Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
medRxiv ; 2024 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-37693606

RESUMEN

The prevalence of childhood obesity is increasing worldwide, along with the associated common comorbidities of type 2 diabetes and cardiovascular disease in later life. Motivated by evidence for a strong genetic component, our prior genome-wide association study (GWAS) efforts for childhood obesity revealed 19 independent signals for the trait; however, the mechanism of action of these loci remains to be elucidated. To molecularly characterize these childhood obesity loci we sought to determine the underlying causal variants and the corresponding effector genes within diverse cellular contexts. Integrating childhood obesity GWAS summary statistics with our existing 3D genomic datasets for 57 human cell types, consisting of high-resolution promoter-focused Capture-C/Hi-C, ATAC-seq, and RNA-seq, we applied stratified LD score regression and calculated the proportion of genome-wide SNP heritability attributable to cell type-specific features, revealing pancreatic alpha cell enrichment as the most statistically significant. Subsequent chromatin contact-based fine-mapping was carried out for genome-wide significant childhood obesity loci and their linkage disequilibrium proxies to implicate effector genes, yielded the most abundant number of candidate variants and target genes at the BDNF, ADCY3, TMEM18 and FTO loci in skeletal muscle myotubes and the pancreatic beta-cell line, EndoC-BH1. One novel implicated effector gene, ALKAL2 - an inflammation-responsive gene in nerve nociceptors - was observed at the key TMEM18 locus across multiple immune cell types. Interestingly, this observation was also supported through colocalization analysis using expression quantitative trait loci (eQTL) derived from the Genotype-Tissue Expression (GTEx) dataset, supporting an inflammatory and neurologic component to the pathogenesis of childhood obesity. Our comprehensive appraisal of 3D genomic datasets generated in a myriad of different cell types provides genomic insights into pediatric obesity pathogenesis.

2.
Lancet Gastroenterol Hepatol ; 9(2): 133-146, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38104569

RESUMEN

BACKGROUND: Many patients with moderately to severely active Crohn's disease do not respond to available therapies or lose response over time. The GALAXI-1 study previously found that three intravenous guselkumab dosages showed superior clinical and endoscopic outcomes over placebo at week 12 in patients with moderately to severely active Crohn's disease. We report the safety and efficacy of subcutaneous guselkumab maintenance regimens to week 48 in the GALAXI-1 study. METHODS: We did a phase 2, randomised, multicentre, double-blind trial. Adult patients with moderately to severely active Crohn's disease were randomly allocated with a computer-generated randomisation schedule to receive one of five treatment groups, with regimens consisting of an intravenous induction phase transitioning to a subcutaneous maintenance phase starting at week 12 in a treat-through design: (1) guselkumab 200→100 mg group (200 mg intravenous at weeks 0, 4, and 8, then 100 mg subcutaneous every 8 weeks; (2) guselkumab 600→200 mg group (600 mg intravenous at weeks 0, 4, and 8, then 200 mg subcutaneous every 4 weeks); (3) guselkumab 1200→200 mg group (1200 mg intravenous at weeks 0, 4, and 8, then 200 mg subcutaneous every 4 weeks); (4) ustekinumab group (approximately 6 mg/kg intravenous at week 0, then 90 mg subcutaneous every 8 weeks); or (5) placebo group (placebo induction followed by either placebo maintenance [for those with CDAI clinical response at week 12] or crossover to ustekinumab [for those without CDAI clinical response at week 12]). Endpoints assessed at week 48 included CDAI remission (CDAI score <150), endoscopic response (≥50% improvement from baseline in SES-CD or SES-CD score ≤2), and endoscopic remission (SES-CD score ≤2) in the primary efficacy analysis population of all randomised patients who received at least one dose of study drug, excluding those discontinued during a temporary study pause. Safety analyses included all randomised patients who received at least one study drug dose. This trial is registered at Clinical Trials.gov (NCT03466411) and is active but not recruiting. FINDINGS: Among 700 patients screened, 309 (112 biologic-naive; 197 biologic-experienced) were included in the primary efficacy analysis population: 61 in the guselkumab 200→100 mg group, 63 in the guselkumab 600→200 mg group, 61 in the guselkumab 1200→200 mg group, 63 in the ustekinumab group, and 61 in the placebo group. 126 (41%) women and 183 (59%) men were included, with median age 36·0 years (IQR 28·0-49·0). At week 48, the numbers of patients with CDAI clinical remission were 39 (64%) in the guselkumab 200→100 mg group, 46 (73%) in the guselkumab 600→200 mg group, 35 (57%) in the guselkumab 1200→200 mg group, and 37 (59%) in the ustekinumab group. The corresponding numbers of patients with endoscopic response were 27 (44%), 29 (46%), 27 (44%), and 19 (30%), respectively, and endoscopic remission was seen in 11 (18%), 11 (17%), 20 (33%), and four (6%) patients, respectively. In the placebo group, 15 patients were in CDAI clinical response at week 12 and continued placebo; of these, nine (60%) were in clinical remission at week 48. 44 patients in the placebo group were not in CDAI clinical response at week 12 and crossed over to ustekinumab; of these, 26 (59%) were in clinical remission at week 48. Up to week 48, adverse events frequencies in the safety population (n=360) were 46 (66%) of 70 patients (464·9 events per 100 patient-years of follow-up) in the placebo group, 163 (74%) of 220 patients (353·1 per 100 patient-years) in the three guselkumab groups combined, and 60 (85%) of 71 patients (350·7 per 100 patient-years) in the ustekinumab group. Among patients treated with guselkumab or ustekinumab, the most frequently reported infections up to week 48 were nasopharyngitis (25 [11%] of 220 guselkumab recipients, 12 [11%] of 114 ustekinumab recipients) and upper respiratory infections (13 [6%] guselkumab recipients, eight [7%] ustekinumab recipients). After week 12, one patient who responded to placebo induction and two guselkumab-treated patients had serious infections. No active tuberculosis, opportunistic infections, or deaths occurred. INTERPRETATION: Patients receiving guselkumab intravenous induction and subcutaneous maintenance treatment achieved high rates of clinical and endoscopic efficacy up to week 48. No new safety concerns were identified. FUNDING: Janssen Research & Development.


Asunto(s)
Productos Biológicos , Enfermedad de Crohn , Masculino , Adulto , Humanos , Femenino , Ustekinumab/uso terapéutico , Enfermedad de Crohn/terapia , Anticuerpos Monoclonales Humanizados/uso terapéutico , Productos Biológicos/uso terapéutico
3.
Gastro Hep Adv ; 2(6): 830-842, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37736163

RESUMEN

BACKGROUND AND AIMS: A key histopathological feature of inflammatory bowel disease is damage to the mucosa, including breakdown of the epithelial barrier. Human enteroids and colonoids are a critical bench-to-bedside tool for studying the epithelium in inflammatory bowel disease. The goal of the current study was to define transcriptional differences in healthy versus diseased subjects that are sustained in enteroids and colonoids, including from disease-spared tissue. METHODS: Biopsies and matching enteroid or colonoid cultures from pediatric patients with ileal Crohn disease (N = 6) and control subjects (N = 17) were subjected to RNA sequencing followed by bioinformatic and machine learning analyses. Late passage enteroids were exposed to cytokines to assess durable transcriptional differences. RESULTS: We observed substantial overlap of pathways upregulated in Crohn disease in enteroids and ileal biopsies, as well as colonoids and rectal biopsies. KEGG pathways for cytokine-cytokine receptor interaction, chemokine signaling, protein export, and Toll-like receptor signaling were upregulated in both ileal and rectal biopsies, as well as enteroids and colonoids. In vitro cytokine exposure reactivated genes previously increased in biopsies. Machine learning predicted biopsy location (100% accuracy) and donor disease status (83% accuracy). A random forest classifier generated using ileal enteroids identified rectal colonoids from ileal Crohn disease subjects with 80% accuracy. CONCLUSION: We confirmed transcriptional profiles of Crohn disease biopsies are expressed in enteroids and colonoids. Furthermore, transcriptomic data from disease-spared rectal tissue can identify patients with ileal Crohn disease. Our data support the use of patient enteroids and colonoids as critical translational tools for the study of inflammatory bowel disease.

4.
PLoS One ; 18(3): e0282267, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36862717

RESUMEN

BACKGROUND: Randomized trials are the gold-standard for clinical evidence generation, but they can sometimes be limited by infeasibility and unclear generalizability to real-world practice. External control arm (ECA) studies may help address this evidence gaps by constructing retrospective cohorts that closely emulate prospective ones. Experience in constructing these outside the context of rare diseases or cancer is limited. We piloted an approach for developing an ECA in Crohn's disease using electronic health records (EHR) data. METHODS: We queried EHR databases and manually screened records at the University of California, San Francisco to identify patients meeting the eligibility criteria of TRIDENT, a recently completed interventional trial involving an ustekinumab reference arm. We defined timepoints to balance missing data and bias. We compared imputation models by their impacts on cohort membership and outcomes. We assessed the accuracy of algorithmic data curation against manual review. Lastly, we assessed disease activity following treatment with ustekinumab. RESULTS: Screening identified 183 patients. 30% of the cohort had missing baseline data. Nonetheless, cohort membership and outcomes were robust to the method of imputation. Algorithms for ascertaining non-symptom-based elements of disease activity using structured data were accurate against manual review. The cohort consisted of 56 patients, exceeding planned enrollment in TRIDENT. 34% of the cohort was in steroid-free remission at week 24. CONCLUSION: We piloted an approach for creating an ECA in Crohn's disease from EHR data by using a combination of informatics and manual methods. However, our study reveals significant missing data when standard-of-care clinical data are repurposed. More work will be needed to improve the alignment of trial design with typical patterns of clinical practice, and thereby enable a future of more robust ECAs in chronic diseases like Crohn's disease.


Asunto(s)
Enfermedad de Crohn , Ustekinumab , Humanos , Ustekinumab/uso terapéutico , Enfermedad de Crohn/tratamiento farmacológico , Proyectos Piloto , Registros Electrónicos de Salud , Estudios Prospectivos , Estudios Retrospectivos
5.
Gut Microbes ; 14(1): 2083417, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35658830

RESUMEN

Complications of short bowel syndrome (SBS) include malabsorption and bacterial overgrowth, requiring prolonged dependence on parenteral nutrition (PN). We hypothesized that the intolerance of whole food in some SBS patients might be due to the effect of dietary fiber on the gut microbiome. Shotgun metagenomic sequencing and targeted metabolomics were performed using biospecimens collected from 55 children with SBS and a murine dietary fiber model. Bioinformatic analyses were performed on these datasets as well as from a healthy human dietary intervention study. Compared to healthy controls, the gut microbiota in SBS had lower diversity and increased Proteobacteria, a pattern most pronounced in children on PN and inversely correlated with whole food consumption. Whole food intake correlated with increased glycoside hydrolases (GH) and bile salt hydrolases (BSH) with reduced fecal conjugated bile acids suggesting that dietary fiber regulates BSH activity via GHs. Mechanistic evidence supporting this notion was generated via fecal and plasma bile acid profiling in a healthy human fiber-free dietary intervention study as well as in a dietary fiber mouse experiment. Gaussian mixture modeling of fecal bile acids was used to identify three clinically relevant SBS phenotypes. Dietary fiber is associated with bile acid deconjugation likely via an interaction between gut microbiota BSHs and GHs in the small intestine, which may lead to whole food intolerance in patients with SBS. This mechanism not only has potential utility in clinical phenotyping and targeted therapeutics in SBS based on bile acid metabolism but may have relevance to other intestinal disease states.


Asunto(s)
Microbioma Gastrointestinal , Amidohidrolasas/metabolismo , Animales , Ácidos y Sales Biliares , Fibras de la Dieta , Microbioma Gastrointestinal/fisiología , Humanos , Ratones
6.
Gastroenterology ; 161(6): 1940-1952, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34529988

RESUMEN

BACKGROUND & AIMS: Significant progress has been made since the first report of inflammatory bowel disease (IBD) in 1859, after decades of research that have contributed to the understanding of the genetic and environmental factors involved in IBD pathogenesis. Today, a range of treatments is available for directed therapy, mostly targeting the overactive immune response. However, the mechanisms by which the immune system contributes to disease pathogenesis and progression are not fully understood. One challenge hindering IBD research is the heterogeneous nature of the disease and the lack of understanding of how immune cells interact with one another in the gut mucosa. Introduction of a technology that enables expansive characterization of the inflammatory environment of human IBD tissues may address this gap in knowledge. METHODS: We used the imaging mass cytometry platform to perform highly multiplex image analysis of IBD and healthy deidentified intestine sections (6 Crohn's disease compared to 6 control ileum; 6 ulcerative colitis compared to 6 control colon). The acquired images were graded for inflammation severity by analysis of adjacent H&E tissue sections. We assigned more than 300,000 cells to unique cell types and performed analyses of tissue integrity, epithelial activity, and immune cell composition. RESULTS: The intestinal epithelia of patients with IBD exhibited increased proliferation rates and expression of HLA-DR compared to control tissues, and both features were positively correlated with the severity of inflammation. The neighborhood analysis determined enrichment of regulatory T cell interactions with CD68+ macrophages, CD4+ T cells, and plasma cells in both forms of IBD, whereas activated lysozyme C+ macrophages were preferred regulatory T cell neighbors in Crohn's disease but not ulcerative colitis. CONCLUSIONS: Altogether, our study shows the power of imaging mass cytometry and its ability to both quantify immune cell types and characterize their spatial interactions within the inflammatory environment by a single analysis platform.


Asunto(s)
Microambiente Celular , Colitis Ulcerosa/patología , Colon/patología , Enfermedad de Crohn/patología , Células Epiteliales/patología , Mucosa Intestinal/patología , Microscopía Confocal , Adolescente , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Biomarcadores/metabolismo , Linfocitos T CD8-positivos , Estudios de Casos y Controles , Comunicación Celular , Proliferación Celular , Niño , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/metabolismo , Colon/inmunología , Colon/metabolismo , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Antígenos HLA-DR/metabolismo , Humanos , Procesamiento de Imagen Asistido por Computador , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Muramidasa/metabolismo , Proteoma , Proteómica , Índice de Severidad de la Enfermedad , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
7.
J Crohns Colitis ; 15(11): 1931-1942, 2021 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-34037715

RESUMEN

BACKGROUND AND AIMS: The objective was to evaluate the pharmacokinetics, safety/tolerability, and efficacy of ustekinumab in children with moderately to severely active Crohn's disease. METHODS: In this Phase 1, multicentre, 16-week, double-blind, induction dose-ranging study [NCT02968108], patients aged 2-<18 years [body weight ≥10 kg] were randomised [1:1] to one of two weight range-based intravenous induction doses: 130 mg vs 390 mg in patients ≥40kg and 3 mg/kg vs 9 mg/kg in patients <40kg. At Week 8, all patients received a single subcutaneous ustekinumab maintenance dose of 90 mg in patients ≥40kg or 2 mg/kg in patients <40kg. RESULTS: A total of 44 patients were randomised and treated with ustekinumab [n = 23 lower dose; n = 21 higher dose]; median [interquartile range] age was 13.0 [12-16] years. Pharmacokinetics were similar to those in adults with Crohn's disease. However, serum ustekinumab concentrations were lower among those with body weight <40 kg compared with patients ≥40 kg and the reference Phase 3 adult population. Through Week 16, 73% of patients reported ≥1 adverse event [82.6% lower vs 62% higher dose]; two discontinued due to adverse events [one in each group]. Serious adverse events occurred in 16% [26% lower, 5% higher dose], with Crohn's disease exacerbation being the most frequent. At Week 16, 22%/29% [lower/higher dose] achieved clinical remission [Paediatric Crohn's Disease Activity Index ≤10]. CONCLUSIONS: The pharmacokinetics/safety profiles were generally consistent with those observed in adults with Crohn's disease. These results suggest a different dosing regimen may be required for patients <40 kg from that employed in this study; additional pharmacokinetic analyses may be needed in this population.


Asunto(s)
Enfermedad de Crohn/tratamiento farmacológico , Ustekinumab/farmacología , Ustekinumab/farmacocinética , Administración Intravenosa , Adolescente , Anticuerpos Monoclonales/farmacocinética , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Niño , Método Doble Ciego , Femenino , Humanos , Inyecciones Subcutáneas , Masculino , Seguridad del Paciente/normas , Seguridad del Paciente/estadística & datos numéricos , Pediatría/métodos , Pediatría/tendencias , Resultado del Tratamiento , Ustekinumab/uso terapéutico
8.
Cell Mol Gastroenterol Hepatol ; 11(3): 667-682, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33069917

RESUMEN

BACKGROUND & AIMS: Inflammatory bowel disease (IBD) is a polygenic disorder characterized principally by dysregulated inflammation impacting the gastrointestinal tract. However, there also is increasing evidence for a clinical association with stress and depression. Given the role of the hypothalamus in stress responses and in the pathogenesis of depression, useful insights could be gleaned from understanding its genetic role in IBD. METHODS: We conducted genetic correlation analyses on publicly available genome-wide association study summary statistics for depression and IBD traits to identify genetic commonalities. We used partitioned linkage disequilibrium score regression, leveraging our ATAC sequencing and promoter-focused Capture C data, to measure enrichment of IBD single-nucleotide polymorphisms within promoter-interacting open chromatin regions of human embryonic stem cell-derived hypothalamic-like neurons (HNs). Using the same data sets, we performed variant-to-gene mapping to implicate putative IBD effector genes in HNs. To contrast these results, we similarly analyzed 3-dimensional genomic data generated in epithelium-derived colonoids from rectal biopsy specimens from donors without pathologic disease noted at the time of colonoscopy. Finally, we conducted enrichment pathway analyses on the implicated genes to identify putative IBD dysfunctional pathways. RESULTS: We found significant genetic correlations (rg) of 0.122 with an adjusted P (Padj) = 1.4 × 10-4 for IBD: rg = 0.122; Padj = 2.5 × 10-3 for ulcerative colitis and genetic correlation (rg) = 0.094; Padj = 2.5 × 10-3 for Crohn's disease, and significant approximately 4-fold (P = .005) and approximately 7-fold (P = .03) enrichment of IBD single-nucleotide polymorphisms in HNs and colonoids, respectively. We implicated 25 associated genes in HNs, among which CREM, CNTF, and RHOA encode key regulators of stress. Seven genes also additionally were implicated in the colonoids. We observed an overall enrichment for immune and hormonal signaling pathways, and a colonoid-specific enrichment for microbiota-relevant terms. CONCLUSIONS: Our results suggest that the hypothalamus warrants further study in the context of IBD pathogenesis.


Asunto(s)
Depresión/genética , Predisposición Genética a la Enfermedad , Hipotálamo/fisiopatología , Enfermedades Inflamatorias del Intestino/genética , Estrés Psicológico/genética , Eje Cerebro-Intestino , Estudios de Casos y Controles , Mapeo Cromosómico , Conjuntos de Datos como Asunto , Depresión/fisiopatología , Estudio de Asociación del Genoma Completo , Humanos , Sistema Hipotálamo-Hipofisario/fisiopatología , Hipotálamo/citología , Enfermedades Inflamatorias del Intestino/fisiopatología , Desequilibrio de Ligamiento , Neuronas , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Estrés Psicológico/fisiopatología
9.
PLoS One ; 15(5): e0231891, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32433684

RESUMEN

OBJECTIVE: The SI gene encodes the sucrase-isomaltase enzyme, a disaccharidase expressed in the intestinal brush border. Hypomorphic SI variants cause recessive congenital sucrase-isomaltase deficiency (CSID) and related gastrointestinal (GI) symptoms. Among children presenting with chronic, idiopathic loose stools, we assessed the prevalence of CSID-associated SI variants relative to the general population and the relative GI symptom burden associated with SI genotype within the study population. METHODS: A prospective study conducted at 18 centers enrolled 308 non-Hispanic white children ≤18 years old who were experiencing chronic, idiopathic, loose stools at least once per week for >4 weeks. Data on demographics, GI symptoms, and genotyping for 37 SI hypomorphic variants were collected. Race/ethnicity-matched SI data from the Exome Aggregation Consortium (ExAC) database was used as the general population reference. RESULTS: Compared with the general population, the cumulative prevalence of hypomorphic SI variants was significantly higher in the study population (4.5% vs. 1.3%, P < .01; OR = 3.5 [95% CI: 6.1, 2.0]). Within the study population, children with a hypomorphic SI variant had a more severe GI symptom burden than those without, including: more frequent episodes of loose stools (P < .01), higher overall stool frequency (P < .01), looser stool form (P = .01) and increased flatulence (P = .02). CONCLUSION: Non-Hispanic white children with chronic idiopathic loose stools have a higher prevalence of CSID-associated hypomorphic SI variants than the general population. The GI symptom burden was greater among the study subjects with a hypomorphic SI variant than those without hypomorphic SI variants.


Asunto(s)
Errores Innatos del Metabolismo de los Carbohidratos/patología , Complejo Sacarasa-Isomaltasa/deficiencia , Complejo Sacarasa-Isomaltasa/genética , Adolescente , Errores Innatos del Metabolismo de los Carbohidratos/epidemiología , Errores Innatos del Metabolismo de los Carbohidratos/genética , Niño , Bases de Datos Factuales , Femenino , Genotipo , Heterocigoto , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Prevalencia , Estudios Prospectivos
10.
Artículo en Inglés | MEDLINE | ID: mdl-31344510

RESUMEN

Despite growing literature characterizing the fecal microbiome and its association with health and disease, few studies have analyzed the microbiome of the small intestine. Here, we examine what is known about the human small intestinal microbiota in terms of community structure and functional properties. We examine temporal dynamics of select bacterial populations in the small intestine, and the effects of dietary carbohydrates and fats on shaping these populations. We then evaluate dysbiosis in the small intestine in several human disease models, including small intestinal bacterial overgrowth, short-bowel syndrome, pouchitis, environmental enteric dysfunction, and irritable bowel syndrome. What is clear is that the bacterial biology, and mechanisms of bacteria-induced pathophysiology, are enormously broad and elegant in the small intestine. Studying the small intestinal microbiota is challenged by rapidly fluctuating environmental conditions in these intestinal segments, as well as the complexity of sample collection and bioinformatic analysis. Because the functionality of the digestive tract is determined primarily by the small intestine, efforts must be made to better characterize this unique and important microbial ecosystem.


Asunto(s)
Disbiosis/microbiología , Conducta Alimentaria/fisiología , Microbioma Gastrointestinal/fisiología , Mucosa Intestinal/microbiología , Intestino Delgado/microbiología , Animales , Síndrome del Asa Ciega/microbiología , Síndrome del Asa Ciega/fisiopatología , Carbohidratos de la Dieta/administración & dosificación , Grasas de la Dieta/administración & dosificación , Modelos Animales de Enfermedad , Disbiosis/complicaciones , Disbiosis/fisiopatología , Humanos , Síndrome del Colon Irritable/microbiología , Síndrome del Colon Irritable/fisiopatología , Reservoritis/microbiología , Reservoritis/fisiopatología , Síndrome del Intestino Corto/microbiología , Síndrome del Intestino Corto/fisiopatología
11.
Dev Cell ; 49(1): 10-29, 2019 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-30930166

RESUMEN

Single-cell gene expression analyses of mammalian tissues have uncovered profound stage-specific molecular regulatory phenomena that have changed the understanding of unique cell types and signaling pathways critical for lineage determination, morphogenesis, and growth. We discuss here the case for a Pediatric Cell Atlas as part of the Human Cell Atlas consortium to provide single-cell profiles and spatial characterization of gene expression across human tissues and organs. Such data will complement adult and developmentally focused HCA projects to provide a rich cytogenomic framework for understanding not only pediatric health and disease but also environmental and genetic impacts across the human lifespan.


Asunto(s)
Desarrollo Embrionario/genética , Redes Reguladoras de Genes/genética , Pediatría/tendencias , Análisis de la Célula Individual/métodos , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica/genética , Humanos , Distribución Tisular/genética
12.
BMC Pediatr ; 19(1): 72, 2019 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-30849955

RESUMEN

BACKGROUND: Total parenteral nutrition (TPN) and biliary atresia (BA) are common causes of cholestasis in infancy. The diagnosis of BA is time sensitive due to an inverse correlation between age at intervention (hepatic portoenterostomy - HPE) and survival without liver transplantation. Clinical, laboratory, and histologic features of BA and parenteral nutrition associated cholestasis (PNAC) are similar, creating a diagnostic dilemma for cholestatic infants on parenteral nutrition. There is limited published information about the natural history of PNAC including time to resolution, or diagnostic tests that distinguish BA from other etiologies of cholestasis. CASE PRESENTATION: We present a case of a child diagnosed with BA whose cholestasis began while receiving TPN. His clinical course was notable for transient resolution of his cholestasis after stopping parenteral nutrition and ultimate intraoperative diagnosis. CONCLUSIONS: Clinicians who care for patients who frequently receive TPN should be aware that clinical, laboratory, imaging, and biopsy findings can be similar between BA and PNAC.


Asunto(s)
Atresia Biliar/diagnóstico , Hígado/patología , Nutrición Parenteral Total/efectos adversos , Atresia Biliar/complicaciones , Bilirrubina/sangre , Colestasis/etiología , Diagnóstico Diferencial , Humanos , Hiperbilirrubinemia/etiología , Lactante , Masculino
13.
Am J Physiol Gastrointest Liver Physiol ; 315(4): G580-G591, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29953253

RESUMEN

Infants with congenital diarrheal disorders caused by enteroendocrine cell dysgenesis, or the loss of intestinal endocrine cells, causes severe malabsorptive diarrhea, though the mechanism is not fully understood. The transcription factor "aristaless-related homeobox" (Arx) is specifically expressed in intestinal endocrine cells. This study seeks to characterize the early malabsorptive phenotype of mice deficient for Arx using cell-type specific gene ablation in Villin-Cre; ArxloxP/Y ( Arxint) mice. In neonatal mice, the loss of intestinal Arx caused the loss of intestinal hormones, such as cholecystokinin, secretin, neurotensin, glucose-dependent insulinotropic peptide, glucagon-like peptide (GLP)-1 and GLP-2 but also upregulation of somatostatin. Arxint mice exhibited steatorrhea with the loss of lipid transport in duodenal enterocytes, upregulation of lysozyme-positive Paneth cells, and a secondary increase in antimicrobial peptides, specifically Reg3ß. When the epithelium from Arxint mice was cultured ex vivo into enteroids, however, the Reg3ß upregulation was lost under the sterile conditions. Thus, Arx is required for the appropriate lineage allocation of multiple enteroendocrine subtypes. We concluded that altered hormonal signaling caused by Arx deficiency results in lipid malabsorption, premature Paneth cell differentiation, and an inflammatory response, including neutrophilic infiltrates and a microbiota-triggered upregulation of Reg3ß. NEW & NOTEWORTHY The enteroendocrine transcription factor aristaless-related homeobox (Arx) plays a key role in lineage specification. Changes in hormonal expression mediated by Arx lead to lipid malabsorption and premature Paneth cell development. Furthermore, global profiling of whole intestine from Arx-deficient mice revealed significant upregulation of antimicrobial peptides. This antimicrobial response in Arx-deficient animals is lost under sterile culture conditions of enteroids.


Asunto(s)
Diarrea/metabolismo , Hormonas Gastrointestinales/metabolismo , Microbioma Gastrointestinal , Intestino Delgado/metabolismo , Síndromes de Malabsorción/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Diarrea/congénito , Enterocitos/citología , Enterocitos/metabolismo , Células Enteroendocrinas/citología , Células Enteroendocrinas/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Intestino Delgado/citología , Intestino Delgado/microbiología , Ratones , Ratones Endogámicos C57BL , Proteínas Asociadas a Pancreatitis/genética , Proteínas Asociadas a Pancreatitis/metabolismo , Células de Paneth/citología , Células de Paneth/metabolismo , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
14.
Handb Exp Pharmacol ; 239: 319-342, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28035530

RESUMEN

Serotonin (5-hydroxytryptamine; 5-HT) is best known as a neurotransmitter critical for central nervous system (CNS) development and function. 95% of the body's serotonin, however, is produced in the intestine where it has been increasingly recognized for its hormonal, autocrine, paracrine, and endocrine actions. This chapter provides the most current knowledge of the critical autocrine and paracrine roles of 5-HT in intestinal motility and inflammation as well as its function as a hormone in osteocyte homeostasis. Therapeutic applications in each of these areas are also discussed.


Asunto(s)
Sistema Nervioso Entérico/metabolismo , Tracto Gastrointestinal/inervación , Enfermedades Inflamatorias del Intestino/metabolismo , Neuronas Serotoninérgicas/metabolismo , Serotonina/metabolismo , Animales , Antiinflamatorios/uso terapéutico , Remodelación Ósea , Sistema Nervioso Entérico/efectos de los fármacos , Sistema Nervioso Entérico/fisiopatología , Fármacos Gastrointestinales/uso terapéutico , Motilidad Gastrointestinal , Tracto Gastrointestinal/efectos de los fármacos , Humanos , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/fisiopatología , Neurogénesis , Osteocitos/metabolismo , Osteoporosis/metabolismo , Osteoporosis/fisiopatología , Receptores de Serotonina/metabolismo , Serotoninérgicos/uso terapéutico , Transducción de Señal
15.
J Pediatr Gastroenterol Nutr ; 60(2): 192-9, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25171319

RESUMEN

OBJECTIVES: Severe congenital diarrhea occurs in approximately half of patients with Aristaless-Related Homeobox (ARX) null mutations. The cause of this diarrhea is unknown. In a mouse model of intestinal Arx deficiency, the prevalence of a subset of enteroendocrine cells is altered, leading to diarrhea. Because polyalanine expansions within the ARX protein are the most common mutations found in ARX-related disorders, we sought to characterize the enteroendocrine population in human tissue of an ARX mutation and in a mouse model of the corresponding polyalanine expansion (Arx). METHODS: Immunohistochemistry and quantitative real-time polymerase chain reaction were the primary modalities used to characterize the enteroendocrine populations. Daily weights were determined for the growth curves, and Oil-Red-O staining on stool and tissue identified neutral fats. RESULTS: An expansion of 7 alanines in the first polyalanine tract of both human ARX and mouse Arx altered enteroendocrine differentiation. In human tissue, cholecystokinin, glucagon-like peptide 1, and somatostatin populations were reduced, whereas the chromogranin A population was unchanged. In the mouse model, cholecystokinin and glucagon-like peptide 1 populations were also lost, although the somatostatin-expressing population was increased. The ARX protein was present in human tissue, whereas the Arx protein was degraded in the mouse intestine. CONCLUSIONS: ARX/Arx is required for the specification of a subset of enteroendocrine cells in both humans and mice. Owing to protein degradation, the Arx mouse recapitulates findings of the intestinal Arx null model, but is not able to further the study of the differential effects of the ARX protein on its transcriptional targets in the intestine.


Asunto(s)
Diarrea/genética , Enfermedades Duodenales/genética , Células Enteroendocrinas/fisiología , Proteínas de Homeodominio/genética , Seudoobstrucción Intestinal/genética , Péptidos/metabolismo , Factores de Transcripción/genética , Adolescente , Animales , Diferenciación Celular/genética , Colecistoquinina/análisis , Cromogranina A/análisis , Diarrea/patología , Modelos Animales de Enfermedad , Enfermedades Duodenales/patología , Duodeno/patología , Células Enteroendocrinas/química , Células Enteroendocrinas/patología , Insuficiencia de Crecimiento/genética , Femenino , Péptido 1 Similar al Glucagón/análisis , Proteínas de Homeodominio/análisis , Humanos , Seudoobstrucción Intestinal/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutagénesis Insercional , Somatostatina/análisis , Esteatorrea/genética , Factores de Transcripción/análisis
16.
Am J Physiol Gastrointest Liver Physiol ; 307(10): G979-91, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25214396

RESUMEN

Enteroendocrine cells secrete over a dozen different hormones responsible for coordinating digestion, absorption, metabolism, and gut motility. Loss of enteroendocrine cells is a known cause of severe congenital diarrhea. Furthermore, enteroendocrine cells regulate glucose metabolism, with the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) playing critical roles in stimulating insulin release by pancreatic ß-cells. Islet1 (Isl1) is a LIM-homeodomain transcription factor expressed specifically in an array of intestinal endocrine cells, including incretin-expressing cells. To examine the impact of intestinal Isl1 on glycemic control, we set out to explore the role of intestinal Isl1 in hormone cell specification and organismal physiology. Mice with intestinal epithelial-specific ablation of Isl1 were obtained by crossing Villin-Cre transgenic animals with mice harboring a Isl1(loxP) allele (Isl1(int) model). Gene ablation of Isl1 in the intestine results in loss of GLP-1, GIP, cholecystokinin (CCK), and somatostatin-expressing cells and an increase in 5-HT (serotonin)-producing cells, while the chromogranin A population was unchanged. This dramatic change in hormonal milieu results in animals with lipid malabsorption and females smaller than their littermate controls. Interestingly, when challenged with oral, not intraperitoneal glucose, the Isl-1 intestinal-deficient animals (Isl1(int)) display impaired glucose tolerance, indicating loss of the incretin effect. Thus the Isl1(int) model confirms that intestinal biology is essential for organism physiology in glycemic control and susceptibility to diabetes.


Asunto(s)
Glucemia/metabolismo , Células Enteroendocrinas/metabolismo , Trastornos del Metabolismo de la Glucosa/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Proteínas con Homeodominio LIM/deficiencia , Factores de Transcripción/deficiencia , Factores de Edad , Animales , Animales Recién Nacidos , Biomarcadores/sangre , Colecistoquinina/metabolismo , Cromogranina A/metabolismo , Diarrea/genética , Diarrea/metabolismo , Grasas de la Dieta/metabolismo , Células Enteroendocrinas/patología , Femenino , Polipéptido Inhibidor Gástrico/metabolismo , Gastrinas/metabolismo , Genotipo , Ghrelina/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Trastornos del Metabolismo de la Glucosa/sangre , Trastornos del Metabolismo de la Glucosa/genética , Prueba de Tolerancia a la Glucosa , Integrasas/genética , Absorción Intestinal , Mucosa Intestinal/patología , Intestino Delgado/patología , Proteínas con Homeodominio LIM/genética , Síndromes de Malabsorción/genética , Síndromes de Malabsorción/metabolismo , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Microfilamentos/genética , Fenotipo , Serotonina/metabolismo , Somatostatina/metabolismo , Factores de Transcripción/genética , Aumento de Peso
17.
PLoS One ; 8(11): e78741, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24236044

RESUMEN

ARX/Arx is a homeodomain-containing transcription factor necessary for the specification and early maintenance of pancreatic endocrine α-cells. Many transcription factors important to pancreas development, including ARX/Arx, are also crucial for proper brain development. Although null mutations of ARX in human patients result in the severe neurologic syndrome XLAG (X-linked lissencephaly associated with abnormal genitalia), the most common mutation is the expansion of the first polyalanine tract of ARX, which results primarily in the clinical syndrome ISSX (infantile spasms). Mouse models of XLAG, ISSX and other human ARX mutations demonstrate a direct genotype-phenotype correlation in ARX-related neurologic disorders. Furthermore, mouse models utilizing a polyalanine tract expansion mutation have illustrated critical developmental differences between null mutations and expansion mutations in the brain, revealing context-specific defects. Although Arx is known to be required for the specification and early maintenance of pancreatic glucagon-producing α-cells, the consequences of the Arx polyalanine expansion on pancreas development remain unknown. Here we report that mice with an expansion mutation in the first polyalanine tract of Arx exhibit impaired α-cell specification and maintenance, with gradual α-cell loss due to apoptosis. This is in contrast to the re-specification of α-cells into ß- and δ-cells that occurs in mice null for Arx. Overall, our analysis of an Arx polyalanine expansion mutation on pancreatic development suggests that impaired α-cell function might also occur in ISSX patients.


Asunto(s)
Apoptosis , Regulación del Desarrollo de la Expresión Génica , Células Secretoras de Glucagón/fisiología , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Animales , Tipificación del Cuerpo , Proteína Doblecortina , Femenino , Expresión Génica , Estudios de Asociación Genética , Glucagón/genética , Glucagón/metabolismo , Proteínas de Homeodominio/metabolismo , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Páncreas/embriología , Páncreas/patología , Péptidos/genética , Espasmos Infantiles/genética , Espasmos Infantiles/patología , Factores de Transcripción/metabolismo
18.
PLoS One ; 8(6): e66214, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23785486

RESUMEN

The specification and differentiation of pancreatic endocrine cell populations (α-, ß-, δ, PP- and ε-cells) is orchestrated by a combination of transcriptional regulators. In the pancreas, Aristaless-related homeobox gene (Arx) is expressed first in the endocrine progenitors and then restricted to glucagon-producing α-cells. While the functional requirement of Arx in early α-cell specification has been investigated, its role in maintaining α-cell identity has yet to be explored. To study this later role of Arx, we have generated mice in which the Arx gene has been ablated specifically in glucagon-producing α-cells. Lineage-tracing studies and immunostaining analysis for endocrine hormones demonstrate that ablation of Arx in neonatal α-cells results in an α-to-ß-like conversion through an intermediate bihormonal state. Furthermore, these Arx-deficient converted cells express ß-cell markers including Pdx1, MafA, and Glut2. Surprisingly, short-term ablation of Arx in adult mice does not result in a similar α-to-ß-like conversion. Taken together, these findings reveal a potential temporal requirement for Arx in maintaining α-cell identity.


Asunto(s)
Eliminación de Gen , Células Secretoras de Glucagón/metabolismo , Proteínas de Homeodominio/genética , Factores de Transcripción/genética , Animales , Animales Recién Nacidos , Biomarcadores/metabolismo , Linaje de la Célula/genética , Femenino , Expresión Génica , Glucagón/genética , Glucagón/metabolismo , Insulina/genética , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Transgénicos
19.
J Pediatr Gastroenterol Nutr ; 56(2): 215-9, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22699838

RESUMEN

OBJECTIVES: Safety and effectiveness of large-volume polyethylene glycol-based solution (PEG-ES) have been documented, but the taste and volume can be barriers to successful colonoscopy preparation. Efficacy and safety of small-volume electrolyte-free (PEG-P) preparation (Miralax) for colonoscopy preparation have been rarely studied, although presently used at many pediatric centers. The primary objective of the present study was to determine whether PEG-P results in a more efficacious and safe colonoscopy preparation as compared with senna. METHODS: The study design was prospective, randomized, and single-blinded. Patients ages 6 to 21 years were randomized to a 2-day clean-out regimen of PEG-P at a dose of 1.5 g/kg divided twice per day for 2 days versus senna 15 mL daily (ages 6-12) or 30 mL daily (ages 12-21) for 2 days. Both preparations required 1 day of clear liquids whereas senna preparation required an additional day of full liquid diet. A blinded endoscopist graded the quality of preparation with a standardized cleanliness tool (Aronchick scale). Serum chemistry panels were obtained. Patients or parents rated symptoms and ease of preparation. The anticipated number of subjects was 166; however, the interim analysis demonstrated inferiority of senna preparation. RESULTS: Thirty patients were evaluated in the present study. Of the patients in the PEG-P arm, 88% (14/16) received an excellent/good score compared with 29% (4/14), with the senna preparation (P = 0.0022). Both preparations were well-tolerated by patient-graded ease of preparation. Demographics and laboratory values did not differ significantly across the 2 groups. No serious adverse events were noted. CONCLUSIONS: PEG-P is an effective colonoscopy preparation whereas senna preparation was insufficient. Both were well-tolerated and appear safe in a pediatric population.


Asunto(s)
Catárticos , Colon , Colonoscopía/métodos , Extractos Vegetales , Polietilenglicoles , Senna , Adolescente , Colon/cirugía , Femenino , Humanos , Masculino , Cooperación del Paciente , Soluciones Farmacéuticas , Polvos
20.
Dev Biol ; 365(1): 175-88, 2012 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-22387004

RESUMEN

Enteroendocrine cells of the gastrointestinal (GI) tract play a central role in metabolism, digestion, satiety and lipid absorption, yet their development remains poorly understood. Here we show that Arx, a homeodomain-containing transcription factor, is required for the normal development of mouse and human enteroendocrine cells. Arx expression is detected in a subset of Neurogenin3 (Ngn3)-positive endocrine progenitors and is also found in a subset of hormone-producing cells. In mice, removal of Arx from the developing endoderm results in a decrease of enteroendocrine cell types including gastrin-, glucagon/GLP-1-, CCK-, secretin-producing cell populations and an increase of somatostatin-expressing cells. This phenotype is also observed in mice with endocrine-progenitor-specific Arx ablation suggesting that Arx is required in the progenitor for enteroendocrine cell development. In addition, depletion of human ARX in developing human intestinal tissue results in a profound deficit in expression of the enteroendocrine cell markers CCK, secretin and glucagon while expression of a pan-intestinal epithelial marker, CDX2, and other non-endocrine markers remained unchanged. Taken together, our findings uncover a novel and conserved role of Arx in mammalian endocrine cell development and provide a potential cause for the chronic diarrhea seen in both humans and mice carrying Arx mutations.


Asunto(s)
Endodermo/embriología , Células Enteroendocrinas/citología , Proteínas de Homeodominio/fisiología , Factores de Transcripción/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Diferenciación Celular , Linaje de la Célula , Endodermo/citología , Humanos , Ratones , Proteínas del Tejido Nervioso/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...