Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Sci Rep ; 12(1): 6593, 2022 04 21.
Artigo em Inglês | MEDLINE | ID: mdl-35449374

RESUMO

Cystic fibrosis (CF) is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR) gene. Defective CFTR leads to accumulation of dehydrated viscous mucus within the small intestine, luminal acidification and altered intestinal motility, resulting in blockage. These changes promote gut microbial dysbiosis, adversely influencing the normal proliferation and differentiation of intestinal epithelial cells. Using Illumina 16S rRNA gene sequencing and immunohistochemistry, we assessed changes in mucosa-attached microbiome and epithelial cell profile in the small intestine of CF mice and a CF patient compared to wild-type mice and non-CF humans. We found increased abundance of pro-inflammatory Escherichia and depletion of beneficial secondary bile-acid producing bacteria in the ileal mucosa-attached microbiome of CFTR-null mice. The ileal mucosa in a CF patient was dominated by a non-aeruginosa Pseudomonas species and lacked numerous beneficial anti-inflammatory and short-chain fatty acid-producing bacteria. In the ileum of both CF mice and a CF patient, the number of absorptive enterocytes, Paneth and glucagon-like peptide 1 and 2 secreting L-type enteroendocrine cells were decreased, whereas stem and goblet cell numbers were increased. These changes in mucosa-attached microbiome and epithelial cell profile suggest that microbiota-host interactions may contribute to intestinal CF disease development with implications for therapy.


Assuntos
Fibrose Cística , Enteropatias , Microbiota , Animais , Bactérias/genética , Contagem de Células , Fibrose Cística/microbiologia , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Células Caliciformes , Humanos , Enteropatias/complicações , Mucosa Intestinal/microbiologia , Intestino Delgado/microbiologia , Camundongos , RNA Ribossômico 16S/genética
2.
J Nutr ; 150(7): 1859-1870, 2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32510158

RESUMO

BACKGROUND: The composition of diets consumed following weight loss (WL) can have a significant impact on satiety and metabolic health. OBJECTIVE: This study was designed to test the effects of including a nondigestible carbohydrate to achieve weight maintenance (WM) following a period of WL. METHODS: Nineteen volunteers [11 females and 8 males, aged 20-62 y; BMI (kg/m2): 27-42] consumed a 3-d maintenance diet (15%:30%:55%), followed by a 21-d WL diet (WL; 30%:30%:40%), followed by 2 randomized 10-d WM diets (20%:30%:50% of energy from protein:fat:carbohydrate) containing either resistant starch type 3 (RS-WM; 22 or 26 g/d for females and males, respectively) or no RS (C-WM) in a within-subject crossover design without washout periods. The primary outcome, WM after WL, was analyzed by body weight. Secondary outcomes of fecal microbiota composition and microbial metabolite concentrations and gut hormones were analyzed in fecal samples and blood plasma, respectively. All outcomes were assessed at the end of each dietary period. RESULTS: Body weight was similar after the RS-WM and C-WM diets (90.7 and 90.8 kg, respectively), with no difference in subjectively rated appetite. During the WL diet period plasma ghrelin increased by 36% (P < 0.001), glucose-dependent insulinotropic polypeptide (GIP) decreased by 33% (P < 0.001), and insulin decreased by 46% (P < 0.001), but no significant differences were observed during the RS-WM and C-WM diet periods. Fasting blood glucose was lower after the RS-WM diet (5.59 ± 0.31 mmol/L) than after the C-WM diet [5.75 ± 0.49 mmol/L; P = 0.015; standard error of the difference between the means (SED): 0.09]. Dietary treatments influenced the fecal microbiota composition (R2 = 0.054, P = 0.031) but not diversity. CONCLUSIONS: The metabolic benefits, for overweight adults, from WL were maintained through a subsequent WM diet with higher total carbohydrate intake. Inclusion of resistant starch in the WM diet altered gut microbiota composition positively and resulted in lower fasting glucose compared with the control, with no apparent change in appetite. This trial was registered at clinicaltrials.gov as NCT01724411.


Assuntos
Fibras na Dieta/farmacologia , Microbioma Gastrointestinal , Sobrepeso/dietoterapia , Redução de Peso , Adulto , Bactérias/classificação , Bactérias/genética , DNA Bacteriano/genética , Dieta Redutora , Fibras na Dieta/administração & dosagem , Fezes/microbiologia , Feminino , Intolerância à Glucose , Humanos , Lipídeos/sangue , Masculino , Pessoa de Meia-Idade , RNA Bacteriano/genética , RNA Ribossômico 16S/genética , Adulto Jovem
3.
Sci Rep ; 7(1): 8174, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28811552

RESUMO

Morbidly obese patients exhibit impaired secretion of gut hormones that may contribute to the development of obesity. After bariatric surgery there is a dramatic increase in gut hormone release. In this study, gastric and duodenal tissues were endoscopically collected from lean, and morbidly obese subjects before and 3 months after laparoscopic sleeve gastrectomy (LSG). Tissue morphology, abundance of chromogranin A, gut hormones, α-defensin, mucin 2, Na+/glucose co-transporter 1 (SGLT1) and transcription factors, Hes1, HATH1, NeuroD1, and Ngn3, were determined. In obese patients, the total number of enteroendocrine cells (EEC) and EECs containing gut hormones were significantly reduced in the stomach and duodenum, compared to lean, and returned to normality post-LSG. No changes in villus height/crypt depth were observed. A significant increase in mucin 2 and SGLT1 expression was detected in the obese duodenum. Expression levels of transcription factors required for differentiation of absorptive and secretory cell lineages were altered. We propose that in obesity, there is deregulation in differentiation of intestinal epithelial cell lineages that may influence the levels of released gut hormones. Post-LSG cellular differentiation profile is restored. An understanding of molecular mechanisms controlling epithelial cell differentiation in the obese intestine assists in the development of non-invasive therapeutic strategies.


Assuntos
Diferenciação Celular , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Fatores de Transcrição/metabolismo , Adulto , Biomarcadores , Índice de Massa Corporal , Diferenciação Celular/genética , Cromogranina A/metabolismo , Duodeno/metabolismo , Células Enteroendócrinas/citologia , Células Enteroendócrinas/metabolismo , Feminino , Hormônios Gastrointestinais/genética , Hormônios Gastrointestinais/metabolismo , Expressão Gênica , Regulação da Expressão Gênica , Humanos , Imuno-Histoquímica , Masculino , Obesidade Mórbida/etiologia , Obesidade Mórbida/metabolismo , Obesidade Mórbida/cirurgia
4.
Exp Physiol ; 99(10): 1335-47, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25172888

RESUMO

The diet of the horse, pasture forage (grass), is fermented by the equine colonic microbiota to short-chain fatty acids, notably acetate, propionate and butyrate. Short-chain fatty acids provide a major source of energy for the horse and contribute to many vital physiological processes. We aimed to determine both the mechanism of butyrate uptake across the luminal membrane of equine colon and the nature of the protein involved. To this end, we isolated equine colonic luminal membrane vesicles. The abundance and activity of cysteine-sensitive alkaline phosphatase and villin, intestinal luminal membrane markers, were significantly enriched in membrane vesicles compared with the original homogenates. In contrast, the abundance of GLUT2 protein and the activity of Na(+)-K(+)-ATPase, known markers of the intestinal basolateral membrane, were hardly detectable. We demonstrated, by immunohistochemistry, that monocarboxylate transporter 1 (MCT1) protein is expressed on the luminal membrane of equine colonocytes. We showed that butyrate transport into luminal membrane vesicles is energized by a pH gradient (out < in) and is not Na(+) dependent. Moreover, butyrate uptake is time and concentration dependent, with a Michaelis-Menten constant of 5.6 ± 0.45 mm and maximal velocity of 614 ± 55 pmol s(-1) (mg protein)(-1). Butyrate transport is significantly inhibited by p-chloromercuribenzoate, phloretin and α-cyano-4-hydroxycinnamic acid, all potent inhibitors of MCT1. Moreover, acetate and propionate, as well as the monocarboxylates pyruvate and lactate, also inhibit butyrate uptake. Data presented here support the conclusion that transport of butyrate across the equine colonic luminal membrane is predominantly accomplished by MCT1.


Assuntos
Butiratos/farmacocinética , Colo/metabolismo , Mucosa Intestinal/metabolismo , Intestino Grosso/metabolismo , Animais , Transporte Biológico , Membrana Celular/metabolismo , Colo/efeitos dos fármacos , Transportador de Glucose Tipo 2/metabolismo , Cavalos , Humanos , Mucosa Intestinal/efeitos dos fármacos , Intestino Grosso/efeitos dos fármacos , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/metabolismo
5.
Am J Physiol Gastrointest Liver Physiol ; 304(3): G271-82, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23203156

RESUMO

CCK is secreted by endocrine cells of the proximal intestine in response to dietary components, including amino acids. CCK plays a variety of roles in digestive processes, including inhibition of food intake, consistent with a role in satiety. In the lingual epithelium, the sensing of a broad spectrum of L-amino acids is accomplished by the heteromeric amino acid (umami) taste receptor (T1R1-T1R3). T1R1 and T1R3 subunits are also expressed in the intestine. A defining characteristic of umami sensing by T1R1-T1R3 is its potentiation by IMP or GMP. Furthermore, T1R1-T1R3 is not activated by Trp. We show here that, in response to L-amino acids (Phe, Leu, Glu, and Trp), but not D-amino acids, STC-1 enteroendocrine cells and mouse proximal small intestinal tissue explants secrete CCK and that IMP enhances Phe-, Leu-, and Glu-induced, but not Trp-induced, CCK secretion. Furthermore, small interfering RNA inhibition of T1R1 expression in STC-1 cells results in significant diminution of Phe-, Leu-, and Glu-stimulated, but not Trp-stimulated, CCK release. In STC-1 cells and mouse intestine, gurmarin inhibits Phe-, Leu-, and Glu-induced, but not Trp-stimulated, CCK secretion. In contrast, the Ca(2+)-sensing receptor antagonist NPS2143 inhibits Phe-stimulated CCK release partially and Trp-induced CCK secretion totally in mouse intestine. However, NPS2143 has no effect on Leu- or Glu-induced CCK secretion. Collectively, our data demonstrate that functional characteristics and cellular location of the gut-expressed T1R1-T1R3 support its role as a luminal sensor for Phe-, Leu-, and Glu-induced CCK secretion.


Assuntos
Aminoácidos/farmacologia , Colecistocinina/metabolismo , Trato Gastrointestinal/fisiologia , Receptores Acoplados a Proteínas G/fisiologia , Aminoácidos/antagonistas & inibidores , Animais , Western Blotting , Membrana Celular/metabolismo , Células Cultivadas , Feminino , Trato Gastrointestinal/efeitos dos fármacos , Imuno-Histoquímica , Fosfatos de Inositol/metabolismo , Isomerismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microscopia Confocal , Naftalenos/farmacologia , Peptídeos/farmacologia , Proteínas de Plantas/farmacologia , Hidrolisados de Proteína/farmacologia , Interferência de RNA , Receptores Acoplados a Proteínas G/efeitos dos fármacos , Estimulação Química
6.
Am J Physiol Regul Integr Comp Physiol ; 303(2): R199-208, 2012 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-22552794

RESUMO

The heteromeric sweet taste receptor T1R2-T1R3 is expressed on the luminal membrane of certain populations of enteroendocrine cells. Sensing of sugars and other sweet compounds by this receptor activates a pathway in enteroendocrine cells, resulting in secretion of a number of gut hormones, including glucagon-like peptide 2 (GLP-2). This subsequently leads to upregulation in the expression of intestinal Na(+)/glucose cotransporter, SGLT1, and increased intestinal glucose absorption. On the basis of the current information available on the horse genome sequence, it has been proposed that the gene for T1R2 (Tas1R2) is absent in the horse. We show here, however, that horses express both the mRNA and protein for T1R2. Equine T1R2 is most closely homologous to that in the pig and the cow. T1R2 protein, along with T1R3, α-gustducin, and GLP-2 proteins are coexpressed in equine intestinal endocrine cells. Intravenous administration of GLP-2, in rats and pigs, leads to an increase in the expression of SGLT1 in absorptive enterocytes and enhancement in blood glucose concentrations. GLP-2 receptor is expressed in enteric neurons, excluding the direct effect of GLP-2 on enterocytes. However, electric stimulation of enteric neurons generates a neural response leading to SGLT1 upregulation, suggesting that sugar in the intestine activates a reflex increase in the functional expression of SGLT1. Horses possess the ability to upregulate SGLT1 expression in response to increased dietary carbohydrates, and to enhance the capacity of the gut to absorb glucose. The gut sweet receptor provides an accessible target for manipulating the equine gut to absorb glucose (and water), allowing greater energy uptake and hydration for hard-working horses.


Assuntos
Células Enteroendócrinas/metabolismo , Glucose/metabolismo , Cavalos/metabolismo , Intestino Delgado/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Sequência de Aminoácidos , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/fisiologia , Gatos , Carboidratos da Dieta/farmacocinética , Metabolismo Energético/efeitos dos fármacos , Metabolismo Energético/fisiologia , Células Enteroendócrinas/citologia , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Feminino , Peptídeo 2 Semelhante ao Glucagon/metabolismo , Intestino Delgado/citologia , Masculino , Dados de Sequência Molecular , Receptores Acoplados a Proteínas G/análise , Transportador 1 de Glucose-Sódio/metabolismo , Suínos , Língua/citologia , Língua/metabolismo , Transducina/metabolismo
7.
Br J Nutr ; 104(5): 637-46, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20338074

RESUMO

In an intensive livestock production, a shorter suckling period allows more piglets to be born. However, this practice leads to a number of disorders including nutrient malabsorption, resulting in diarrhoea, malnutrition and dehydration. A number of strategies have been proposed to overcome weaning problems. Artificial sweeteners, routinely included in piglets' diet, were thought to enhance feed palatability. However, it is shown in rodent models that when included in the diet, they enhance the expression of Na+/glucose co-transporter (SGLT1) and the capacity of the gut to absorb glucose. Here, we show that supplementation of piglets' feed with a combination of artificial sweeteners saccharin and neohesperidin dihydrochalcone enhances the expression of SGLT1 and intestinal glucose transport function. Artificial sweeteners are known to act on the intestinal sweet taste receptor T1R2/T1R3 and its partner G-protein, gustducin, to activate pathways leading to SGLT1 up-regulation. Here, we demonstrate that T1R2, T1R3 and gustducin are expressed together in the enteroendocrine cells of piglet intestine. Furthermore, gut hormones secreted by the endocrine cells in response to dietary carbohydrates, glucagon-like peptides (GLP)-1, GLP-2 and glucose-dependent insulinotrophic peptide (GIP), are co-expressed with type 1 G-protein-coupled receptors (T1R) and gustducin, indicating that L- and K-enteroendocrine cells express these taste elements. In a fewer endocrine cells, T1R are also co-expressed with serotonin. Lactisole, an inhibitor of human T1R3, had no inhibitory effect on sweetener-induced SGLT1 up-regulation in piglet intestine. A better understanding of the mechanism(s) involved in sweetener up-regulation of SGLT1 will allow the identification of nutritional targets with implications for the prevention of weaning-related malabsorption.


Assuntos
Suplementos Nutricionais , Intestino Delgado/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Edulcorantes/farmacologia , Suínos/metabolismo , Transducina/metabolismo , Animais , Derivados de Benzeno/farmacologia , Transporte Biológico/efeitos dos fármacos , Chalconas/farmacologia , Carboidratos da Dieta/metabolismo , Células Enteroendócrinas/metabolismo , Feminino , Polipeptídeo Inibidor Gástrico/metabolismo , Peptídeos Semelhantes ao Glucagon/metabolismo , Hesperidina/análogos & derivados , Hesperidina/farmacologia , Masculino , Sacarina/farmacologia , Serotonina/metabolismo , Regulação para Cima , Desmame
8.
Gastroenterology ; 133(6): 1916-27, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18054563

RESUMO

BACKGROUND & AIMS: Butyrate oxidation is impaired in intestinal mucosa of patients with inflammatory bowel diseases (IBD). Butyrate uptake by colonocytes involves the monocarboxylate transporter (MCT) 1. We aimed to investigate the role of MCT1 in butyrate oxidation deficiency during colonic inflammation. METHODS: Colonic tissues were collected from patients with IBD or healthy controls and from rats with dextran sulfate sodium (DSS)-induced colitis. The intestinal epithelial cell line HT-29 was treated with interferon-gamma (IFN-gamma) and tumor necrosis factor-alpha (TNF-alpha). MCT1 expression was analyzed by real-time reverse-transcription polymerase chain reaction, Western blot, and immunofluorescence. Butyrate uptake and oxidation in HT-29 cells was assessed using [(14)C]-butyrate. The mechanism of MCT1 gene regulation was analyzed by nuclear run-on and reporter gene assays. RESULTS: MCT1 messenger RNA (mRNA) and protein levels were markedly decreased in inflamed colonic mucosa of IBD patients and rats. In HT-29 cells, down-regulation of MCT1 mRNA and protein abundance by IFN-gamma and TNF-alpha correlated with a decrease in butyrate uptake and subsequent oxidation. IFN-gamma and TNF-alpha did not affect MCT1 mRNA stability but rather down-regulated gene transcription. We demonstrate that the cytokine response element is located in the proximal -111/+213 core region of the MCT1 promoter. CONCLUSIONS: The data suggest that butyrate oxidation deficiency in intestinal inflammation is a consequence of reduction in MCT1-mediated butyrate uptake. This reinforces the proposition that butyrate oxidation deficiency in IBD is not a primary defect.


Assuntos
Butiratos/metabolismo , Doenças Inflamatórias Intestinais/imunologia , Transportadores de Ácidos Monocarboxílicos/genética , Transportadores de Ácidos Monocarboxílicos/imunologia , Simportadores/genética , Simportadores/imunologia , Adulto , Idoso , Animais , Células Cultivadas , Colite/imunologia , Modelos Animais de Doenças , Regulação para Baixo , Feminino , Expressão Gênica , Humanos , Mucosa Intestinal/imunologia , Masculino , Pessoa de Meia-Idade , Transportadores de Ácidos Monocarboxílicos/biossíntese , Oxirredução , Ratos , Ratos Sprague-Dawley , Simportadores/biossíntese
9.
DNA Cell Biol ; 25(1): 49-62, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16405400

RESUMO

Butyrate is a naturally occurring product of colonic microbial fermentation of dietary carbohydrates that escape hydrolysis in the small intestine. Butyrate plays a significant role in the maintenance of colonic tissue homeostasis by regulating the expression of genes associated with the processes of proliferation, differentiation, and apoptosis. Using microarray analysis, we assessed changes in the expression of 19,400 genes in response to butyrate in a human colonic epithelial cell line. Among these, we have identified 221 potentially butyrate- responsive genes specifically associated with the processes of proliferation, differentiation, and apoptosis. Of these genes, 59 are upregulated and 162 downregulated, in accordance with the known modes of action of butyrate. The changes in the expression levels (up- or downregulation) of many of these genes were found to be opposite to that reported in colon cancer tissue, where the intracellular concentration of butyrate would be reduced due to the decline in expression of the colonic butyrate transporter, MCT1.


Assuntos
Butiratos/metabolismo , Colo/metabolismo , Células Epiteliais/metabolismo , Perfilação da Expressão Gênica , Mucosa Intestinal/metabolismo , Apoptose , Diferenciação Celular , Linhagem Celular , Proliferação de Células , Colo/citologia , Regulação da Expressão Gênica , Humanos , Mucosa Intestinal/citologia , Análise de Sequência com Séries de Oligonucleotídeos
10.
Gastroenterology ; 128(3): 676-86, 2005 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-15765403

RESUMO

BACKGROUND & AIMS: Butyrate serves as the major source of energy for colonic epithelial cells, and has profound effects on their proliferation, differentiation, and apoptosis. Transport of butyrate across the colonocyte luminal membrane is mediated by the monocarboxylate transporter, MCT1; the expression of which is down-regulated dramatically during colon carcinogenesis. We have proposed that the decline in MCT1 expression during colon carcinogenesis may reduce the intracellular availability of butyrate required to regulate expression of genes associated with the processes maintaining tissue homeostasis within the colonic mucosa. METHODS: To test this hypothesis we used the technique of RNA interference to inhibit MCT1 expression specifically, and determined the consequences of this inhibition on the ability of butyrate to exert its recognized effects in vitro using flow cytometry, immunofluorescence, Northern analysis, and Western analysis. RESULTS: We show that inhibition of MCT1 expression, and hence butyrate uptake, has profound inhibitory effects on the ability of butyrate to regulate expression of key target genes: p21waf1/cip1 (p21), intestinal alkaline phosphatase (IAP), and cyclin D1, and their associated processes of proliferation and differentiation. In contrast, inhibition of MCT1 expression had no effect on the ability of butyrate to modulate expression of either bcl-XL or bak, and this was reflected in a corresponding lack of effect on butyrate induction of apoptosis. CONCLUSIONS: Collectively, these results show the importance of MCT1 to the ability of butyrate to induce cell-cycle arrest and differentiation, and suggest fundamental differences in the mechanisms by which butyrate modulates specific aspects of cell function.


Assuntos
Colo/metabolismo , Homeostase , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/metabolismo , Fosfatase Alcalina/genética , Antígenos de Neoplasias/genética , Apoptose/efeitos dos fármacos , Butiratos/farmacologia , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/genética , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular , Colo/efeitos dos fármacos , Ciclina D1/genética , Inibidor de Quinase Dependente de Ciclina p21 , Proteínas Ligadas por GPI , Expressão Gênica/efeitos dos fármacos , Células HT29 , Humanos , Transportadores de Ácidos Monocarboxílicos/antagonistas & inibidores , Interferência de RNA , Simportadores/antagonistas & inibidores
11.
Eur J Biochem ; 270(16): 3377-88, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12899695

RESUMO

Dietary sugars regulate expression of the intestinal Na+/glucose cotransporter, SGLT1, in many species. Using sheep intestine as a model, we showed that lumenal monosaccharides, both metabolisable and nonmetabolisable, regulate SGLT1 expression. This regulation occurs not only at the level of transcription, but also at the post-transcriptional level. Introduction of d-glucose and some d-glucose analogues into ruminant sheep intestine resulted in > 50-fold enhancement of SGLT1 expression. We aimed to determine if transport of sugar into the enterocytes is required for SGLT1 induction, and delineate the signal-transduction pathways involved. A membrane impermeable d-glucose analogue, di(glucos-6-yl)poly(ethylene glycol) 600, was synthesized and infused into the intestines of ruminant sheep. SGLT1 expression was determined using transport studies, Northern and Western blotting, and immunohistochemistry. An intestinal cell line, STC-1, was used to investigate the signalling pathways. Intestinal infusion with di(glucos-6-yl)poly(ethylene glycol) 600 led to induction of functional SGLT1, but the compound did not inhibit Na+/glucose transport into intestinal brush-border membrane vesicles. Studies using cells showed that increased medium glucose up-regulated SGLT1 abundance and SGLT1 promoter activity, and increased intracellular cAMP levels. Glucose-induced activation of the SGLT1 promoter was mimicked by the protein kinase A (PKA) agonist, 8Br-cAMP, and was inhibited by H-89, a PKA inhibitor. Pertussis toxin, a G-protein (Gi)-specific inhibitor, enhanced SGLT1 protein abundance to levels observed in response to glucose or 8Br-cAMP. We conclude that lumenal glucose is sensed by a glucose sensor, distinct from SGLT1, residing on the external face of the lumenal membrane. The glucose sensor initiates a signalling pathway, involving a G-protein-coupled receptor linked to a cAMP-PKA pathway resulting in enhancement of SGLT1 expression.


Assuntos
Glucose/metabolismo , Mucosa Intestinal/metabolismo , Animais , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Feminino , Glucose/análogos & derivados , Glucosídeos/síntese química , Glucosídeos/metabolismo , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Proteínas de Transporte de Monossacarídeos/genética , Proteínas de Transporte de Monossacarídeos/metabolismo , Polietilenoglicóis/síntese química , Polietilenoglicóis/metabolismo , Regiões Promotoras Genéticas , Receptores de Superfície Celular/metabolismo , Ovinos/metabolismo , Sódio/metabolismo , Transportador 1 de Glucose-Sódio
12.
J Physiol ; 539(Pt 2): 361-71, 2002 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11882670

RESUMO

Butyrate is the principal source of energy for colonic epithelial cells, and has profound effects on their proliferation, differentiation and apoptosis. Transport of butyrate across the colonocyte luminal membrane is mediated by the monocarboxylate transporter 1 (MCT1). We have examined the regulation of expression of human colonic MCT1 by butyrate, in cultured colonic epithelial cells (AA/C1). Treatment with sodium butyrate (NaBut) resulted in a concentration- and time-dependent upregulation of both MCT1 mRNA and protein. At 2 mM butyrate, the magnitude of induction of mRNA (5.7-fold) entirely accounted for the 5.2-fold increase in protein abundance, and was mediated by both activation of transcription and enhanced mRNA stability. The other monocarboxylates found naturally in the colon, acetate and propionate, had no effect. The properties of butyrate uptake by AA/C1 cells were characteristic of MCT1. Induction of the MCT1 protein resulted in a corresponding increase in the maximal rate of butyrate transport. The V(max) for uptake of [U-(14)C]butyrate was increased 5-fold following pre-incubation with 2 mM NaBut, with no significant change in the apparent K(m). In conclusion, this study is the first to show substrate-induced regulation of human colonic MCT1. The basis of this regulation is a butyrate-induced increase in MCT1 mRNA abundance, resulting from the dual control of MCT1 gene transcription and stability of the MCT1 transcript. We suggest that butyrate-induced increases in the expression and resulting activity of MCT1 serve as a mechanism to maximise intracellular availability of butyrate, to act both as a source of energy and to influence processes maintaining cellular homeostasis in the colonic epithelium.


Assuntos
Butiratos/metabolismo , Colo/metabolismo , Transportadores de Ácidos Monocarboxílicos/metabolismo , Simportadores/metabolismo , Northern Blotting , Western Blotting , Linhagem Celular , Núcleo Celular/metabolismo , Polaridade Celular , Colo/citologia , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Humanos , Membranas/metabolismo , Microscopia Eletrônica , Transportadores de Ácidos Monocarboxílicos/biossíntese , Transportadores de Ácidos Monocarboxílicos/genética , Processamento de Proteína Pós-Traducional/fisiologia , RNA Mensageiro/biossíntese , Simportadores/biossíntese , Simportadores/genética , Transcrição Gênica/fisiologia , Regulação para Cima/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA