Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
Sci Rep ; 14(1): 11911, 2024 05 24.
Artigo em Inglês | MEDLINE | ID: mdl-38789719

RESUMO

Lack of understanding of the pathophysiology of gastrointestinal (GI) complications in type 1 diabetes (T1D), including altered intestinal transcriptomes and protein expression represents a major gap in the management of these patients. Human enteroids have emerged as a physiologically relevant model of the intestinal epithelium but establishing enteroids from individuals with long-standing T1D has proven difficult. We successfully established duodenal enteroids using endoscopic biopsies from pediatric T1D patients and compared them with aged-matched enteroids from healthy subjects (HS) using bulk RNA sequencing (RNA-seq), and functional analyses of ion transport processes. RNA-seq analysis showed significant differences in genes and pathways associated with cell differentiation and proliferation, cell fate commitment, and brush border membrane. Further validation of these results showed higher expression of enteroendocrine cells, and the proliferating cell marker Ki-67, significantly lower expression of NHE3, lower epithelial barrier integrity, and higher fluid secretion in response to cAMP and elevated calcium in T1D enteroids. Enteroids established from pediatric T1D duodenum identify characteristics of an abnormal intestinal epithelium and are distinct from HS. Our data supports the use of pediatric enteroids as an ex-vivo model to advance studies of GI complications and drug discovery in T1D patients.


Assuntos
Diabetes Mellitus Tipo 1 , Duodeno , Mucosa Intestinal , Humanos , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Diabetes Mellitus Tipo 1/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Criança , Duodeno/metabolismo , Duodeno/patologia , Feminino , Masculino , Proliferação de Células , Adolescente , Células Enteroendócrinas/metabolismo , Células Enteroendócrinas/patologia , Trocador 3 de Sódio-Hidrogênio/metabolismo , Trocador 3 de Sódio-Hidrogênio/genética , Diferenciação Celular , Organoides/metabolismo , Organoides/patologia , Antígeno Ki-67/metabolismo
2.
Cancer Res ; 81(14): 3791-3805, 2021 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-34035083

RESUMO

Despite the connection of secretory cells, including goblet and enteroendocrine (EEC) cells, to distinct mucus-containing colorectal cancer histologic subtypes, their role in colorectal cancer progression has been underexplored. Here, our analysis of The Cancer Genome Atlas (TCGA) and single-cell RNA-sequencing data demonstrates that EEC progenitor cells are enriched in BRAF-mutant colorectal cancer patient tumors, cell lines, and patient-derived organoids. In BRAF-mutant colorectal cancer, EEC progenitors were blocked from differentiating further by DNA methylation and silencing of NEUROD1, a key gene required for differentiation of intermediate EECs. Mechanistically, secretory cells and the factors they secrete, such as trefoil factor 3, promoted colony formation and activation of cell survival pathways in the entire cell population. Lysine-specific demethylase 1 (LSD1) was identified as a critical regulator of secretory cell specification in vitro and in a colon orthotopic xenograft model, where LSD1 loss blocks formation of EEC progenitors and reduces tumor growth and metastasis. These findings reveal an important role for EEC progenitors in supporting colorectal cancer. SIGNIFICANCE: This study establishes enteroendocrine progenitors as a targetable population that promotes BRAF-mutant colorectal cancer and can be blocked by LSD1 inhibition to suppress tumor growth.


Assuntos
Neoplasias Colorretais/genética , Metilação de DNA , Células Enteroendócrinas/metabolismo , Histona Desmetilases/metabolismo , Proteínas Proto-Oncogênicas B-raf/genética , Animais , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Células Enteroendócrinas/patologia , Células HT29 , Xenoenxertos , Histona Desmetilases/deficiência , Histona Desmetilases/genética , Humanos , Camundongos , Proteínas Proto-Oncogênicas B-raf/metabolismo , Células-Tronco/metabolismo , Células-Tronco/patologia
3.
Histochem Cell Biol ; 155(4): 451-462, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33404704

RESUMO

Chagas disease is caused by the parasite, Trypanosoma cruzi that causes chronic cardiac and digestive dysfunction. Megacolon, an irreversible dilation of the left colon, is the main feature of the gastrointestinal form of Chagas disease. Patients have severe constipation, a consequence of enteric neuron degeneration associated with chronic inflammation. Dysmotility, infection, neuronal loss and a chronic exacerbated inflammation, all observed in Chagas disease, can affect enteroendocrine cells (EEC) expression, which in turn, could influence the inflammatory process. In this study, we investigated the distribution and chemical coding of EEC in the dilated and non-dilated portion of T. cruzi-induced megacolon and in non-infected individuals (control colon). Using immunohistochemistry, EECs were identified by applying antibodies to chromogranin A (CgA), glucagon-like peptide 1 (GLP-1), 5-hydroxytryptamine (5-HT), peptide YY (PYY) and somatostatin (SST). Greater numbers of EEC expressing GLP-1 and SST occurred in the dilated portion compared to the non-dilated portion of the same patients with Chagas disease and in control colon, but numbers of 5-HT and PYY EEC were not significantly different. However, it was noticeable that EEC in which 5-HT and PYY were co-expressed were common in control colon, but were rare in the non-dilated and absent in the dilated portion of chagasic megacolon. An increase in the number of CgA immunoreactive EEC in chagasic patients reflected the increases in EEC numbers summarised above. Our data suggests that the denervation and associated chronic inflammation are accompanied by changes in the number and coding of EEC that could contribute to disorders of motility and defence in the chagasic megacolon.


Assuntos
Doença de Chagas/patologia , Células Enteroendócrinas/patologia , Megacolo/patologia , Trypanosoma cruzi/isolamento & purificação , Doença de Chagas/imunologia , Doença de Chagas/parasitologia , Feminino , Humanos , Imuno-Histoquímica , Inflamação/imunologia , Inflamação/parasitologia , Inflamação/patologia , Masculino , Megacolo/imunologia , Megacolo/parasitologia
4.
J Clin Endocrinol Metab ; 106(2): e966-e981, 2021 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-33135737

RESUMO

CONTEXT: The mechanisms underlying Roux-en-Y gastric bypass (RYGB) surgery-induced weight loss and the immediate postoperative beneficial metabolic effects associated with the operation remain uncertain. Enteroendocrine cell (EEC) secretory function has been proposed as a key factor in the marked metabolic benefits from RYGB surgery. OBJECTIVE: To identify novel gut-derived peptides with therapeutic potential in obesity and/or diabetes by profiling EEC-specific molecular changes in obese patients following RYGB-induced weight loss. SUBJECTS AND METHODS: Genome-wide expression analysis was performed in isolated human small intestinal EECs obtained from 20 gut-biopsied obese subjects before and after RYGB. Targets of interest were profiled for preclinical and clinical metabolic effects. RESULTS: Roux-en-Y gastric bypass consistently increased expression levels of the inverse ghrelin receptor agonist, liver-expressed antimicrobial peptide 2 (LEAP2). A secreted endogenous LEAP2 fragment (LEAP238-47) demonstrated robust insulinotropic properties, stimulating insulin release in human pancreatic islets comparable to the gut hormone glucagon-like peptide-1. LEAP238-47 showed reciprocal effects on growth hormone secretagogue receptor (GHSR) activity, suggesting that the insulinotropic action of the peptide may be directly linked to attenuation of tonic GHSR activity. The fragment was infused in healthy human individuals (n = 10), but no glucoregulatory effect was observed in the chosen dose as compared to placebo. CONCLUSIONS: Small intestinal LEAP2 expression was upregulated after RYGB. The corresponding circulating LEAP238-47 fragment demonstrated strong insulinotropic action in vitro but failed to elicit glucoregulatory effects in healthy human subjects.


Assuntos
Peptídeos Catiônicos Antimicrobianos/metabolismo , Proteínas Sanguíneas/metabolismo , Derivação Gástrica/métodos , Trato Gastrointestinal/metabolismo , Ilhotas Pancreáticas/metabolismo , Obesidade/cirurgia , Fragmentos de Peptídeos/metabolismo , Transcriptoma , Adolescente , Adulto , Peptídeos Catiônicos Antimicrobianos/genética , Biomarcadores/análise , Proteínas Sanguíneas/genética , Estudos de Casos e Controles , Estudos Cross-Over , Método Duplo-Cego , Células Enteroendócrinas/metabolismo , Células Enteroendócrinas/patologia , Feminino , Seguimentos , Humanos , Ilhotas Pancreáticas/patologia , Masculino , Obesidade/patologia , Fragmentos de Peptídeos/genética , Prognóstico , Estudos Prospectivos , Adulto Jovem
5.
Mol Metab ; 37: 100990, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32278655

RESUMO

OBJECTIVE: Glucagon-like peptide-1 is a nutrient-sensitive hormone secreted from enteroendocrine L cells within the small and large bowel. Although GLP-1 levels rise rapidly in response to food ingestion, the greatest density of L cells is localized to the distal small bowel and colon. Here, we assessed the importance of the distal gut in the acute L cell response to diverse secretagogues. METHODS: Circulating levels of glucose and plasma GLP-1 were measured in response to the administration of L cell secretagogues in wild-type mice and in mice with (1) genetic reduction of Gcg expression throughout the small bowel and large bowel (GcgGut-/-) and (2) selective reduction of Gcg expression in the distal gut (GcgDistalGut-/-). RESULTS: The acute GLP-1 response to olive oil or arginine administration was markedly diminished in GcgGut-/- but preserved in GcgDistalGut-/- mice. In contrast, the increase in plasma GLP-1 levels following the administration of the GPR119 agonist AR231453, or the melanocortin-4 receptor (MC4R) agonist LY2112688, was markedly diminished in the GcgDistalGut-/- mice. The GLP-1 response to LPS was also markedly attenuated in the GcgGut-/- mice and remained submaximal in the GcgDistalGut-/- mice. Doses of metformin sufficient to lower glucose and increase GLP-1 levels in the GcgGut+/+ mice retained their glucoregulatory activity, yet they failed to increase GLP-1 levels in the GcgGut-/- mice. Surprisingly, the actions of metformin to increase plasma GLP-1 levels were substantially attenuated in the GcgDistalGut-/- mice. CONCLUSION: These findings further establish the importance of the proximal gut for the acute response to nutrient-related GLP-1 secretagogues. In contrast, we identify essential contributions of the distal gut to (i) the rapid induction of circulating GLP-1 levels in response to pharmacological selective agonism of G-protein-coupled receptors, (ii) the increased GLP-1 levels following the activation of Toll-Like Receptors with LPS, and iii) the acute GLP-1 response to metformin. Collectively, these results reveal that distal gut Gcg + endocrine cells are rapid responders to structurally and functionally diverse GLP-1 secretagogues.


Assuntos
Células Enteroendócrinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glucagon/metabolismo , Animais , Glicemia/análise , Colo/metabolismo , Colo/fisiologia , Células Enteroendócrinas/patologia , Feminino , Expressão Gênica/genética , Regulação da Expressão Gênica/genética , Glucagon/genética , Peptídeo 1 Semelhante ao Glucagon/sangue , Peptídeo 1 Semelhante ao Glucagon/genética , Receptor do Peptídeo Semelhante ao Glucagon 2/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Intestino Delgado/metabolismo , Intestino Delgado/fisiologia , Masculino , Metformina/farmacologia , Camundongos , Camundongos Knockout , Proglucagon/metabolismo
6.
Nutrients ; 11(8)2019 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-31394793

RESUMO

Diet plays an important role not only in the pathophysiology of irritable bowel syndrome (IBS), but also as a tool that improves symptoms and quality of life. The effects of diet seem to be a result of an interaction with the gut bacteria and the gut endocrine cells. The density of gut endocrine cells is low in IBS patients, and it is believed that this abnormality is the direct cause of the symptoms seen in IBS patients. The low density of gut endocrine cells is probably caused by a low number of stem cells and low differentiation progeny toward endocrine cells. A low fermentable oligo-, di-, monosaccharide, and polyol (FODMAP) diet and fecal microbiota transplantation (FMT) restore the gut endocrine cells to the level of healthy subjects. It has been suggested that our diet acts as a prebiotic that favors the growth of a certain types of bacteria. Diet also acts as a substrate for gut bacteria fermentation, which results in several by-products. These by-products might act on the stem cells in such a way that the gut stem cells decrease, and consequently, endocrine cell numbers decrease. Changing to a low-FODMAP diet or changing the gut bacteria through FMT improves IBS symptoms and restores the density of endocrine cells.


Assuntos
Dieta , Hormônios Gastrointestinais/fisiologia , Microbioma Gastrointestinal/fisiologia , Síndrome do Intestino Irritável/dietoterapia , Síndrome do Intestino Irritável/etiologia , Diferenciação Celular , Dieta com Restrição de Carboidratos , Células Enteroendócrinas/patologia , Transplante de Microbiota Fecal , Fermentação , Humanos , Síndrome do Intestino Irritável/patologia , Monossacarídeos/administração & dosagem , Polissacarídeos/administração & dosagem , Prebióticos/administração & dosagem , Qualidade de Vida , Células-Tronco/citologia
7.
Cell Rep ; 27(6): 1934-1947.e5, 2019 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-31067475

RESUMO

Intestinal-type gastric cancer is preceded by premalignant lesions, including chronic atrophic gastritis and intestinal metaplasia. In this study, we constructed a single-cell atlas for 32,332 high-quality cells from gastric antral mucosa biopsies of patients spanning a cascade of gastric premalignant lesions and early gastric cancer (EGC) using single-cell RNA sequencing. We then constructed a single-cell network underlying cellular and molecular characteristics of gastric epithelial cells across different lesions. We found that gland mucous cells tended to acquire an intestinal-like stem cell phenotype during metaplasia, and we identified OR51E1 as a marker for unique endocrine cells in the early-malignant lesion. We also found that HES6 might mark the pre-goblet cell cluster, potentially aiding identification of metaplasia at the early stage. Finally, we identified a panel of EGC-specific signatures, with clinical implications for the precise diagnosis of EGC. Our study offers unparalleled insights into the human gastric cellulome in premalignant and early-malignant lesions.


Assuntos
Redes Reguladoras de Genes , Lesões Pré-Cancerosas/genética , Análise de Célula Única , Neoplasias Gástricas/genética , Transcriptoma/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem da Célula , Células Enteroendócrinas/metabolismo , Células Enteroendócrinas/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Regulação Neoplásica da Expressão Gênica , Células Caliciformes/metabolismo , Humanos , Lesões Pré-Cancerosas/patologia , RNA Neoplásico/genética , RNA Neoplásico/metabolismo , Proteínas Repressoras/metabolismo , Neoplasias Gástricas/patologia , Transcrição Gênica
8.
Diabetes ; 67(9): 1701-1709, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30135131

RESUMO

Diabetes is caused by combined abnormalities in insulin production and action. The pathophysiology of these defects has been studied extensively and is reasonably well understood. Their causes are elusive and their manifestations pleiotropic, likely reflecting the triple threat of genes, environment, and lifestyle. Treatment, once restricted to monotherapy with secretagogues or insulin, now involves complex combinations of expensive regimens that stem the progression but do not fundamentally alter the underlying causes of the disease. As advances in our understanding of insulin action and ß-cell failure reach a critical stage, here I draw on lessons learned from our research on insulin regulation of gene expression and pancreatic ß-cell dedifferentiation to address the question of how we can translate this exciting biology into mechanism-based interventions to reverse the course of diabetes.


Assuntos
Complicações do Diabetes/prevenção & controle , Diabetes Mellitus Tipo 1/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Hipoglicemiantes/uso terapêutico , Células Secretoras de Insulina/efeitos dos fármacos , Insulina/uso terapêutico , Modelos Biológicos , Animais , Distinções e Prêmios , Desdiferenciação Celular/efeitos dos fármacos , Transdiferenciação Celular/efeitos dos fármacos , Reprogramação Celular/efeitos dos fármacos , Terapia Combinada/efeitos adversos , Diabetes Mellitus Tipo 1/complicações , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/terapia , Diabetes Mellitus Tipo 2/complicações , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Desenho de Fármacos , Quimioterapia Combinada/efeitos adversos , Células Enteroendócrinas/efeitos dos fármacos , Células Enteroendócrinas/metabolismo , Células Enteroendócrinas/patologia , Fatores de Transcrição Forkhead/antagonistas & inibidores , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Humanos , Hipoglicemiantes/efeitos adversos , Hipoglicemiantes/química , Hipoglicemiantes/farmacologia , Insulina/efeitos adversos , Insulina/metabolismo , Insulina/farmacologia , Resistência à Insulina , Secreção de Insulina , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Fígado/patologia
9.
Cell Physiol Biochem ; 47(4): 1442-1452, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29940597

RESUMO

BACKGROUND/AIMS: Type II diabetes is a complex, chronic, and progressive disease. Glucagon-like peptide-1 (7-6) amide (GLP-1) is a gut hormone released from the L cells which stimulates insulin secretion, and promotes insulin gene expression and ß-cell growth and differentiation. Elevated levels of hormone secreted by L cells are an important reason for diabetes improvement. GLP-1 secretion has been reported to be regulated by farnesoid X receptor (FXR), a transcriptional sensor for bile acids which also acts on glucose metabolism. Herein, we attempted to evaluate the effect of FXR on GLP-1 secretion in mouse enteroendocrine L cell lines, STC-1 and GLUTag, and to investigate the underlying mechanism. METHODS: ELISA and Western blot assays were employed to examine the levels of GLP-1 and FXR, and the effect of FXR on GLP-1 secretion; online database, including BioGRID and KEGG were used to identify the potential interactions between FXR and proteins and involved pathways; GST pull-down and Co-Immunoprecipitation (Co-IP) assays were performed to validate FXR-CREB interaction; Luciferase reporter gene assays were used for CREB transcriptional activity determination. RESULTS: FXR inversely regulated GLP-1 secretion in the mouse enteroendocrine L cell lines, GLUTag and STC-1. A total of 24 nonredundant human proteins were shown to be related to FXR by BioGRID; KEGG pathway analysis showed that FXR was related to glucagon signaling pathway, particularly with the transcriptional activators CREB, PGC1α, Sirt1 and CBP. CREB could positively regulate GLP-1 secretion in GLUTag and STC-1 cells. FXR combined with CREB to inhibit its transcriptional activity, thus inhibiting proprotein convertase subtilisin/ kexin type 1 (PCSK1) protein level and GLP-1 secretion. CONCLUSION: In the present study, we demonstrated a negative regulation of GLP-1 secretion by FXR in L cell lines, GLUTag and STC-1; FXR exerts its function in L cells through interacting with CREB, a crucial transcriptional regulator of cAMP-CREB signaling pathway, to inhibit its transcriptional activity. Targeting FXR to rescue GLP-1 secretion may be a promising strategy for type II diabetes.


Assuntos
Proteína de Ligação a CREB/metabolismo , Células Enteroendócrinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Fragmentos de Peptídeos/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Proteína de Ligação a CREB/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patologia , Células Enteroendócrinas/patologia , Células L , Camundongos , Receptores Citoplasmáticos e Nucleares/genética
10.
Cell Tissue Res ; 374(2): 339-348, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29869714

RESUMO

The antimicrobial glycoprotein neutrophil gelatinase-associated lipocalin (NGAL) is strongly expressed in several infectious, inflammatory and malignant disorders, among these inflammatory bowel disease (IBD). Fecal and serum NGAL is elevated during active IBD and we have recently shown that fecal NGAL is a novel biomarker for IBD with a test performance comparable to the established fecal biomarker calprotectin. This study examines expression of NGAL in the healthy gut and in Crohn's disease (CD), with emphasis on the previously unexplored small intestine. Pinch biopsies were taken from active and inactive CD in jejunum, ileum and colon and from the same sites in healthy controls. Microarray gene expression showed that the NGAL gene, LCN2, was the second most upregulated among 1820 differentially expressed genes in terminal ileum comparing active CD and controls (FC 5.86, p = 0.027). Based on immunohistochemistry and in situ hybridization findings, this upregulation most likely represented increased expression in epithelial cells. Double immunofluorescence showed NGAL expression in 49% (range 19-70) of Paneth cells (PCs) in control ileum with no change during inflammation. In healthy jejunum, the NGAL expression in PCs was weak to none but markedly increased during active CD. We further found NGAL also in metaplastic PCs in colon. Finally, we show for the first time that NGAL is expressed in enteroendocrine cells in small intestine as well as in colon.


Assuntos
Doença de Crohn/genética , Doença de Crohn/patologia , Sistema Digestório/patologia , Lipocalina-2/genética , Adulto , Sistema Digestório/metabolismo , Células Enteroendócrinas/metabolismo , Células Enteroendócrinas/patologia , Humanos , Lipocalina-2/metabolismo , Pessoa de Meia-Idade , Celulas de Paneth/metabolismo , Celulas de Paneth/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Regulação para Cima/genética , Adulto Jovem
11.
J Cell Biochem ; 119(9): 7506-7514, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29761876

RESUMO

As incretins are known to play an important role in type 2 diabetics (T2D) improvement observed after Roux-en-Y gastric bypass (RYGB), our aim was to assess whether increasing the length of RYGB biliopancreatic limb in T2D would modify the incretin staining cell density found after the gastric outlet. Small intestine biopsies (n = 38) were harvested during RYGB at two different distances from the duodenal angle; either 60-90 cm (n = 28), from non-diabetic (n = 18) patients, and T2D (n = 10), or 200 cm (n = 10) from T2D. GIP and GLP-1 staining cells were identified by immunohistochemistry and GLP-1/GIP co-staining cells by immunofluorescence. Incretin staining cell density at the proximal small intestine of T2D and non-diabetic individuals was similar. At 200 cm, T2D patients depicted a significantly lower GIP staining cell density (0.181 ± 0.016 vs 0.266 ± 0.033, P = 0.038) with a similar GLP-1 staining cell density when compared to the proximal gut. GIP/GLP-1 co-staining cells was similar in all studied groups. In T2D patients, the incretin staining cells density in the distal intestine is significantly different from the proximal gut. Thus, a longer RYGB biliopancreatic limb produces a distinctive incretin cell pattern at the gastro-enteric anastomosis that can result in different endocrine profiles.


Assuntos
Diabetes Mellitus Tipo 2/patologia , Células Enteroendócrinas/patologia , Derivação Gástrica , Intestino Delgado/patologia , Obesidade/patologia , Adulto , Diabetes Mellitus Tipo 2/complicações , Células Enteroendócrinas/metabolismo , Feminino , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Humanos , Incretinas/metabolismo , Intestino Delgado/metabolismo , Masculino , Obesidade/complicações , Obesidade/cirurgia
12.
Gastroenterology ; 154(8): 2045-2059.e6, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29654747

RESUMO

Diarrhea is common in infants (children less than 2 years of age), usually acute, and, if chronic, commonly caused by allergies and occasionally by infectious agents. Congenital diarrheas and enteropathies (CODEs) are rare causes of devastating chronic diarrhea in infants. Evaluation of CODEs is a lengthy process and infrequently leads to a clear diagnosis. However, genomic analyses and the development of model systems have increased our understanding of CODE pathogenesis. With these advances, a new diagnostic approach is needed. We propose a revised approach to determine causes of diarrhea in infants, including CODEs, based on stool analysis, histologic features, responses to dietary modifications, and genetic tests. After exclusion of common causes of diarrhea in infants, the evaluation proceeds through analyses of stool characteristics (watery, fatty, or bloody) and histologic features, such as the villus to crypt ratio in intestinal biopsies. Infants with CODEs resulting from defects in digestion, absorption, transport of nutrients and electrolytes, or enteroendocrine cell development or function have normal villi to crypt ratios; defects in enterocyte structure or immune-mediated conditions result in an abnormal villus to crypt ratios and morphology. Whole-exome and genome sequencing in the early stages of evaluation can reduce the time required for a definitive diagnosis of CODEs, or lead to identification of new variants associated with these enteropathies. The functional effects of gene mutations can be analyzed in model systems such as enteroids or induced pluripotent stem cells and are facilitated by recent advances in gene editing procedures. Characterization and investigation of new CODE disorders will improve management of patients and advance our understanding of epithelial cells and other cells in the intestinal mucosa.


Assuntos
Diarreia Infantil/diagnóstico , Enterócitos/patologia , Células Enteroendócrinas/patologia , Enteropatias/diagnóstico , Biópsia , Doença Crônica , Procedimentos Clínicos , Diarreia Infantil/classificação , Diarreia Infantil/etiologia , Diarreia Infantil/patologia , Endoscopia do Sistema Digestório , Enterócitos/metabolismo , Células Enteroendócrinas/metabolismo , Testes Genéticos/métodos , Humanos , Lactente , Recém-Nascido , Enteropatias/classificação , Enteropatias/etiologia , Enteropatias/patologia , Mutação , Sequenciamento Completo do Genoma
13.
Mol Cell Biochem ; 447(1-2): 21-32, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29363059

RESUMO

Glucagon-like peptide-1 (GLP-1) is involved in the regulation of insulin secretion and glucose homeostasis. GLP-1 release is stimulated when berberine interacts with a novel G protein family (TAS2Rs) in enteroendocrine cells. In this study, we used STC-1 cells and examined a marked increase in Ca2+ in response to various bitter compounds. Ca2+ responses to traditional Chinese medicine extracts, including berberine, phellodendrine and coptisine, in STC-1 cells were suppressed by the phospholipase C (PLC) inhibitor U-73122, suggesting the involvement of bitter taste receptors in changing the physiological status of enteroendocrine cells in a PLC-dependent manner. STC-1 cells showed berberine-up-regulated preproglucagon (GLP-1 precursor) mRNA and GLP-1 secretion. A QPCR analysis demonstrated that TAS2R38, a subtype of the bitter taste receptor, was associated with GLP-1 secretion. Berberine-mediated GLP-1 secretion was attenuated in response to small interfering RNA silencing of TAS2R38. The current studies demonstrated that Gα-gustducin co-localized with GLP-1 and Tas2r106 in the STC-1 cells. We further utilized inhibitors of PLC and TRPM5, which are known to participate in taste signal transduction, to investigate the underlying pathways mediated in berberine-induced GLP-1 secretion. Berberine-induced GLP-1 release from enteroendocrine cells is modulated in a PLC-dependent manner through a process involving the activation of bitter taste receptors. Together, our data demonstrated a berberine-mediated GLP-1 secretion pathway in mouse enteroendocrine cells that could be of therapeutic relevance to hyperglycemia and the role of bitter taste receptors in the function of the small intestine.


Assuntos
Berberina/farmacologia , Células Enteroendócrinas/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Regulação para Cima/efeitos dos fármacos , Linhagem Celular Tumoral , Células Enteroendócrinas/patologia , Estrenos/farmacologia , Humanos , Hiperglicemia/tratamento farmacológico , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Intestino Delgado/metabolismo , Intestino Delgado/patologia , Pirrolidinonas/farmacologia , Fosfolipases Tipo C/antagonistas & inibidores , Fosfolipases Tipo C/metabolismo
14.
JCI Insight ; 2(21)2017 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-29093273

RESUMO

The secretion of insulin and glucagon from the pancreas and the incretin hormones glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic peptide (GIP) from the gastrointestinal tract is essential for glucose homeostasis. Several novel treatment strategies for type 2 diabetes (T2D) mimic GLP-1 actions or inhibit incretin degradation (DPP4 inhibitors), but none is thus far aimed at increasing the secretion of endogenous incretins. In order to identify new potential therapeutic targets for treatment of T2D, we performed a meta-analysis of a GWAS and an exome-wide association study of circulating insulin, glucagon, GIP, and GLP-1 concentrations measured during an oral glucose tolerance test in up to 7,828 individuals. We identified 6 genome-wide significant functional loci associated with plasma incretin concentrations in or near the SLC5A1 (encoding SGLT1), GIPR, ABO, GLP2R, F13A1, and HOXD1 genes and studied the effect of these variants on mRNA expression in pancreatic islet and on metabolic phenotypes. Immunohistochemistry showed expression of GIPR, ABO, and HOXD1 in human enteroendocrine cells and expression of ABO in pancreatic islets, supporting a role in hormone secretion. This study thus provides candidate genes and insight into mechanisms by which secretion and breakdown of GIP and GLP-1 are regulated.


Assuntos
Células Enteroendócrinas/metabolismo , Polipeptídeo Inibidor Gástrico/genética , Variação Genética , Peptídeo 1 Semelhante ao Glucagon/genética , Glucagon/metabolismo , Insulina/metabolismo , Sistema ABO de Grupos Sanguíneos/genética , Idoso , Idoso de 80 Anos ou mais , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/terapia , Dipeptidil Peptidase 4/efeitos dos fármacos , Células Enteroendócrinas/patologia , Feminino , Polipeptídeo Inibidor Gástrico/metabolismo , Hormônios Gastrointestinais , Trato Gastrointestinal/metabolismo , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 2/genética , Glucose/metabolismo , Teste de Tolerância a Glucose , Proteínas de Homeodomínio/genética , Humanos , Incretinas/metabolismo , Insulina/genética , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , RNA Mensageiro/metabolismo , Receptores dos Hormônios Gastrointestinais/genética , Transportador 1 de Glucose-Sódio/genética
15.
Int J Mol Med ; 40(4): 943-952, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28849091

RESUMO

Irritable bowel syndrome (IBS) is a common chronic gastrointestinal (GI) disorder that is characterized by a combination of abdominal pain or discomfort, bloating and alterations in bowel movements. This review presents recent developments concerning the roles of diet and GI endocrine cells in the pathophysiology of IBS and of individual dietary guidance in the management of IBS. Patients with IBS typically report that food aggravates their IBS symptoms. The interactions between specific types of foodstuffs rich in fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAPs) and GI endocrine cells induce changes in cell densities. Providing individual dietary guidance about a low FODMAP intake, high soluble­fiber intake, and changing the proportions of protein, fat and carbohydrates helps to reduce the symptoms experienced by patients with IBS and to improve their quality of life. These improvements are due to restoring the densities of the GI endocrine cells back to normal. The reported observations emphasize the role of GI endocrine cells in the pathophysiology of IBS and support the provision of dietary guidance as a first-line treatment for managing IBS.


Assuntos
Carboidratos da Dieta/administração & dosagem , Fibras na Dieta/administração & dosagem , Células Enteroendócrinas/metabolismo , Síndrome do Intestino Irritável/dietoterapia , Recomendações Nutricionais , Contagem de Células , Dieta/métodos , Carboidratos da Dieta/metabolismo , Fibras na Dieta/metabolismo , Dissacarídeos/administração & dosagem , Dissacarídeos/metabolismo , Células Enteroendócrinas/efeitos dos fármacos , Células Enteroendócrinas/patologia , Fermentação , Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/metabolismo , Regulação da Expressão Gênica , Humanos , Síndrome do Intestino Irritável/genética , Síndrome do Intestino Irritável/metabolismo , Síndrome do Intestino Irritável/fisiopatologia , Monossacarídeos/administração & dosagem , Monossacarídeos/metabolismo , Oligossacarídeos/administração & dosagem , Oligossacarídeos/metabolismo , Qualidade de Vida
16.
Cell Stem Cell ; 21(1): 78-90.e6, 2017 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-28686870

RESUMO

Several cell populations have been reported to possess intestinal stem cell (ISC) activity during homeostasis and injury-induced regeneration. Here, we explored inter-relationships between putative mouse ISC populations by comparative RNA-sequencing (RNA-seq). The transcriptomes of multiple cycling ISC populations closely resembled Lgr5+ ISCs, the most well-defined ISC pool, but Bmi1-GFP+ cells were distinct and enriched for enteroendocrine (EE) markers, including Prox1. Prox1-GFP+ cells exhibited sustained clonogenic growth in vitro, and lineage-tracing of Prox1+ cells revealed long-lived clones during homeostasis and after radiation-induced injury in vivo. Single-cell mRNA-seq revealed two subsets of Prox1-GFP+ cells, one of which resembled mature EE cells while the other displayed low-level EE gene expression but co-expressed tuft cell markers, Lgr5 and Ascl2, reminiscent of label-retaining secretory progenitors. Our data suggest that the EE lineage, including mature EE cells, comprises a reservoir of homeostatic and injury-inducible ISCs, extending our understanding of cellular plasticity and stemness.


Assuntos
Antígenos de Diferenciação/metabolismo , Células Enteroendócrinas/metabolismo , Mucosa Intestinal/lesões , Mucosa Intestinal/metabolismo , Jejuno/lesões , Jejuno/metabolismo , Células-Tronco/metabolismo , Animais , Antígenos de Diferenciação/genética , Células Enteroendócrinas/patologia , Regulação da Expressão Gênica , Mucosa Intestinal/patologia , Jejuno/patologia , Camundongos , Camundongos Transgênicos , Células-Tronco/patologia
17.
Mol Med Rep ; 15(4): 2106-2112, 2017 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-28259987

RESUMO

The present study aimed to determine whether there is an association between abnormalities in enteroendocrine cells in dextran sulfate sodium (DSS)­induced colitis and the clonogenic and/or proliferative activities of stem cells. A total of 48 male Wistar rats were divided into four groups. Animals in the control group were provided with normal drinking water, whereas DSS colitis was induced in the remaining three groups. The rats with DSS­induced colitis were randomized into the following three groups: i) DSS group, which received 0.5 ml 0.5% carboxymethyl cellulose (CMC; vehicle); ii) DSS­G group, which was treated with 3-[(dodecylthiocarbonyl)-methyl]-glutarimide at 20 mg/kg body weight in 0.5% CMC; and iii) DSS­Q group, which was treated with dehydroxymethylepoxyquinomicin at 15 mg/kg body weight in 0.5% CMC. Treatments were administered intraperitoneally twice daily for 5 days in all groups. Subsequently, tissue samples from the colon were stained with hematoxylin­eosin, or immunostained for chromogranin A (CgA), Musashi 1 (Msi1), Math­1, neurogenin 3 (Neurog3) and neurogenic differentiation D1 (NeuroD1). The densities of CgA, Msi1­, Math­1­, Neurog3­ and NeuroD1-immunoreactive cells were determined. DTCM­G, and DHMEQ ameliorated the inflammation in DSS­induced colitis. The density of CgA­, Neurog3­ and NeuroD1­immunoreactive cells was significantly higher in the DSS group compared with in the control group, and the density of CgA cells was correlated with the densities of Neurog3­ and NeuroD1-immunoreactive cells. There were no significant differences in the densities of Msi1­ and Math­1­immunoreactive cells among the four experimental groups. The elevated densities of enteroendocrine cells detected in DSS­induced colitis may be due to the increased differentiation of early enteroendocrine progenitors during secretory lineage. It is probable that the DSS­induced inflammatory processes trigger certain signaling pathways, which control differentiation of the stem­cell secretory lineage into mature enteroendocrine cells.


Assuntos
Colite/induzido quimicamente , Colite/patologia , Colo/patologia , Sulfato de Dextrana , Células Enteroendócrinas/patologia , Células-Tronco/patologia , Animais , Diferenciação Celular , Células Enteroendócrinas/citologia , Masculino , Ratos Wistar , Células-Tronco/citologia
18.
Med Mol Morphol ; 50(2): 94-102, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28070680

RESUMO

The adenomatous polyposis coli (APC) is a multifunctional protein as well as a tumor suppressor. To determine the functions of the C-terminal domain of APC, we explored APC 1638T/1638T (APC1638T) mice that express a truncated APC lacking the C-terminal domain. The APC1638T mice were tumor free and exhibited growth retardation. In the present study, we compared small intestinal crypt-villus cells homeostasis in APC +/+ (WT) mice and APC1638T mice. The body weight of APC1638T mice was significantly smaller than that of WT mice at all ages. The length of small intestine of APC1638T mice was significantly shorter than that of WT mice. The crypt-villus axis was significantly elongated, and the number of intestinal epithelial cells also increased in APC1638T mice compared with those in WT mice. However, the number of intestinal epithelial cells per 100 µm of villi was not different between WT and APC1638T mice. Migration and proliferation of intestinal epithelial cells in APC1638T mice were faster than that in WT mice. The population of Goblet cells, Paneth cells, and enteroendocrine cells was significantly altered in APC1638T mice. These results indicate that C-terminal domain of APC has a role in the regulation of intestinal epithelium homeostasis.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Células Enteroendócrinas/patologia , Células Caliciformes/patologia , Mucosa Intestinal/patologia , Intestino Delgado/patologia , Celulas de Paneth/patologia , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Sequência de Bases , Tamanho Corporal , Contagem de Células , Movimento Celular , Proliferação de Células , Células Enteroendócrinas/metabolismo , Feminino , Expressão Gênica , Células Caliciformes/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Celulas de Paneth/metabolismo , Domínios Proteicos , Deleção de Sequência
19.
Expert Rev Gastroenterol Hepatol ; 11(2): 139-148, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27927062

RESUMO

INTRODUCTION: The etiology of irritable bowel syndrome (IBS) is unknown, but several factors appear to play a role in its pathophysiology, including abnormalities of the gastrointestinal endocrine cells. The present review illuminates the possible role of gastrointestinal hormones in the pathophysiology of IBS and the possibility of utilizing the current knowledge in treating the disease. Areas covered: Research into the intestinal endocrine cells and their possible role in the pathophysiology of IBS is discussed. Furthermore, the mechanisms underlying the abnormalities in the gastrointestinal endocrine cells in IBS patients are revealed. Expert commentary: The abnormalities observed in the gastrointestinal endocrine cells in IBS patients explains their visceral hypersensitivity, gastrointestinal dysmotility, and abnormal intestinal secretion, as well as the interchangeability of symptoms over time. Clarifying the role of the intestinal stem cells in the pathophysiology of IBS may lead to new treatment methods for IBS.


Assuntos
Células Enteroendócrinas/metabolismo , Hormônios Gastrointestinais/metabolismo , Mucosa Intestinal/metabolismo , Síndrome do Intestino Irritável/metabolismo , Células-Tronco/metabolismo , Animais , Células Enteroendócrinas/patologia , Motilidade Gastrointestinal , Humanos , Secreções Intestinais/metabolismo , Intestinos/patologia , Intestinos/fisiopatologia , Síndrome do Intestino Irritável/patologia , Síndrome do Intestino Irritável/fisiopatologia , Fenótipo , Transdução de Sinais , Células-Tronco/patologia , Dor Visceral/metabolismo , Dor Visceral/patologia , Dor Visceral/fisiopatologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA