Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Diabetologia ; 64(1): 181-194, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33052459

RESUMEN

AIMS/HYPOTHESIS: Melanocortin 4 receptor (MC4R) mutation is the most common cause of known monogenic obesity in humans. Unexpectedly, humans and rodents with MC4R deficiency do not develop hyperglycaemia despite chronic obesity and insulin resistance. To explain the underlying mechanisms for this phenotype, we determined the role of MC4R in glucose homeostasis in the presence and absence of obesity in mice. METHODS: We used global and hypothalamus-specific MC4R-deficient mice to investigate the brain regions that contribute to glucose homeostasis via MC4R. We performed oral, intraperitoneal and intravenous glucose tolerance tests in MC4R-deficient mice that were either obese or weight-matched to their littermate controls to define the role of MC4R in glucose regulation independently of changes in body weight. To identify the integrative pathways through which MC4R regulates glucose homeostasis, we measured renal and adrenal sympathetic nerve activity. We also evaluated glucose homeostasis in adrenaline (epinephrine)-deficient mice to investigate the role of adrenaline in mediating the effects of MC4R in glucose homeostasis. We employed a graded [13C6]glucose infusion procedure to quantify renal glucose reabsorption in MC4R-deficient mice. Finally, we measured the levels of renal glucose transporters in hypothalamus-specific MC4R-deficient mice and adrenaline-deficient mice using western blotting to ascertain the molecular mechanisms underlying MC4R control of glucose homeostasis. RESULTS: We found that obese and weight-matched MC4R-deficient mice exhibited improved glucose tolerance due to elevated glucosuria, not enhanced beta cell function. Moreover, MC4R deficiency selectively in the paraventricular nucleus of the hypothalamus (PVH) is responsible for reducing the renal threshold for glucose as measured by graded [13C6]glucose infusion technique. The MC4R deficiency suppressed renal sympathetic nerve activity by 50% in addition to decreasing circulating adrenaline and renal GLUT2 levels in mice, which contributed to the elevated glucosuria. We further report that adrenaline-deficient mice recapitulated the increased excretion of glucose in urine observed in the MC4R-deficient mice. Restoration of circulating adrenaline in both the MC4R- and adrenaline-deficient mice reversed their phenotype of improved glucose tolerance and elevated glucosuria, demonstrating the role of adrenaline in mediating the effects of MC4R on glucose reabsorption. CONCLUSIONS/INTERPRETATION: These findings define a previously unrecognised function of hypothalamic MC4R in glucose reabsorption mediated by adrenaline and renal GLUT2. Taken together, our findings indicate that elevated glucosuria due to low sympathetic tone explains why MC4R deficiency does not cause hyperglycaemia despite inducing obesity and insulin resistance. Graphical abstract.


Asunto(s)
Hexosas/metabolismo , Homeostasis/fisiología , Receptor de Melanocortina Tipo 4/fisiología , Bases de Schiff/metabolismo , Animales , Glucemia/metabolismo , Cruzamientos Genéticos , Epinefrina/deficiencia , Epinefrina/fisiología , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 2/fisiología , Glucosuria/fisiopatología , Hipotálamo/química , Insulina/sangre , Resistencia a la Insulina/fisiología , Riñón/inervación , Riñón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/fisiopatología , Receptor de Melanocortina Tipo 4/deficiencia , Sistema Nervioso Simpático/fisiopatología
2.
Diabetologia ; 61(10): 2087-2097, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30132032

RESUMEN

The concentration of glucose in plasma is held within narrow limits (4-10 mmol/l), primarily to ensure fuel supply to the brain. Kidneys play a role in glucose homeostasis in the body by ensuring that glucose is not lost in the urine. Three membrane proteins are responsible for glucose reabsorption from the glomerular filtrate in the proximal tubule: sodium-glucose cotransporters SGLT1 and SGLT2, in the apical membrane, and GLUT2, a uniporter in the basolateral membrane. 'Knockout' of these transporters in mice and men results in the excretion of filtered glucose in the urine. In humans, intravenous injection of the plant glucoside phlorizin also results in excretion of the full filtered glucose load. This outcome and the finding that, in an animal model, phlorizin reversed the symptoms of diabetes, has stimulated the development and successful introduction of SGLT2 inhibitors, gliflozins, in the treatment of type 2 diabetes mellitus. Here we summarise the current state of our knowledge about the physiology of renal glucose handling and provide background to the development of SGLT2 inhibitors for type 2 diabetes treatment.


Asunto(s)
Transportador de Glucosa de Tipo 2/fisiología , Glucosa/metabolismo , Riñón/metabolismo , Transportador 1 de Sodio-Glucosa/fisiología , Transportador 2 de Sodio-Glucosa/fisiología , Animales , Glucemia/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Diseño de Fármacos , Transportador de Glucosa de Tipo 2/genética , Glucosuria/metabolismo , Células HEK293 , Homeostasis , Humanos , Hipoglucemiantes/farmacología , Túbulos Renales/metabolismo , Túbulos Renales Proximales/metabolismo , Ratones , Ratones Noqueados , Florizina/farmacología , Transportador 1 de Sodio-Glucosa/genética , Transportador 2 de Sodio-Glucosa/genética , Inhibidores del Cotransportador de Sodio-Glucosa 2/farmacología
3.
J Hepatol ; 61(6): 1328-1336, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25076362

RESUMEN

BACKGROUND & AIMS: Microsomal prostaglandin E synthase-2 (mPGES-2) deletion does not influence in vivo PGE2 production and the function of this enzyme remains elusive. The present study was undertaken to investigate the role of mPGES-2 in streptozotocin (STZ)-induced type-1 diabetes and organ injuries. METHODS: mPGES-2 wild type (WT) and knockout (KO) mice were treated by a single intraperitoneal injection of STZ at the dose of 120 mg/kg to induce type-1 diabetes. Subsequently, glycemic status and organ injuries were evaluated. RESULTS: Following 4 days of STZ administration, mPGES-2 KO mice exhibited severe lethality in contrast to the normal phenotype observed in WT control mice. In a separate experiment, the analysis was performed at day 3 of the STZ treatment in order to avoid lethality. Blood glucose levels were similar between STZ-treated KO and WT mice. However, the livers of KO mice were yellowish with severe global hepatic steatosis, in parallel with markedly elevated liver enzymes and remarkable stomach expansion. However, the morphology of the other organs was largely normal. The STZ-treated KO mice displayed extensive hepatocyte apoptosis compared with WT mice in parallel with markedly enhanced inflammation and oxidative stress. More interestingly, a liver-specific 50% upregulation of GLUT2 was found in the KO mice accompanied with a markedly enhanced STZ accumulation and this induction of GLUT2 was likely to be associated with the insulin/SREBP-1c pathway. Primary cultured hepatocytes of KO mice exhibited an increased sensitivity to STZ-induced injury and higher cellular STZ content, which was markedly blunted by the selective GLUT2 inhibitor phloretin. CONCLUSIONS: mPGES-2 deletion enhanced STZ-induced liver toxicity possibly via GLUT2-mediated STZ uptake, independently of diabetes mellitus.


Asunto(s)
Enfermedad Hepática Inducida por Sustancias y Drogas/fisiopatología , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/fisiopatología , Transportador de Glucosa de Tipo 2/fisiología , Prostaglandina-Endoperóxido Sintasas/deficiencia , Estreptozocina/efectos adversos , Animales , Apoptosis/fisiología , Enfermedad Hepática Inducida por Sustancias y Drogas/etiología , Ciclooxigenasa 2/deficiencia , Ciclooxigenasa 2/genética , Ciclooxigenasa 2/fisiología , Diabetes Mellitus Tipo 1/complicaciones , Modelos Animales de Enfermedad , Insulina/fisiología , Hígado/enzimología , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Prostaglandina-Endoperóxido Sintasas/genética , Prostaglandina-Endoperóxido Sintasas/fisiología , Transducción de Señal/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/fisiología
4.
Crit Care Med ; 42(1): 57-65, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23963126

RESUMEN

OBJECTIVES: Providing effective enteral nutrition is important during critical illness. In health, glucose is absorbed from the small intestine via sodium-dependent glucose transporter-1 and glucose transporter-2, which may both be regulated by intestinal sweet taste receptors. We evaluated the effect of critical illness on glucose absorption and expression of intestinal sodium-dependent glucose transporter-1, glucose transporter-2, and sweet taste receptors in humans and mice. DESIGN: Prospective observational study in humans and mice. SETTING: ICU and university-affiliated research laboratory. SUBJECTS: Human subjects were 12 critically ill patients and 12 healthy controls. In the laboratory 16-week-old mice were studied. INTERVENTIONS: Human subjects underwent endoscopy. Glucose (30 g) and 3-O-methylglucose (3 g), used to estimate glucose absorption, were infused intraduodenally over 30 minutes. Duodenal mucosa was biopsied before and after infusion. Mice were randomized to cecal ligation and puncture to model critical illness (n = 16) or sham laparotomy (control) (n = 8). At day 5, mice received glucose (100 mg) and 3-O-methylglucose (10 mg) infused intraduodenally prior to mucosal tissue collection. MEASUREMENTS AND MAIN RESULTS: Quantitative polymerase chain reaction was performed to measure absolute (human) and relative levels of sodium-dependent glucose transporter-1, glucose transporter-2, and taste receptor type 1 member 2 (T1R2) transcripts. Blood samples were assayed for 3-O-methylglucose to estimate glucose absorption. Glucose absorption was three-fold lower in critically ill humans than in controls (p = 0.002) and reduced by a similar proportion in cecal ligation and puncture mice (p = 0.004). In critically ill patients, duodenal levels of sodium-dependent glucose transporter-1, glucose transporter-2, and T1R2 transcript were reduced 49% (p < 0.001), 50% (p = 0.009), and 85% (p = 0.007), whereas in the jejunum of cecal ligation and puncture mice sodium-dependent glucose transporter-1, glucose transporter-2, and T1R2 transcripts were reduced by 55% (p < 0.001), 50% (p = 0.002), and 69% (p = 0.004). CONCLUSIONS: Critical illness is characterized by markedly diminished glucose absorption, associated with reduced intestinal expression of glucose transporters (sodium-dependent glucose transporter-1 and glucose transporter-2) and sweet taste receptor transcripts. These changes are paralleled in cecal ligation and puncture mice.


Asunto(s)
Enfermedad Crítica , Glucosa/metabolismo , Absorción Intestinal/fisiología , Intestinos/fisiopatología , 3-O-Metilglucosa/metabolismo , Adulto , Anciano , Animales , Modelos Animales de Enfermedad , Duodeno/fisiopatología , Femenino , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 1/fisiología , Transportador de Glucosa de Tipo 2/metabolismo , Transportador de Glucosa de Tipo 2/fisiología , Humanos , Masculino , Ratones , Persona de Mediana Edad , Estudios Prospectivos , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores Acoplados a Proteínas G/metabolismo , Receptores Acoplados a Proteínas G/fisiología , Transportador 1 de Sodio-Glucosa/metabolismo , Transportador 1 de Sodio-Glucosa/fisiología , Adulto Joven
5.
J Surg Res ; 183(2): 606-11, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23531453

RESUMEN

BACKGROUND: When studied in enterocyte-like cell lines (Caco-2 and RIE cells), agonists and antagonists of the sweet taste receptor (STR) augment and decrease glucose uptake, respectively. We hypothesize that exposure to STR agonists and antagonists in vivo will augment glucose absorption in the rat. MATERIALS AND METHODS: About 30-cm segments of jejunum in anesthetized rats were perfused with iso-osmolar solutions containing 10, 35, and 100 mM glucose solutions (n = 6 rats, each group) with and without the STR agonist 2 mM acesulfame potassium and the STR inhibitor 10 µM U-73122 (inhibitor of the phospholipase C pathway). Carrier-mediated absorption of glucose was calculated by using stereospecific and nonstereospecific (14)C-d-glucose and (3)H-l-glucose, respectively. RESULTS: Addition of the STR agonist acesulfame potassium to the 10, 35, and 100 mM glucose solutions had no substantive effects on glucose absorption from 2.1 ± 0.2 to 2.0 ± 0.3, 5.8 ± 0.2 to 4.8 ± 0.2, and 15.5 ± 2.3 to 15.7 ± 2.7 µmoL/min/30-cm intestinal segment (P > 0.05), respectively. Addition of the STR inhibitor (U-73122) also had no effect on absorption in the 10, 35, and 100 mM solutions from 2.3 ± 0.1 to 2.1 ± 0.2, 7.7 ± 0.5 to 7.2 ± 0.5, and 15.7 ± 0.9 to 15.2 ± 1.1 µmoL/min/30-cm intestinal segment, respectively. CONCLUSIONS: Provision of glucose directly into rat jejunum does not augment glucose absorption via STR-mediated mechanisms within the jejunum in the rat. Our experiments show either no major role of STRs in mediating postprandial augmentation of glucose absorption or that proximal gastrointestinal tract stimulation of STR or other luminal factors may be required for absorption of glucose to be augmented by STR.


Asunto(s)
Células Quimiorreceptoras/fisiología , Enterocitos/citología , Glucosa/metabolismo , Absorción Intestinal/fisiología , Yeyuno/metabolismo , Animales , Células Quimiorreceptoras/efectos de los fármacos , Enterocitos/metabolismo , Estrenos/farmacología , Transportador de Glucosa de Tipo 2/fisiología , Absorción Intestinal/efectos de los fármacos , Yeyuno/citología , Masculino , Modelos Animales , Pirrolidinonas/farmacología , Ratas , Ratas Endogámicas Lew , Tiazinas/farmacología , Fosfolipasas de Tipo C/antagonistas & inhibidores
6.
FASEB J ; 24(6): 1747-58, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20097878

RESUMEN

The physiological contribution of glucose in thermoregulation is not completely established nor whether this control may involve a regulation of the melanocortin pathway. Here, we assessed thermoregulation and leptin sensitivity of hypothalamic arcuate neurons in mice with inactivation of glucose transporter type 2 (Glut2)-dependent glucose sensing. Mice with inactivation of Glut2-dependent glucose sensors are cold intolerant and show increased susceptibility to food deprivation-induced torpor and abnormal hypothermic response to intracerebroventricular administration of 2-deoxy-d-glucose compared to control mice. This is associated with a defect in regulated expression of brown adipose tissue uncoupling protein I and iodothyronine deiodinase II and with a decreased leptin sensitivity of neuropeptide Y (NPY) and proopiomelanocortin (POMC) neurons, as observed during the unfed-to-refed transition or following i.p. leptin injection. Sites of central Glut-2 expression were identified by a genetic tagging approach and revealed that glucose-sensitive neurons were present in the lateral hypothalamus, the dorsal vagal complex, and the basal medulla but not in the arcuate nucleus. NPY and POMC neurons were, however, connected to nerve terminals from Glut2-expressing neurons. Thus, our data suggest that glucose controls thermoregulation and the leptin sensitivity of NPY and POMC neurons through activation of Glut2-dependent glucose-sensing neurons located outside of the arcuate nucleus.


Asunto(s)
Regulación de la Temperatura Corporal , Transportador de Glucosa de Tipo 2/fisiología , Glucosa/metabolismo , Leptina/farmacología , Neuronas/efectos de los fármacos , Neuropéptido Y/metabolismo , Proopiomelanocortina/metabolismo , Tejido Adiposo Pardo/metabolismo , Animales , Western Blotting , Femenino , Glucosa/análisis , Humanos , Técnicas para Inmunoenzimas , Integrasas , Yoduro Peroxidasa/genética , Yoduro Peroxidasa/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Neuronas/metabolismo , Neuropéptido Y/genética , Proopiomelanocortina/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Desacopladora 1 , Yodotironina Deyodinasa Tipo II
7.
J Membr Biol ; 234(2): 57-73, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20091162

RESUMEN

Transport through lipids and aquaporins is osmotic and entirely driven by the difference in osmotic pressure. Water transport in cotransporters and uniporters is different: Water can be cotransported, energized by coupling to the substrate flux by a mechanism closely associated with protein. In the K(+)/Cl(-) and the Na(+)/K(+)/2Cl(-) cotransporters, water is entirely cotransported, while water transport in glucose uniporters and Na(+)-coupled transporters of nutrients and neurotransmitters takes place by both osmosis and cotransport. The molecular mechanism behind cotransport of water is not clear. It is associated with the substrate movements in aqueous pathways within the protein; a conventional unstirred layer mechanism can be ruled out, due to high rates of diffusion in the cytoplasm. The physiological roles of the various modes of water transport are reviewed in relation to epithelial transport. Epithelial water transport is energized by the movements of ions, but how the coupling takes place is uncertain. All epithelia can transport water uphill against an osmotic gradient, which is hard to explain by simple osmosis. Furthermore, genetic removal of aquaporins has not given support to osmosis as the exclusive mode of transport. Water cotransport can explain the coupling between ion and water transport, a major fraction of transepithelial water transport and uphill water transport. Aquaporins enhance water transport by utilizing osmotic gradients and cause the osmolarity of the transportate to approach isotonicity.


Asunto(s)
Proteínas Portadoras/metabolismo , Agua/metabolismo , Animales , Acuaporinas/fisiología , Transporte Biológico/fisiología , Difusión , Epitelio/metabolismo , Transportador de Glucosa de Tipo 2/fisiología , Humanos , Proteínas de Transporte de Membrana/metabolismo , Ósmosis , Transportador 1 de Sodio-Glucosa/fisiología , Simportadores de Cloruro de Sodio-Potasio/fisiología , Miembro 2 de la Familia de Transportadores de Soluto 12 , Simportadores/fisiología
8.
J Endod ; 46(1): 81-88, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31767340

RESUMEN

INTRODUCTION: Pulp capping materials allow healing of injured pulp with a layer of reparative dentin. Glucose is needed to cure the injured area. Glucose is transported by glucose transporter (Glut) 2 and Glut4, which are transmembrane proteins that act as gatekeepers. We hypothesized that the transport of glucose via Glut2/Glut4 might contribute to the production of a dentin bridge during wound healing. Therefore, we explored Glut2 and Glut4 expression during reparative dentinogenesis after mineral trioxide aggregate capping. METHODS: The upper left first molar of 8-week-old Wistar rats underwent pulpotomy with mineral trioxide aggregate. At 1, 3, 5, 7, and 14 days after treatment, localization and colocalization of Glut2, Glut4, nestin (odontoblast marker), and antiendothelial cell antigen 1 (RECA-1; endothelial cell marker) were analyzed with immunohistochemical staining. Messenger RNA expression levels of Slc2a2 (encoding Glut2), Slc2a4 (encoding Glut4), Igf-1r (encoding insulinlike growth factor 1 receptor), and nestin were analyzed in the extracted teeth using real-time polymerase chain reaction. RESULTS: Glut2 and Glut4 were localized within odontoblasts and endothelial cells in normal control teeth. Three days after pulpotomy, Glut2- and Glut4-positive cells were detected; 7 days after pulpotomy, immunoreactivity for Glut2 and Glut4 was confined to newly differentiated odontoblastlike cells arranged beneath reparative dentin. Messenger RNA expression levels of Slc2a2 and Slc2a4 were significantly up-regulated after pulpotomy. CONCLUSIONS: Glut2 and Glut4 regulate glucose transport during wound healing beneath the injured area. This may contribute to the development of new vital pulp therapy for patients with deep caries.


Asunto(s)
Transportador de Glucosa de Tipo 2 , Transportador de Glucosa de Tipo 4 , Glucosa , Pulpotomía , Cicatrización de Heridas , Compuestos de Aluminio , Animales , Compuestos de Calcio , Pulpa Dental , Recubrimiento de la Pulpa Dental , Combinación de Medicamentos , Células Endoteliales , Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/fisiología , Transportador de Glucosa de Tipo 4/fisiología , Humanos , Diente Molar , Óxidos , Ratas , Ratas Wistar , Silicatos
9.
Am J Physiol Regul Integr Comp Physiol ; 297(5): R1570-81, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19776250

RESUMEN

In mammals, GLUT2 plays an essential role in glucose homeostasis. From an evolutionary perspective, relatively little is known about the biology of GLUT2, or other GLUTs, in nonmammalian vertebrates. Here, we have conducted studies to functionally characterize GLUT2 in zebrafish. First, we cloned the zebrafish ortholog of GLUT2 (zfGLUT2) encoding a protein of 504 amino acids with high-sequence identity to other known vertebrate GLUT2 proteins. The zfGLUT2 gene consists of 11 exons and 10 introns, spanning 20 kb and mapping to a region of chromosome 2 that exhibits conserved synteny with human chromosome 3. When expressed in Xenopus oocytes, zfGLUT2 transported 2-deoxyglucose (2-DG) with similar affinity than mammalian GLUT2 (K(m) of 11 mM). Transport of 2-DG was competed mostly by D-fructose and D-mannose and was inhibited by cytochalasin B. During early development, zfGLUT2 expression was detected already at 10 h postfertilization and remained elevated in 5-day larvae, when it was clearly localized to the liver and intestinal bulb. In the adult, zfGLUT2 expression was highest in testis, brain, skin, kidney, and intestine, followed by liver and muscle. In the intestine, zfGLUT2 transcripts were detected in absorptive enterocytes, and its mRNA levels were altered by fasting and refeeding, suggesting that its expression in the intestine may be regulated by the nutritional status. These results indicate that the structure and function of GLUT2 has been remarkably well conserved during vertebrate evolution and open the way for the use of zebrafish as a model species in which to study the biology and pathophysiology of GLUT2.


Asunto(s)
Evolución Molecular , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/fisiología , Pez Cebra/genética , Secuencia de Aminoácidos , Animales , Exones/genética , Glucosa/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Intrones/genética , Modelos Animales , Datos de Secuencia Molecular , Trucha
10.
Nephron ; 143(1): 24-27, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30636250

RESUMEN

Diabetic nephropathy (DN), a distinct manifestation of diabetic kidney disease, affects approximately 30% of patients with diabetes. While most attention has been focused on glomerular changes related to DN, there is growing evidence that tubulopathy is a key feature in the pathogenesis of this disease. The renal proximal tubule cells (RPTCs) are particularly sensitive to the deleterious effect of chronic hyperglycemia. However, the cellular changes that control the dysfunction of the RPTCs are not fully understood. Controlling glucose reabsorption in the proximal tubules via inhibition of glucose transporters (GLUT) has emerged as a promising therapeutic in ameliorating DN. Overactivation of the renal endocannabinoid (eCB) system via the cannabinoid-1 receptor (CB1R) contributes to the development of DN, and its blockade by globally acting or peripherally restricted CB1R antagonists has been shown to ameliorate renal dysfunction in different murine models for diabetes. Recently, we have utilized various pharmacological and genetic tools to show that the eCB/CB1R system contributes to the development of DN via regulating the expression, translocation, and activity of the facilitative GLUT2 located in the RPTCs. These findings have the potential to be translated into therapy, and support the rationale for the preclinical development of novel renal-specific CB1R and/or GLUT2 inhibitors for the treatment of DN.


Asunto(s)
Nefropatías Diabéticas/etiología , Endocannabinoides/fisiología , Glucosa/metabolismo , Túbulos Renales Proximales/metabolismo , Animales , Nefropatías Diabéticas/tratamiento farmacológico , Nefropatías Diabéticas/metabolismo , Transportador de Glucosa de Tipo 2/antagonistas & inhibidores , Transportador de Glucosa de Tipo 2/fisiología , Humanos , Receptor Cannabinoide CB1/antagonistas & inhibidores , Receptor Cannabinoide CB1/fisiología , Inhibidores del Cotransportador de Sodio-Glucosa 2/uso terapéutico
11.
J Clin Invest ; 115(12): 3545-53, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16322792

RESUMEN

Ripglut1;glut2-/- mice have no endogenous glucose transporter type 2 (glut2) gene expression but rescue glucose-regulated insulin secretion. Control of glucagon plasma levels is, however, abnormal, with fed hyperglucagonemia and insensitivity to physiological hypo- or hyperglycemia, indicating that GLUT2-dependent sensors control glucagon secretion. Here, we evaluated whether these sensors were located centrally and whether GLUT2 was expressed in glial cells or in neurons. We showed that ripglut1;glut2-/- mice failed to increase plasma glucagon levels following glucoprivation induced either by i.p. or intracerebroventricular 2-deoxy-D-glucose injections. This was accompanied by failure of 2-deoxy-D-glucose injections to activate c-Fos-like immunoreactivity in the nucleus of the tractus solitarius and the dorsal motor nucleus of the vagus. When glut2 was expressed by transgenesis in glial cells but not in neurons of ripglut1;glut2-/- mice, stimulated glucagon secretion was restored as was c-Fos-like immunoreactive labeling in the brainstem. When ripglut1;glut2-/- mice were backcrossed into the C57BL/6 genetic background, fed plasma glucagon levels were also elevated due to abnormal autonomic input to the alpha cells; glucagon secretion was, however, stimulated by hypoglycemic stimuli to levels similar to those in control mice. These studies identify the existence of central glucose sensors requiring glut2 expression in glial cells and therefore functional coupling between glial cells and neurons. These sensors may be activated at different glycemic levels depending on the genetic background.


Asunto(s)
Astrocitos/metabolismo , Regulación de la Expresión Génica , Glucagón/metabolismo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/fisiología , Glucosa/metabolismo , Animales , Glucemia/metabolismo , Northern Blotting , Southern Blotting , Western Blotting , ADN Complementario/metabolismo , Desoxiglucosa/química , Glucagón/sangre , Glucagón/química , Células Secretoras de Glucagón/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Hipoglucemia/patología , Inmunohistoquímica , Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Modelos Biológicos , Modelos Genéticos , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Factores de Tiempo , Transgenes
12.
FASEB J ; 21(2): 366-77, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17172639

RESUMEN

We tested whether the dominant intestinal sugar transporter GLUT2 was inhibited by intestinal luminal compounds that are inefficiently absorbed and naturally present in foods. Because of their abundance in fruits and vegetables, flavonoids were selected as model compounds. Robust inhibition of glucose and fructose transport by GLUT2 expressed in Xenopus laevis oocytes was produced by the flavonols myricetin, fisetin, the widely consumed flavonoid quercetin, and its glucoside precursor isoquercitrin [corrected]. IC50s for quercetin, myricetin, and isoquercitirin [corrected]were approximately 200- to 1000-fold less than glucose or fructose concentrations, and noncompetitive inhibition was observed. The two other major intestinal sugar transporters, GLUT5 and SGLT1, were unaffected by flavonoids. Sugar transport by GLUT2 overexpressed in pituitary cells and naturally present in Caco-2E intestinal cells was similarly inhibited by quercetin. GLUT2 was detected on the apical side of Caco-2E cells, indicating that GLUT2 was in the correct orientation to be inhibited by luminal compounds. Quercetin itself was not transported by the three major intestinal glucose transporters. Because the flavonoid quercetin, a food component with an excellent pharmacology safety profile, might act as a potent luminal inhibitor of sugar absorption independent of its own transport, flavonols show promise as new pharmacologic agents in the obesity epidemic.


Asunto(s)
Flavonoides/farmacología , Fructosa/metabolismo , Transportador de Glucosa de Tipo 2/fisiología , Glucosa/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Células CACO-2 , Línea Celular Tumoral , Femenino , Flavonoides/química , Flavonoles , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 5/fisiología , Humanos , Mucosa Intestinal/metabolismo , Ratones , Microscopía Confocal , Modelos Biológicos , Estructura Molecular , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Quercetina/química , Quercetina/farmacología , Transportador 1 de Sodio-Glucosa/genética , Transportador 1 de Sodio-Glucosa/fisiología , Xenopus laevis
13.
J Dairy Sci ; 91(3): 1005-10, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18292256

RESUMEN

Ghrelin is an endogenous ligand of the growth hormone secretagogue receptor and a potent orexigenic (appetite-stimulating) agent in humans and rodents, but little is known about its effect in dairy cows. Ten multiparous dairy cows 35 d in milk were subjected to an i.v. glucose challenge (300 mg of D-glucose/kg of body weight). Before infusion and at regular intervals after infusion, plasma glucose, insulin, nonesterified fatty acids (NEFA), growth hormone, epinephrine, and ghrelin concentrations were monitored. Plasma insulin rose (27.2 mU/L at 10 min) and NEFA, epinephrine, and ghrelin declined (nadir = 0.22 mmol/L, 22.2 microg/L, and 272 microg/L at 31, 13, and 22 min, respectively) after the glucose infusion. Ghrelin declined for 22 min before returning to suprabasal levels at approximately 75 min postinfusion. Sequential changes of the hormones and metabolites suggested a glucose transporter, type 2- and glucose transporter, type 4-mediated disposal of glucose, and an insulin-mediated reduction in NEFA. Ghrelin and epinephrine declined after glucose infusion and before the insulin peak, but the effect of insulin as a controlling factor in the hyperglycemic reduction in these hormones cannot be discounted. The post-nadir surge in ghrelin may be regulated by the decline in circulating concentrations of glucose and NEFA (an energy-deficit signal). The profile of change in plasma ghrelin in lactating dairy cows after a glucose challenge was similar to that in monogastric animals.


Asunto(s)
Bovinos/sangre , Ghrelina/sangre , Prueba de Tolerancia a la Glucosa/veterinaria , Animales , Glucemia/análisis , Epinefrina/sangre , Ácidos Grasos no Esterificados/sangre , Femenino , Transportador de Glucosa de Tipo 2/fisiología , Transportador de Glucosa de Tipo 4/fisiología , Insulina/sangre , Cinética
14.
Physiol Genomics ; 31(2): 264-72, 2007 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-17636114

RESUMEN

Single nucleotide polymorphisms (SNPs) in two genes regulating insulin secretion, SLC2A2 (encoding GLUT2) and ABCC8 (encoding SUR1), were associated with the conversion from impaired glucose tolerance (IGT) to type 2 diabetes (T2D) in the Finnish Diabetes Prevention Study (DPS). We determined whether physical activity (PA), assessed annually with a questionnaire, modified the association of SNPs in SLC2A2 and ABCC8 with the conversion to T2D in the combined intervention and control groups of the DPS. Finnish overweight subjects with IGT (N = 479) were followed for an average of 4.1 yr. The interaction of the SNPs with the change in PA on the conversion to T2D was assessed using Cox regression with adjustments for the other components of the intervention (dietary changes, weight reduction). The carriers of the common homozygous genotype of rs5393, rs5394, or rs5404 of SLC2A2 and rs3758947 of ABCC8 who were in the lower third of the change in moderate-to-vigorous PA during the follow-up had a 2.6- to 3.7-fold increased risk of developing T2D compared with the upper third, whereas the rare allele carriers seemed to be unresponsive to changes in moderate-to-vigorous PA (for the interaction of genotype with change in PA, P = 0.022-0.027 for the SNPs in SLC2A2, and P = 0.007 for rs3758947). We conclude that moderate-to-vigorous PA may modify the risk of developing T2D associated with genes regulating insulin secretion (SLC2A2, ABCC8) in persons with IGT.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Diabetes Mellitus Tipo 2/prevención & control , Terapia por Ejercicio , Intolerancia a la Glucosa/terapia , Transportador de Glucosa de Tipo 2/genética , Polimorfismo de Nucleótido Simple , Canales de Potasio de Rectificación Interna/genética , Canales de Potasio/genética , Receptores de Droga/genética , Transportadoras de Casetes de Unión a ATP/fisiología , Terapia Combinada , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/genética , Progresión de la Enfermedad , Exones/genética , Femenino , Finlandia/epidemiología , Predisposición Genética a la Enfermedad , Intolerancia a la Glucosa/dietoterapia , Intolerancia a la Glucosa/genética , Transportador de Glucosa de Tipo 2/fisiología , Promoción de la Salud , Humanos , Insulina/metabolismo , Secreción de Insulina , Masculino , Persona de Mediana Edad , Sobrepeso , Canales de Potasio/fisiología , Canales de Potasio de Rectificación Interna/fisiología , Regiones Promotoras Genéticas/genética , Receptores de Droga/fisiología , Riesgo , Receptores de Sulfonilureas , Pérdida de Peso
15.
Diabetes ; 55(4): 988-95, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16567520

RESUMEN

A role for glucose in the control of feeding has been proposed, but its precise physiological importance is unknown. Here, we evaluated feeding behavior in glut2-null mice, which express a transgenic glucose transporter in their beta-cells to rescue insulin secretion (ripglut1;glut2-/- mice). We showed that in the absence of GLUT2, daily food intake was increased and feeding initiation and termination following a fasting period were abnormal. This was accompanied by suppressed regulation of hypothalamic orexigenic and anorexigenic neuropeptides expression during the fast-to-refed transition. In these conditions, however, there was normal regulation of the circulating levels of insulin, leptin, or glucose but a loss of regulation of plasma ghrelin concentrations. To evaluate whether the abnormal feeding behavior was due to suppressed glucose sensing, we evaluated feeding in response to intraperitoneal or intracerebroventricular glucose or 2-deoxy-D-glucose injections. We showed that in GLUT2-null mice, feeding was no longer inhibited by glucose or activated by 2-deoxy-D-glucose injections and the regulation of hypothalamic neuropeptide expression by intracerebroventricular glucose administration was lost. Together, these data demonstrate that absence of GLUT2 suppressed the function of central glucose sensors, which control feeding probably by regulating the hypothalamic melanocortin pathway. Furthermore, inactivation of these glucose sensors causes overeating.


Asunto(s)
Conducta Alimentaria/fisiología , Transportador de Glucosa de Tipo 2/fisiología , Animales , Secuencia de Bases , Cartilla de ADN , Ghrelina , Transportador de Glucosa de Tipo 2/deficiencia , Transportador de Glucosa de Tipo 2/genética , Hipotálamo/fisiología , Insulina/sangre , Leptina/sangre , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neuropéptidos/genética , Hormonas Peptídicas/sangre , Proopiomelanocortina/genética , ARN Mensajero/genética , ARN Mensajero/aislamiento & purificación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , alfa-MSH/fisiología
16.
Mol Metab ; 6(1): 61-72, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28123938

RESUMEN

OBJECTIVE: Intestinal glucose absorption is orchestrated by specialized glucose transporters such as SGLT1 and GLUT2. However, the role of GLUT2 in the regulation of glucose absorption remains to be fully elucidated. METHODS: We wanted to evaluate the role of GLUT2 on glucose absorption and glucose homeostasis after intestinal-specific deletion of GLUT2 in mice (GLUT2ΔIEC mice). RESULTS: As anticipated, intestinal GLUT2 deletion provoked glucose malabsorption as visualized by the delay in the distribution of oral sugar in tissues. Consequences of intestinal GLUT2 deletion in GLUT2ΔIEC mice were limiting body weight gain despite normal food intake, improving glucose tolerance, and increasing ketone body production. These features were reminiscent of calorie restriction. Other adaptations to intestinal GLUT2 deletion were reduced microvillus length and altered gut microbiota composition, which was associated with improved inflammatory status. Moreover, a reduced density of glucagon-like peptide-1 (GLP-1) positive cells was compensated by increased GLP-1 content per L-cell, suggesting a preserved enteroendocrine function in GLUT2ΔIEC mice. CONCLUSIONS: Intestinal GLUT2 modulates glucose absorption and constitutes a control step for the distribution of dietary sugar to tissues. Consequently, metabolic and gut homeostasis are improved in the absence of functional GLUT2 in the intestine, thus mimicking calorie restriction.


Asunto(s)
Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Glucosa/metabolismo , Animales , Glucemia/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/fisiología , Homeostasis , Absorción Intestinal , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Transportador 1 de Sodio-Glucosa/metabolismo , Distribución Tisular
17.
Diabetes ; 66(8): 2144-2149, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28385801

RESUMEN

Intestinal glucose stimulates secretion of the incretin hormone glucagon-like peptide 1 (GLP-1). The mechanisms underlying this pathway have not been fully investigated in humans. In this study, we showed that a 30-min intraduodenal glucose infusion activated half of all duodenal L cells in humans. This infusion was sufficient to increase plasma GLP-1 levels. With an ex vivo model using human gut tissue specimens, we showed a dose-responsive GLP-1 secretion in the ileum at ≥200 mmol/L glucose. In ex vivo tissue from the duodenum and ileum, but not the colon, 300 mmol/L glucose potently stimulated GLP-1 release. In the ileum, this response was independent of osmotic influences and required delivery of glucose via GLUT2 and mitochondrial metabolism. The requirement of voltage-gated Na+ and Ca2+ channel activation indicates that membrane depolarization occurs. KATP channels do not drive this, as tolbutamide did not trigger release. The sodium-glucose cotransporter 1 (SGLT1) substrate α-MG induced secretion, and the response was blocked by the SGLT1 inhibitor phlorizin or by replacement of extracellular Na+ with N-methyl-d-glucamine. This is the first report of the mechanisms underlying glucose-induced GLP-1 secretion from human small intestine. Our findings demonstrate a dominant role of SGLT1 in controlling glucose-stimulated GLP-1 release in human ileal L cells.


Asunto(s)
Duodeno/metabolismo , Péptido 1 Similar al Glucagón/metabolismo , Glucosa/administración & dosificación , Íleon/metabolismo , Edulcorantes/administración & dosificación , Canales de Calcio/fisiología , Relación Dosis-Respuesta a Droga , Glucosa/fisiología , Transportador de Glucosa de Tipo 2/fisiología , Glutamatos/metabolismo , Humanos , Infusiones Parenterales , Metilglucósidos/metabolismo , Mitocondrias/metabolismo , Florizina/metabolismo , Transportador 1 de Sodio-Glucosa/antagonistas & inhibidores , Transportador 1 de Sodio-Glucosa/metabolismo
18.
J Physiol Biochem ; 61(4): 529-37, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16669350

RESUMEN

GLUT2 is a facilitative glucose transporter located in the plasma membrane of the liver, pancreatic, intestinal, kidney cells as well as in the portal and the hypothalamus areas. Due to its low affinity and high capacity, GLUT2 transports dietary sugars, glucose, fructose and galactose in a large range of physiological concentrations, displaying large bidirectional fluxes in and out the cells. This review focuses on the roles of GLUT2. The first identified function of GLUT2 is its capacity to fuel metabolism and to provide metabolites stimulating the transcription of glucose sensitive genes. Recently, two other functions of GLUT2 are uncovered. First, the insertion of GLUT2 into the apical membrane of enterocytes induces the acute regulation of intestinal sugar absorption after a meal. Second, the GLUT2 protein itself initiates a protein signalling pathway triggering a glucose signal from the plasma membrane to the transcription machinery.


Asunto(s)
Metabolismo de los Hidratos de Carbono , Sacarosa en la Dieta/metabolismo , Transportador de Glucosa de Tipo 2/metabolismo , Animales , Transporte Biológico , Metabolismo de los Hidratos de Carbono/fisiología , Membrana Celular/metabolismo , Enterocitos/metabolismo , Transportador de Glucosa de Tipo 2/genética , Transportador de Glucosa de Tipo 2/fisiología , Humanos , Absorción Intestinal , Transducción de Señal , Transcripción Genética
19.
J Endocrinol ; 224(3): 205-14, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25486965

RESUMEN

Oral ingestion of carbohydrate triggers glucagon-like peptide 1 (GLP1) secretion, but the molecular mechanism remains elusive. By measuring GLP1 concentrations in murine portal vein, we found that the ATP-sensitive K(+) (KATP) channel is not essential for glucose-induced GLP1 secretion from enteroendocrine L cells, while the sodium-glucose co-transporter 1 (SGLT1) is required, at least in the early phase (5 min) of secretion. By contrast, co-administration of the α-glucosidase inhibitor (α-GI) miglitol plus maltose evoked late-phase secretion in a glucose transporter 2-dependent manner. We found that GLP1 secretion induced by miglitol plus maltose was significantly higher than that by another α-GI, acarbose, plus maltose, despite the fact that acarbose inhibits maltase more potently than miglitol. As miglitol activates SGLT3, we compared the effects of miglitol on GLP1 secretion with those of acarbose, which failed to depolarize the Xenopus laevis oocytes expressing human SGLT3. Oral administration of miglitol activated duodenal enterochromaffin (EC) cells as assessed by immunostaining of phosphorylated calcium-calmodulin kinase 2 (phospho-CaMK2). In contrast, acarbose activated much fewer enteroendocrine cells, having only modest phospho-CaMK2 immunoreactivity. Single administration of miglitol triggered no GLP1 secretion, and GLP1 secretion by miglitol plus maltose was significantly attenuated by atropine pretreatment, suggesting regulation via vagal nerve. Thus, while α-GIs generally delay carbohydrate absorption and potentiate GLP1 secretion, miglitol also activates duodenal EC cells, possibly via SGLT3, and potentiates GLP1 secretion through the parasympathetic nervous system.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Células Enteroendocrinas/efectos de los fármacos , Péptido 1 Similar al Glucagón/metabolismo , Inhibidores de Glicósido Hidrolasas/farmacología , Proteínas de Transporte de Sodio-Glucosa/metabolismo , 1-Desoxinojirimicina/farmacología , Acarbosa/farmacología , Animales , Células Enteroendocrinas/metabolismo , Transportador de Glucosa de Tipo 2/fisiología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Transporte de Sodio-Glucosa/genética , Transportador 1 de Sodio-Glucosa/fisiología , Xenopus laevis
20.
Diabetes ; 61(3): 692-701, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22315324

RESUMEN

In this study, we investigated the significance of ß(2)-adrenergic receptor (ß(2)AR) in age-related impaired insulin secretion and glucose homeostasis. We characterized the metabolic phenotype of ß(2)AR-null C57Bl/6N mice (ß(2)AR(-/-)) by performing in vivo and ex vivo experiments. In vitro assays in cultured INS-1E ß-cells were carried out in order to clarify the mechanism by which ß(2)AR deficiency affects glucose metabolism. Adult ß(2)AR(-/-) mice featured glucose intolerance, and pancreatic islets isolated from these animals displayed impaired glucose-induced insulin release, accompanied by reduced expression of peroxisome proliferator-activated receptor (PPAR)γ, pancreatic duodenal homeobox-1 (PDX-1), and GLUT2. Adenovirus-mediated gene transfer of human ß(2)AR rescued these defects. Consistent effects were evoked in vitro both upon ß(2)AR knockdown and pharmacologic treatment. Interestingly, with aging, wild-type (ß(2)AR(+/+)) littermates developed impaired insulin secretion and glucose tolerance. Moreover, islets from 20-month-old ß(2)AR(+/+) mice exhibited reduced density of ß(2)AR compared with those from younger animals, paralleled by decreased levels of PPARγ, PDX-1, and GLUT2. Overexpression of ß(2)AR in aged mice rescued glucose intolerance and insulin release both in vivo and ex vivo, restoring PPARγ/PDX-1/GLUT2 levels. Our data indicate that reduced ß(2)AR expression contributes to the age-related decline of glucose tolerance in mice.


Asunto(s)
Insulina/metabolismo , Receptores Adrenérgicos beta 2/fisiología , Factores de Edad , Animales , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 2/análisis , Transportador de Glucosa de Tipo 2/fisiología , Proteínas de Homeodominio/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , PPAR gamma/fisiología , Fenotipo , Transactivadores/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA