Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 141
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Histochem Cell Biol ; 155(4): 451-462, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33404704

RESUMEN

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.


Asunto(s)
Enfermedad de Chagas/patología , Células Enteroendocrinas/patología , Megacolon/patología , Trypanosoma cruzi/aislamiento & purificación , Enfermedad de Chagas/inmunología , Enfermedad de Chagas/parasitología , Femenino , Humanos , Inmunohistoquímica , Inflamación/inmunología , Inflamación/parasitología , Inflamación/patología , Masculino , Megacolon/inmunología , Megacolon/parasitología
2.
Gastroenterology ; 154(8): 2045-2059.e6, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29654747

RESUMEN

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.


Asunto(s)
Diarrea Infantil/diagnóstico , Enterocitos/patología , Células Enteroendocrinas/patología , Enfermedades Intestinales/diagnóstico , Biopsia , Enfermedad Crónica , Vías Clínicas , Diarrea Infantil/clasificación , Diarrea Infantil/etiología , Diarrea Infantil/patología , Endoscopía del Sistema Digestivo , Enterocitos/metabolismo , Células Enteroendocrinas/metabolismo , Pruebas Genéticas/métodos , Humanos , Lactante , Recién Nacido , Enfermedades Intestinales/clasificación , Enfermedades Intestinales/etiología , Enfermedades Intestinales/patología , Mutación , Secuenciación Completa del Genoma
3.
J Cell Biochem ; 119(9): 7506-7514, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29761876

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 2/patología , Células Enteroendocrinas/patología , Derivación Gástrica , Intestino Delgado/patología , Obesidad/patología , Adulto , Diabetes Mellitus Tipo 2/complicaciones , Células Enteroendocrinas/metabolismo , Femenino , Péptido 1 Similar al Glucagón/metabolismo , Humanos , Incretinas/metabolismo , Intestino Delgado/metabolismo , Masculino , Obesidad/complicaciones , Obesidad/cirugía
4.
Cell Physiol Biochem ; 47(4): 1442-1452, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29940597

RESUMEN

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.


Asunto(s)
Proteína de Unión a CREB/metabolismo , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Fragmentos de Péptidos/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Proteína de Unión a CREB/genética , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Células Enteroendocrinas/patología , Células L , Ratones , Receptores Citoplasmáticos y Nucleares/genética
5.
Cell Tissue Res ; 374(2): 339-348, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29869714

RESUMEN

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.


Asunto(s)
Enfermedad de Crohn/genética , Enfermedad de Crohn/patología , Sistema Digestivo/patología , Lipocalina 2/genética , Adulto , Sistema Digestivo/metabolismo , Células Enteroendocrinas/metabolismo , Células Enteroendocrinas/patología , Humanos , Lipocalina 2/metabolismo , Persona de Mediana Edad , Células de Paneth/metabolismo , Células de Paneth/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba/genética , Adulto Joven
6.
Mol Cell Biochem ; 447(1-2): 21-32, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29363059

RESUMEN

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.


Asunto(s)
Berberina/farmacología , Células Enteroendocrinas/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Receptores Acoplados a Proteínas G/biosíntesis , Regulación hacia Arriba/efectos de los fármacos , Línea Celular Tumoral , Células Enteroendocrinas/patología , Estrenos/farmacología , Humanos , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Hiperglucemia/patología , Intestino Delgado/metabolismo , Intestino Delgado/patología , Pirrolidinonas/farmacología , Fosfolipasas de Tipo C/antagonistas & inhibidores , Fosfolipasas de Tipo C/metabolismo
7.
Biochim Biophys Acta ; 1862(4): 696-704, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26775031

RESUMEN

OBJECTIVE: Serum autotaxin (ATX) activity is significantly increased in cholestatic patients. Our study aimed to unravel the source(s) of ATX in cholestasis. MATERIALS AND METHODS: ATX activity and protein were measured in sera of healthy (n=33) and cholestatic patients (n=152), including women with intrahepatic cholestasis of pregnancy. ATX mRNA and protein expression were analyzed in various tissues from mice and men. Induction of ATX activity was assessed in mouse models of extrahepatic (bile duct ligation) and intrahepatic cholestasis (Atp8b1(G308V/G308V), 0.1% cholate-supplemented diet). ATX clearance in cholestatic and control mice was assessed after intravenous injection of recombinant ATX. Human hepatic clearance was estimated by comparing ATX activity in portal and hepatic vein serum. RESULTS: Serum ATX activity and ATX protein concentration tightly correlated under all conditions in patients and controls (p<0.0001). In humans Atx mRNA was highly expressed in small intestine, whereas in mice Atx was expressed mainly in brain and placenta but not in small intestine. Extensive ATX protein expression was identified in human, but not murine intestinal enteroendocrine cells. In murine models of cholestasis and cholestatic pregnancy plasma ATX activity was only mildly elevated (up to 2.1-fold). Atx tissue expression and rATX clearance after parenteral administration did not differ between cholestatic and control mice. CONCLUSION: Serum ATX activity during cholestasis and itch is enhanced by increased protein concentration rather than enzymatic induction. In mice, clearance of ATX is not affected by cholestasis. Small intestinal ATX expression by enteroendocrine cells might represent an important source of cholestasis-induced serum ATX activity in men.


Asunto(s)
Colestasis/sangre , Células Enteroendocrinas/enzimología , Regulación Enzimológica de la Expresión Génica , Hidrolasas Diéster Fosfóricas/sangre , Complicaciones del Embarazo/sangre , Animales , Colestasis/enzimología , Colestasis/patología , Células Enteroendocrinas/patología , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Embarazo , Complicaciones del Embarazo/patología , ARN Mensajero/biosíntesis
8.
Med Mol Morphol ; 50(2): 94-102, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28070680

RESUMEN

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.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Células Enteroendocrinas/patología , Células Caliciformes/patología , Mucosa Intestinal/patología , Intestino Delgado/patología , Células de Paneth/patología , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Secuencia de Bases , Tamaño Corporal , Recuento de Células , Movimiento Celular , Proliferación Celular , Células Enteroendocrinas/metabolismo , Femenino , Expresión Génica , Células Caliciformes/metabolismo , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Masculino , Ratones , Ratones Transgénicos , Células de Paneth/metabolismo , Dominios Proteicos , Eliminación de Secuencia
9.
Am J Physiol Gastrointest Liver Physiol ; 309(12): G975-87, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26492922

RESUMEN

There are two major stem cell populations in the intestinal crypt region that express either Bmi1 or Lgr5; however, it has been shown that other populations in the crypt can regain stemness. In this study, we demonstrate that the transcription factor NK2 homeobox 2 (Nkx2.2) is expressed in enteroendocrine cells located in the villus and crypt of the intestinal epithelium and is coexpressed with the stem cell markers Bmi1 and Lgr5 in a subset of crypt cells. To determine whether Nkx2.2-expressing enteroendocrine cells display cellular plasticity and stem cell potential, we performed genetic lineage tracing of the Nkx2.2-expressing population using Nkx2.2(Cre/+);R26RTomato mice. These studies demonstrated that Nkx2.2+ cells are able to give rise to all intestinal epithelial cell types in basal conditions. The proliferative capacity of Nkx2.2-expressing cells was also demonstrated in vitro using crypt organoid cultures. Injuring the intestine with irradiation, systemic inflammation, and colitis did not enhance the lineage potential of Nkx2.2-expressing cells. These findings demonstrate that a rare mature enteroendocrine cell subpopulation that is demarcated by Nkx2.2 expression display stem cell properties during normal intestinal epithelial homeostasis, but is not easily activated upon injury.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Proliferación Celular , Células Enteroendocrinas/metabolismo , Proteínas de Homeodominio/metabolismo , Mucosa Intestinal/metabolismo , Células Madre Pluripotentes/metabolismo , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores/metabolismo , Células Cultivadas , Células Enteroendocrinas/patología , Células Enteroendocrinas/efectos de la radiación , Genotipo , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/genética , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Complejo Represivo Polycomb 1/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Factores de Transcripción/genética , Irradiación Corporal Total , Proteínas de Pez Cebra , Proteína Fluorescente Roja
10.
Gastroenterology ; 147(1): 162-171.e6, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24662331

RESUMEN

BACKGROUND & AIMS: Type 1 diabetes is caused by an aberrant response against pancreatic ß cells. Intestinal K cells are glucose-responsive endocrine cells that might be engineered to secrete insulin. We generated diabetes-prone non-obese diabetic (NOD) mice that express insulin, via a transgene, in K cells. We assessed the effects on immunogenicity and diabetes development. METHODS: Diabetes incidence and glucose homeostasis were assessed in NOD mice that expressed mouse preproinsulin II from a transgene in K cells and nontransgenic NOD mice (controls); pancreas and duodenum tissues were collected and analyzed by histology. We evaluated T cell responses to insulin, levels of circulating autoantibodies against insulin, and the percentage of circulating antigen-specific T cells. Inflammation of mesenteric and pancreatic lymph node cells was also evaluated. RESULTS: The transgenic mice tended to have lower blood levels of glucose than control mice, associated with increased plasma levels of immunoreactive insulin and proinsulin. Fewer transgenic mice developed diabetes than controls. In analyses of pancreas and intestine tissues from the transgenic mice, insulin-producing K cells were not affected by the immune response and the mice had reduced destruction of endogenous ß cells. Fewer transgenic mice were positive for insulin autoantibodies compared with controls. Cells isolated from mesenteric lymph nodes of the transgenic mice had significantly lower rates of proliferation and T cells from transgenic mice tended to secrete lower levels of inflammatory cytokines than from controls. At 15 weeks, transgenic mice had fewer peripheral CD8(+) T cells specific for NRP-V7 than control mice. CONCLUSIONS: NOD mice with intestinal K cells engineered to express insulin have reduced blood levels of glucose, are less likely to develop diabetes, and have reduced immunity against pancreatic ß cells compared with control NOD mice. This approach might be developed to treat patients with type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/prevención & control , Células Enteroendocrinas/metabolismo , Glucosa/metabolismo , Células Secretoras de Insulina/inmunología , Insulina/metabolismo , Animales , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Citocinas/metabolismo , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Duodeno/metabolismo , Duodeno/patología , Células Enteroendocrinas/patología , Femenino , Homeostasis/fisiología , Insulina/inmunología , Ratones , Ratones Endogámicos NOD , Ratones Transgénicos , Páncreas/metabolismo , Páncreas/patología , Linfocitos T/metabolismo , Linfocitos T/patología
11.
Gastroenterology ; 146(3): 754-764.e3, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24316261

RESUMEN

BACKGROUND & AIMS: The alimentary tract contains a diffuse endocrine system comprising enteroendocrine cells that secrete peptides or biogenic amines to regulate digestion, insulin secretion, food intake, and energy homeostasis. Lineage analysis in the stomach revealed that a significant fraction of endocrine cells in the gastric corpus did not arise from Neurogenin3 (Neurog3)-expressing cells, unlike enteroendocrine cells elsewhere in the digestive tract. We aimed to isolate enriched serotonin-secreting and enterochromaffin-like (ECL) cells from the stomach and to clarify their cellular origin. METHODS: We used Neurogenic differentiation 1 (NeuroD1) and Neurog3 lineage analysis and examined the differentiation of serotonin-producing and ECL cells in stomach tissues of NeuroD1-cre;ROSA(tdTom), tryptophan hydroxylase 1 (Tph1)-cyan fluorescent protein (CFP), c-Kit(wsh/wsh), and Neurog3Cre;ROSA(tdTom) mice by immunohistochemistry. We used fluorescence-activated cell sorting to isolate each cell type for gene expression analysis. We also performed RNA sequencing analysis of ECL cells. RESULTS: Neither serotonin-secreting nor ECL cells of the corpus arose from cells expressing NeuroD1. Serotonin-secreting cells expressed a number of mast cell genes but not genes associated with endocrine differentiation; they did not develop in c-Kit(wsh/wsh) mice and were labeled with transplanted bone marrow cells. RNA sequencing analysis of ECL cells revealed high expression levels of many genes common to endocrine cells, including transcription factors, hormones, ion channels, and solute transporters but not markers of bone marrow cells. CONCLUSIONS: Serotonin-expressing cells of the gastric corpus of mice appear to be bone marrow-derived mucosal mast cells. Gene expression analysis of ECL cells indicated that they are endocrine cells of epithelial origin that do not express the same transcription factors as their intestinal enteroendocrine cell counterparts.


Asunto(s)
Linaje de la Célula , Células Enterocromafines/patología , Células Enteroendocrinas/patología , Serotonina/metabolismo , Estómago/patología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Diferenciación Celular , Células Enterocromafines/metabolismo , Células Enteroendocrinas/metabolismo , Mucosa Gástrica/metabolismo , Mastocitos/metabolismo , Mastocitos/patología , Ratones , Ratones Transgénicos , Modelos Animales , Proteínas del Tejido Nervioso/metabolismo
12.
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
13.
J Biol Chem ; 288(3): 1929-38, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23192339

RESUMEN

Gastric inhibitory polypeptide (GIP) is an incretin released from enteroendocrine K-cells in response to nutrient ingestion. GIP potentiates glucose-stimulated insulin secretion and induces energy accumulation into adipose tissue, resulting in obesity. Plasma GIP levels are reported to be increased in the obese state. However, the molecular mechanisms of GIP secretion and high fat diet (HFD)-induced GIP hypersecretion remain unclear, primarily due to difficulties in separating K-cells from other intestinal epithelial cells in vivo. In this study, GIP-GFP knock-in mice that enable us to visualize K-cells by enhanced GFP were established. Microarray analysis of isolated K-cells from these mice revealed that transcriptional regulatory factor X6 (Rfx6) is expressed exclusively in K-cells. In vitro experiments using the mouse intestinal cell line STC-1 showed that knockdown of Rfx6 decreased mRNA expression, cellular content, and secretion of GIP. Rfx6 bound to the region in the gip promoter that regulates gip promoter activity, and overexpression of Rfx6 increased GIP mRNA expression. HFD induced obesity and GIP hypersecretion in GIP-GFP heterozygous mice in vivo. Immunohistochemical and flow cytometry analysis showed no significant difference in K-cell number between control fat diet-fed (CFD) and HFD-fed mice. However, GIP content in the upper small intestine and GIP mRNA expression in K-cells were significantly increased in HFD-fed mice compared with those in CFD-fed mice. Furthermore, expression levels of Rfx6 mRNA were increased in K-cells of HFD-fed mice. These results suggest that Rfx6 increases GIP expression and content in K-cells and is involved in GIP hypersecretion in HFD-induced obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Proteínas de Unión al ADN/genética , Células Enteroendocrinas/metabolismo , Polipéptido Inhibidor Gástrico/genética , Obesidad/genética , Factores de Transcripción/genética , Tejido Adiposo/patología , Animales , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/metabolismo , Dieta Alta en Grasa/efectos adversos , Células Enteroendocrinas/patología , Polipéptido Inhibidor Gástrico/metabolismo , Expresión Génica , Técnicas de Sustitución del Gen , Genes Reporteros , Glucosa/metabolismo , Proteínas Fluorescentes Verdes , Insulina/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Ratones , Ratones Transgénicos , Obesidad/etiología , Obesidad/metabolismo , Obesidad/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Cultivo Primario de Células , Regiones Promotoras Genéticas , Unión Proteica , ARN Interferente Pequeño , Factores de Transcripción del Factor Regulador X , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/metabolismo
14.
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
15.
Int J Exp Pathol ; 95(1): 1-7, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24354351

RESUMEN

Models of the development and early progression of colorectal cancer are based upon understanding the cycle of stem cell turnover, proliferation, differentiation and death. Existing crypt compartmental models feature a linear pathway of cell types, with little regulatory mechanism. Previous work has shown that there are perturbations in the enteroendocrine cell population of macroscopically normal crypts, a compartment not included in existing models. We show that existing models do not adequately recapitulate the dynamics of cell fate pathways in the crypt. We report the progressive development, iterative testing and fitting of a developed compartmental model with additional cell types, and which includes feedback mechanisms and cross-regulatory mechanisms between cell types. The fitting of the model to existing data sets suggests a need to invoke cross-talk between cell types as a feature of colon crypt cycle models.


Asunto(s)
Comunicación Celular/fisiología , Colon/patología , Células Enteroendocrinas/patología , Modelos Teóricos , Células Madre/patología , Apoptosis/fisiología , Diferenciación Celular/fisiología , Transformación Celular Neoplásica/patología , Colon/fisiopatología , Neoplasias Colorrectales/patología , Neoplasias Colorrectales/fisiopatología , Progresión de la Enfermedad , Células Enteroendocrinas/fisiología , Humanos , Células Madre/fisiología
16.
Klin Khir ; (6): 14-6, 2014 Jun.
Artículo en Ruso | MEDLINE | ID: mdl-25252544

RESUMEN

The results of investigation of endocrinal and containing serotonin cells in gastric and rectal mucosa in 48 patients in nonspecific ulcerative colitis (NUC), are presented. In accordance to histochemical and immunohistochemical investigations, quantity of endocrinal cells and those, containing serotonin, in gastric and rectal mucosa is reducing, persistence of these changes depends on NUC severity.


Asunto(s)
Colitis Ulcerosa/metabolismo , Células Enteroendocrinas/metabolismo , Mucosa Gástrica/metabolismo , Mucosa Intestinal/metabolismo , Serotonina/metabolismo , Adolescente , Adulto , Estudios de Casos y Controles , Recuento de Células , Colitis Ulcerosa/patología , Interpretación Estadística de Datos , Células Enteroendocrinas/patología , Femenino , Mucosa Gástrica/patología , Humanos , Inmunohistoquímica , Mucosa Intestinal/patología , Masculino , Persona de Mediana Edad , Adulto Joven
17.
Sci Rep ; 14(1): 11911, 2024 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-38789719

RESUMEN

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.


Asunto(s)
Diabetes Mellitus Tipo 1 , Duodeno , Mucosa Intestinal , Humanos , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Niño , Duodeno/metabolismo , Duodeno/patología , Femenino , Masculino , Proliferación Celular , Adolescente , Células Enteroendocrinas/metabolismo , Células Enteroendocrinas/patología , Intercambiador 3 de Sodio-Hidrógeno/metabolismo , Intercambiador 3 de Sodio-Hidrógeno/genética , Diferenciación Celular , Organoides/metabolismo , Organoides/patología , Antígeno Ki-67/metabolismo
18.
Gastroenterology ; 140(5): 1564-74, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21300064

RESUMEN

BACKGROUND & AIMS: Glucagon-like peptide (GLP)-1, an intestinal incretin produced by L cells through proglucagon processing, is secreted after nutrient ingestion and acts on endocrine pancreas beta cells to enhance insulin secretion. Peroxisome proliferator-activated receptor (PPAR) ß/δ is a nuclear receptor that improves glucose homeostasis and pancreas islet function in diabetic animal models. Here, we investigated whether PPARß/δ activation regulates L cell GLP-1 production. METHODS: Proglucagon regulation and GLP-1 release were evaluated in murine GLUTag and human NCI-H716 L cells and in vivo using wild-type, PPARß/δ-null, and ob/ob C57Bl/6 mice treated with the PPARß/δ synthetic agonists GW501516 or GW0742. RESULTS: PPARß/δ activation increased proglucagon expression and enhanced glucose- and bile acid-induced GLP-1 release by intestinal L cells in vitro and ex vivo in human jejunum. In vivo treatment with GW0742 increased proglucagon messenger RNA levels in the small intestine in wild-type but not in PPARß/δ-deficient mice. Treatment of wild-type and ob/ob mice with GW501516 enhanced the increase in plasma GLP-1 level after an oral glucose load and improved glucose tolerance. Concomitantly, proglucagon and GLP-1 receptor messenger RNA levels increased in the small intestine and pancreas, respectively. Finally, PPARß/δ agonists activate the proglucagon gene transcription by interfering with the ß-catenin/TCF-4 pathway. CONCLUSIONS: Our data show that PPARß/δ activation potentiates GLP-1 production by the small intestine. Pharmacologic targeting of PPARß/δ is a promising approach in the treatment of patients with type 2 diabetes mellitus, especially in combination with dipeptidyl peptidase IV inhibitors.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Células Enteroendocrinas/metabolismo , Regulación de la Expresión Génica , Péptido 1 Similar al Glucagón/biosíntesis , PPAR-beta/metabolismo , ARN Mensajero/genética , Animales , Glucemia/metabolismo , Western Blotting , Células Cultivadas , Diabetes Mellitus Experimental/patología , Modelos Animales de Enfermedad , Células Enteroendocrinas/patología , Péptido 1 Similar al Glucagón/genética , Humanos , Masculino , Ratones , Reacción en Cadena de la Polimerasa , ARN Mensajero/biosíntesis , Ratas
19.
Am J Physiol Regul Integr Comp Physiol ; 302(6): R657-66, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22160540

RESUMEN

Glucose in the gut lumen activates gut endocrine cells to release 5-HT, glucagon-like peptide 1/2 (GLP-1/2), and glucose-dependent insulinotropic polypeptide (GIP), which act to change gastrointestinal function and regulate postprandial plasma glucose. There is evidence that both release and action of incretin hormones is reduced in type 2 diabetes (T2D). We measured cellular activation of enteroendocrine and enterochromaffin cells, enteric neurons, and vagal afferent neurons in response to intestinal glucose in a model of type 2 diabetes mellitus, the UCD-T2DM rat. Prediabetic (PD), recent-diabetic (RD, 2 wk postonset), and 3-mo diabetic (3MD) fasted UCD-T2DM rats were given an orogastric gavage of vehicle (water, 0.5 ml /100 g body wt) or glucose (330 µmol/100 g body wt); after 6 min tissue was removed and cellular activation was determined by immunohistochemistry for phosphorylated calcium calmodulin-dependent kinase II (pCaMKII). In PD rats, pCaMKII immunoreactivity was increased in duodenal 5-HT (P < 0.001), K (P < 0.01) and L (P < 0.01) cells in response to glucose; glucose-induced activation of all three cell types was significantly reduced in RD and 3MD compared with PD rats. Immunoreactivity for GLP-1, but not GIP, was significantly reduced in RD and 3MD compared with PD rats (P < 0.01). Administration of glucose significantly increased pCaMKII in enteric and vagal afferent neurons in PD rats; glucose-induced pCaMKII immunoreactivity was attenuated in enteric and vagal afferent neurons (P < 0.01, P < 0.001, respectively) in RD and 3MD. These data suggest that glucose sensing in enteroendocrine and enterochromaffin cells and activation of neural pathways is markedly impaired in UCD-T2DM rats.


Asunto(s)
Vías Aferentes/fisiopatología , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/fisiopatología , Células Enteroendocrinas/metabolismo , Glucosa/metabolismo , Nervio Vago/fisiopatología , Animales , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/metabolismo , Diabetes Mellitus Tipo 2/patología , Modelos Animales de Enfermedad , Células Enterocromafines/metabolismo , Células Enterocromafines/patología , Células Enteroendocrinas/patología , Polipéptido Inhibidor Gástrico/metabolismo , Péptidos Similares al Glucagón/metabolismo , Resistencia a la Insulina/fisiología , Obesidad/metabolismo , Obesidad/fisiopatología , Ratas , Ratas Sprague-Dawley , Ratas Zucker , Serotonina/metabolismo
20.
Mol Cancer ; 10: 27, 2011 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-21401950

RESUMEN

BACKGROUND: Previous reports have suggested that the VEGF receptor neuropilin-1 (NRP-1) is expressed in a singly dispersed subpopulation of cells in the normal colonic epithelium, but that expression becomes dysregulated during colorectal carcinogenesis, with higher levels in tumour suggestive of a poor prognosis. We noted that the spatial distribution and morphology if NRP-1 expressing cells resembles that of enteroendocrine cells (EEC) which are altered in response to disease state including cancer and irritable bowel syndrome (IBS). We have shown that NRP-1 is down-regulated by butyrate in colon cancer cell lines in vitro and we hypothesized that butyrate produced in the lumen would have an analogous effect on the colon mucosa in vivo. Therefore we sought to investigate whether NRP-1 is expressed in EEC and how NRP-1 and EEC respond to butyrate and other short-chain fatty acids (SCFA - principally acetate and propionate). Additionally we sought to assess whether there is a field effect around adenomas. METHODOLOGY: Biopsies were collected at the mid-sigmoid, at the adenoma and at the contralateral wall (field) of 28 subjects during endoscopy. Samples were fixed for IHC and stained for either NRP-1 or for chromogranin A (CgA), a marker of EEC. Stool sampling was undertaken to assess individuals' butyrate, acetate and propionate levels. RESULT: NRP-1 expression was inversely related to SCFA concentration at the colon landmark (mid-sigmoid), but expression was lower and not related to SCFA concentration at the field. Likewise CgA+ cell number was also inversely related to SCFA at the landmark, but was lower and unresponsive at the field. Crypt cellularity was unaltered by field effect. A colocalisation analysis showed only a small subset of NRP-1 localised with CgA. Adenomas showed extensive, weaker staining for NRP-1 which contrastingly correlated positively with butyrate level. Field effects cause this relationship to be lost. Adenoma tissue shows dissociation of the co-regulation of NRP-1 and EEC. CONCLUSION: NRP-1 is inversely associated with levels of butyrate and other SCFA in vivo and is expressed in a subset of CgA expressing cells. EEC number is related to butyrate level in the same way.


Asunto(s)
Butiratos/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Células Enteroendocrinas/metabolismo , Células Enteroendocrinas/patología , Ácidos Grasos Volátiles/metabolismo , Neuropilinas/metabolismo , Adenoma/complicaciones , Adenoma/metabolismo , Adenoma/patología , Anciano , Recuento de Células , Cromogranina A/metabolismo , Pólipos del Colon/complicaciones , Pólipos del Colon/metabolismo , Pólipos del Colon/patología , Neoplasias Colorrectales/complicaciones , Demografía , Células Epiteliales/metabolismo , Epitelio/metabolismo , Epitelio/patología , Humanos , Transporte de Proteínas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA