interquartile range, 5-12 y) in Argentina. At baseline, symptoms were assessed by the celiac symptom index questionnaire. Patients collected stool each Friday and Saturday and urine samples each Sunday for 4 weeks. We used a commercial enzyme-linked immunosorbent assay to measure GIP in stool and point-of-care tests to measure GIP in urine samples. RESULTS: Overall, 159 of 420 stool and urine samples (37.9%) were positive for GIP; 88.7% of patients had at least 1 fecal or urine sample that was positive for GIP (median, 3 excretions). On weekends (urine samples), 69.8% of patients excreted GIP at least once, compared with 62.3% during weekdays (stool). The number of patients with a sample that was positive for GIP increased over the 4-week study period (urine samples in week 1 vs week 4: P < .05). Patients with symptoms had more weeks in which GIP was detected in stool than patients without symptoms (P < .05). The number of samples that were positive for GIP correlated with titers of deamidated gliadin peptide IgA in patients' blood samples, but not with levels of tissue transglutaminase. CONCLUSIONS: Patients with celiac disease on a long-term GFD still frequently are exposed to gluten. Assays to detect GIP in stool and urine might be used to assist dietitians in assessment of GFD compliance.


Assuntos
Doença Celíaca , Gliadina , Adulto , Dieta Livre de Glúten , Glutens , Humanos , Peptídeos , Estudos Prospectivos
16.
Gastroenterology ; 159(3): 884-903.e31, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32416141

RESUMO

BACKGROUND & AIMS: There is controversy over the association between celiac disease (CeD) and inflammatory bowel diseases (IBD). We performed a systematic review and meta-analysis to assess evidence for an association between CeD and IBD. METHODS: We searched databases including MEDLINE, EMBASE, CENTRAL, Web of Science, CINAHL, DARE, and SIGLE through June 25, 2019 for studies assessing the risk of CeD in patients with IBD, and IBD in patients with CeD, compared with controls of any type. We used the Newcastle-Ottawa Scale to evaluate the risk of bias and GRADE to assess the certainty of the evidence. RESULTS: We identified 9791 studies and included 65 studies in our analysis. Moderate certainty evidence found an increased risk of CeD in patients with IBD vs controls (risk ratio [RR] 3.96; 95% confidence interval [CI] 2.23-7.02) and increased risk of IBD in patients with CeD vs controls (RR 9.88; 95% CI 4.03-24.21). There was low-certainty evidence for the risk of anti-Saccharomyces antibodies, a serologic marker of IBD, in patients with CeD vs controls (RR 6.22; 95% CI 2.44-15.84). There was low-certainty evidence for no difference in risk of HLA-DQ2 or DQ8 in patients with IBD vs controls (RR 1.04; 95% CI 0.42-2.56), and very low-certainty evidence for an increased risk of anti-tissue transglutaminase in patients with IBD vs controls (RR 1.52; 95% CI 0.52-4.40). Patients with IBD had a slight decrease in risk of anti-endomysial antibodies vs controls (RR 0.70; 95% CI 0.18-2.74), but these results are uncertain. CONCLUSIONS: In a systematic review and meta-analysis, we found an increased risk of IBD in patients with CeD and increased risk of CeD in patients with IBD, compared with other patient populations. High-quality prospective cohort studies are needed to assess the risk of CeD-specific and IBD-specific biomarkers in patients with IBD and CeD.


Assuntos
Doença Celíaca/epidemiologia , Colite Ulcerativa/epidemiologia , Doença de Crohn/epidemiologia , Mucosa Intestinal/imunologia , Autoanticorpos/sangue , Autoanticorpos/imunologia , Estudos de Casos e Controles , Doença Celíaca/sangue , Doença Celíaca/imunologia , Colite Ulcerativa/sangue , Colite Ulcerativa/complicações , Colite Ulcerativa/imunologia , Doença de Crohn/sangue , Doença de Crohn/complicações , Doença de Crohn/imunologia , Proteínas de Ligação ao GTP/imunologia , Humanos , Imunoglobulina A/sangue , Imunoglobulina A/imunologia , Prevalência , Proteína 2 Glutamina gama-Glutamiltransferase , Fatores de Risco , Saccharomyces/imunologia , Transglutaminases/imunologia
17.
Clin Gastroenterol Hepatol ; 19(11): 2343-2352.e8, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-32827724

RESUMO

BACKGROUND & AIMS: Many patients with irritable bowel syndrome (IBS) perceive that their symptoms are triggered by wheat-containing foods. We assessed symptoms and gastrointestinal transit before and after a gluten-free diet (GFD) in unselected patients with IBS and investigated biomarkers associated with symptoms. METHODS: We performed a prospective study of 50 patients with IBS (ROME III, all subtypes), with and without serologic reactivity to gluten (antigliadin IgG and IgA), and 25 healthy subjects (controls) at a university hospital in Hamilton, Ontario, Canada, between 2012 and 2016. Gastrointestinal transit, gut symptoms, anxiety, depression, somatization, dietary habits, and microbiota composition were studied before and after 4 weeks of a GFD. HLA-DQ2/DQ8 status was determined. GFD compliance was assessed by a dietitian and by measuring gluten peptides in stool. RESULTS: There was no difference in symptoms among patients at baseline, but after the GFD, patients with antigliadin IgG and IgA reported less diarrhea than patients without these antibodies (P = .03). Compared with baseline, IBS symptoms improved in 18 of 24 patients (75%) with antigliadin IgG and IgA and in 8 of 21 patients (38%) without the antibodies. Although constipation, diarrhea, and abdominal pain were reduced in patients with antigliadin IgG and IgA, only pain decreased in patients without these antibodies. Gastrointestinal transit normalized in a higher proportion of patients with antigliadin IgG and IgA. Anxiety, depression, somatization, and well-being increased in both groups. The presence of antigliadin IgG was associated with overall reductions in symptoms (adjusted odds ratio compared with patients without this antibody, 128.9; 95% CI, 1.16-1427.8; P = .04). Symptoms were reduced even in patients with antigliadin IgG and IgA who reduced gluten intake but were not strictly compliant with the GFD. In controls, a GFD had no effect on gastrointestinal symptoms or gut function. CONCLUSIONS: Antigliadin IgG can be used as a biomarker to identify patients with IBS who might have reductions in symptoms, particularly diarrhea, on a GFD. Larger studies are needed to validate these findings. ClinicalTrials.gov: NCT03492333.


Assuntos
Doença Celíaca , Síndrome do Intestino Irritável , Diarreia , Dieta Livre de Glúten , Humanos , Imunoglobulina G , Estudos Prospectivos
18.
Clin Gastroenterol Hepatol ; 19(2): 391-393, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33059041

RESUMO

The World Health Organization declared coronavirus disease-2019 (COVID-19) a global pandemic in March 2020. Since then, there are more than 34 million cases of COVID-19 leading to more than 1 million deaths worldwide. Numerous studies suggest that celiac disease (CeD), a chronic immune-mediated gastrointestinal condition triggered by gluten, is associated with an increased risk of respiratory infections.1-3 However, how it relates to the risk of COVID-19 is unknown. To address this gap, we conducted a cross-sectional study to evaluate whether patients with self-reported CeD are at an increased risk of contracting COVID-19.


Assuntos
COVID-19/epidemiologia , Doença Celíaca/epidemiologia , Adulto , Estudos de Casos e Controles , Doença Celíaca/dietoterapia , Doença Celíaca/fisiopatologia , Dieta Livre de Glúten , Feminino , Humanos , Masculino , Razão de Chances , Fatores de Risco , SARS-CoV-2 , Inquéritos e Questionários
19.
Am J Gastroenterol ; 116(5): 943-945, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33767100

RESUMO

ABSTRACT: Nonceliac gluten sensitivity, or the more preferred term, nonceliac wheat sensitivity (NCWS), is a heterogenous condition that is diagnosed purely on the basis of symptoms and without an understanding of disease mechanisms and triggers. Biomarkers to identify patients and implementation of dietary treatment in a personalized manner are needed. Mansueto et al. identified a population of NCWS patients with associated autoimmune markers and immune activation. The presence of these markers could be used, in combination with other serological tests, to help develop better diagnostic strategies for NCWS.


Assuntos
Hipersensibilidade a Trigo , Autoimunidade , Biomarcadores , Humanos , Hipersensibilidade a Trigo/diagnóstico
20.
Appl Soft Comput ; 101: 107039, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33519324

RESUMO

Virus diseases are a continued threat to human health in both community and healthcare settings. The current virus disease COVID-19 outbreak raises an unparalleled public health issue for the world at large. Wuhan is the city in China from where this virus came first and, after some time the whole world was affected by this severe disease. It is a challenge for every country's people and higher authorities to fight with this battle due to the insufficient number of resources. On-going assessment of the epidemiological features and future impacts of the COVID-19 disease is required to stay up-to-date of any changes to its spread dynamics and foresee needed resources and consequences in different aspects as social or economic ones. This paper proposes a prediction model of confirmed and death cases of COVID-19. The model is based on a deep learning algorithm with two long short-term memory (LSTM) layers. We consider the available infection cases of COVID-19 in India from January 22, 2020, till October 9, 2020, and parameterize the model. The proposed model is an inference to obtain predicted coronavirus cases and deaths for the next 30 days, taking the data of the previous 260 days of duration of the pandemic. The proposed deep learning model has been compared with other popular prediction methods (Support Vector Machine, Decision Tree and Random Forest) showing a lower normalized RMSE. This work also compares COVID-19 with other previous diseases (SARS, MERS, h1n1, Ebola, and 2019-nCoV). Based on the mortality rate and virus spread, this study concludes that the novel coronavirus (COVID-19) is more dangerous than other diseases.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA