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

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Cell Physiol ; 234(4): 4396-4408, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30352123

RESUMO

GLUT12 was cloned from the mammary cancer cell line MCF-7, but its physiological role still needs to be elucidated. To gain more knowledge of GLUT12 function in the intestine, we investigated GLUT12 subcellular localization in the small intestine and its regulation by sugars, hormones, and intracellular mediators in Caco-2 cells and mice. Immunohistochemical methods were used to determine GLUT12 subcellular localization in human and murine small intestine. Brush border membrane vesicles were isolated for western blot analyses. Functional studies were performed in Caco-2 cells by measuring α-methyl-d-glucose (αMG) uptake in the absence of sodium. GLUT12 is located in the apical cytoplasm, below the brush border membrane, and in the perinuclear region of murine and human enterocytes. In Caco-2 cells, GLUT12 translocation to the apical membrane and α-methyl- d-glucose uptake by the transporter are stimulated by protons, glucose, insulin, tumor necrosis factor-α (TNF-α), protein kinase C, and AMP-activated protein kinase. In contrast, hypoxia decreases GLUT12 expression in the apical membrane. Upregulation of TNF-α and hypoxia-inducible factor-1α ( HIF-1α) genes is found in the jejunal mucosa of diet-induced obese mice. In these animals, GLUT12 expression in the brush border membrane is slightly decreased compared with lean animals. Moreover, an intraperitoneal injection of insulin does not induce GLUT12 translocation to the membrane, as it occurs in lean animals. GLUT12 rapid translocation to the enterocytes' apical membrane in response to glucose and insulin could be related to GLUT12 participation in sugar absorption during postprandial periods. In obesity, in which insulin sensitivity is reduced, the contribution of GLUT12 to sugar absorption is affected.


Assuntos
Colo/metabolismo , Enterócitos/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Absorção Intestinal , Intestino Delgado/metabolismo , Metilglucosídeos/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Células CACO-2 , Hipóxia Celular , Colo/citologia , Colo/efeitos dos fármacos , Modelos Animais de Doenças , Enterócitos/efeitos dos fármacos , Regulação da Expressão Gênica , Proteínas Facilitadoras de Transporte de Glucose/efeitos dos fármacos , Proteínas Facilitadoras de Transporte de Glucose/genética , Humanos , Insulina/farmacologia , Intestino Delgado/citologia , Intestino Delgado/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Obesidade/genética , Obesidade/metabolismo , Proteína Quinase C/metabolismo , Transporte Proteico , Ratos Wistar , Fator de Necrose Tumoral alfa/farmacologia
2.
J Cell Physiol ; 234(4): 4352-4361, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30246472

RESUMO

We have previously demonstrated in Caco-2 cells that tumor necrosis factor-α (TNF-α) inhibits sugar uptake, acting from the apical membrane, by decreasing the expression of the Na+ -glucose cotransporter SGLT1 in the brush border membrane. The goal was to investigate the hypothesis that TNF-α from abdominal adipose tissue (adipocytes and macrophages) would decrease sugar and amino acid transport acting from the basolateral membrane of the enterocytes. TNF-α placed in the basal compartment of Caco-2 cells decreased α-methyl- d-glucose (αMG) and glutamine uptake. The apical medium derived from these Caco-2 cells apically placed in another set of cells, also reduced sugar and glutamine transport. Reverse-transcription polymerase chain reaction analysis demonstrated upregulation of TNF-α, IL-1ß, and MCP1 expression in Caco-2 cells exposed to basal TNF-α. Similarly, αMG uptake was inhibited after Caco-2 cells were incubated, in the basal compartment, with medium from visceral human mesenchymal stem cells-derived adipocytes of overweight individuals. The apical medium collected from those Caco-2 cells, and placed in the upper side of other set of cells, also decreased sugar uptake. Basal presence of medium derived from lipopolysaccharide-activated macrophages and nonactivated macrophages decreased αMG uptake as well. Diet-induced obese mice showed an increase in the visceral adipose tissue surrounding the intestine. In this physiological condition, there was a reduction on αMG uptake in jejunal everted rings. Altogether, these results suggest that basolateral TNF-α, which can be produced by adipocytes and macrophages during obesity, would be able to activate TNF-α and other proinflammatory proteins expression in the small intestine and diminish intestinal sugar and amino acids transport.


Assuntos
Adipócitos/metabolismo , Absorção Intestinal , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Metilglucosídeos/metabolismo , Comunicação Parácrina , Transportador 1 de Glucose-Sódio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Animais , Células CACO-2 , Meios de Cultivo Condicionados/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Glutamina/metabolismo , Humanos , Masculino , Camundongos Endogâmicos C57BL , Obesidade/metabolismo , Via Secretória , Transdução de Sinais , Células THP-1 , Fator de Necrose Tumoral alfa/genética
3.
J Cell Physiol ; 233(3): 2426-2433, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28771713

RESUMO

The aim of the present work was to investigate in Caco-2 cells whether eicosapentaenoic acid (EPA), an omega-3 polyunsaturated fatty acid, could block the inhibitory effect of tumor necrosis factor-α (TNF-α) on sugar transport, and identify the intracellular signaling pathways involved. After pre-incubation of the Caco-2 cells with TNF-α and EPA for 1 hr, EPA prevented the inhibitory effect of the cytokine on α-methyl-d-glucose (αMG) uptake (15 min) and on SGLT1 expression at the brush border membrane, measured by Western blot. The ERK1/2 inhibitor PD98059 and the AMPK activator AICAR also prevented the inhibitory effect of TNF-α on both αMG uptake and SGLT1 expression. Interestingly, the AMPK inhibitor, Compound C, abolished the ability of EPA to prevent TNF-α-induced reduction of sugar uptake and transporter expression. The GPR120 antagonist, AH7614, also blocked the preventive effect of EPA on TNF-α-induced decrease of αMG uptake and AMPK phosphorylation. In summary, TNF-α inhibits αMG uptake by decreasing SGLT1 expression in the brush border membrane through the activation of ERK1/2 pathway. EPA prevents the inhibitory effect of TNF-α through the involvement of GPR120 and AMPK activation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Açúcares da Dieta/metabolismo , Ácido Eicosapentaenoico/farmacologia , Células Epiteliais/efeitos dos fármacos , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Metilglucosídeos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Fator de Necrose Tumoral alfa/farmacologia , Transporte Biológico , Células CACO-2 , Ativação Enzimática , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Mucosa Intestinal/metabolismo , Transdução de Sinais , Transportador 1 de Glucose-Sódio/metabolismo
4.
Am J Physiol Cell Physiol ; 308(12): C1008-22, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25855082

RESUMO

GLUT12 is a member of the facilitative family of glucose transporters. The goal of this study was to characterize the functional properties of GLUT12, expressed in Xenopus laevis oocytes, using radiotracer and electrophysiological methods. Our results showed that GLUT12 is a facilitative sugar transporter with substrate selectivity: d-glucose ≥ α-methyl-d-glucopyranoside (α-MG) > 2-deoxy-d-glucose(2-DOG) > d-fructose = d-galactose. α-MG is a characteristic substrate of the Na(+)/glucose (SGLT) family and has not been shown to be a substrate of any of the GLUTs. In the absence of sugar, (22)Na(+) was transported through GLUT12 at a higher rate (40%) than noninjected oocytes, indicating that there is a Na(+) leak through GLUT12. Genistein, an inhibitor of GLUT1, also inhibited sugar uptake by GLUT12. Glucose uptake was increased by the PKA activator 8-bromoadenosine 3',5'-cyclic monophosphate (8-Br-cAMP) but not by the PKC activator phorbol-12-myristate-13-acetate (PMA). In high K(+) concentrations, glucose uptake was blocked. Addition of glucose to the external solution induced an inward current with a reversal potential of approximately -15 mV and was blocked by Cl(-) channel blockers, indicating the current was carried by Cl(-) ions. The sugar-activated Cl(-) currents were unaffected by genistein. In high external K(+) concentrations, sugar-activated Cl(-) currents were also blocked, indicating that GLUT12 activity is voltage dependent. Furthermore, glucose-induced current was increased by the PKA activator 8-Br-cAMP but not by the PKC activator PMA. These new features of GLUT12 are very different from those described for other GLUTs, indicating that GLUT12 must have a specific physiological role within glucose homeostasis, still to be discovered.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Glucose/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Transporte Biológico , Canais de Cloreto/antagonistas & inibidores , Canais de Cloreto/metabolismo , Cloretos/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Ativação Enzimática , Ativadores de Enzimas/farmacologia , Genisteína/farmacologia , Glucose/análogos & derivados , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Humanos , Concentração de Íons de Hidrogênio , Cinética , Potenciais da Membrana , Oócitos , Técnicas de Patch-Clamp , Sódio/metabolismo , Xenopus laevis
5.
Mol Pharm ; 12(6): 2158-66, 2015 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-25923048

RESUMO

The abundance and function of transporter proteins at the plasma membrane are likely to be crucial in drug responsiveness. Functional detection of human concentrative nucleoside transporters (hCNTs) is of interest for predicting drug sensitivity because of their ability to transport most nucleoside-derived drugs. In the present study, two fluorescent nucleoside analogues, uridine-furan and etheno-cytidine, were evaluated as tools to study in vivo nucleoside transporter-related functions. These two molecules showed high affinity interactions with hCNT1 and hCNT3 and were shown to be substrates of both transporters. Both fluorescence microscopy and flow cytometry experiments showed that uridine-furan uptake was better suited for distinguishing cells that express hCNT1 or hCNT3. These data highlight the usefulness of fluorescent nucleoside derivatives, as long as they fulfill the requirements of confocal microscopy and flow cytometry, for in vivo analysis of hCNT-related function.


Assuntos
Citometria de Fluxo/métodos , Microscopia Confocal/métodos , Proteínas de Transporte de Nucleosídeos/metabolismo , Nucleosídeos/química , Humanos
6.
Cytokine ; 64(1): 181-7, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23910014

RESUMO

PURPOSE: During intestinal inflammation TNFα levels are increased and as a consequence malabsorption of nutrients may occur. We have previously demonstrated that TNFα inhibits galactose, fructose and leucine intestinal absorption in animal models. In continuation with our work, the purpose of the present study was to investigate in the human intestinal epithelial cell line Caco-2, the effect of TNFα on sugar transport and to identify the intracellular mechanisms involved. METHODS: Caco-2 cells were grown on culture plates and pre-incubated during different periods with various TNFα concentrations before measuring the apical uptake of galactose, α-methyl-glucoside (MG) or fructose for 15 min. To elucidate the signaling pathway implicated, cells were pre-incubated for 30min with the PKA inhibitor H-89 or the PKC inhibitor chelerythrine, before measuring the sugar uptake. The expression in the apical membrane of the transporters implicated in the sugars uptake process (SGLT1 and GLUT5) was determined by Western blot. RESULTS: TNFα inhibited 0.1mM MG uptake after pre-incubation of the cells for 6-48h with the cytokine and in the absence of cytokine pre-incubation. In contrast, 5mM fructose uptake was stimulated by TNFα only after long pre-incubation times (24 and 48 h). These effects were mediated by the binding of the cytokine to its specific receptor TNFR1, present in the apical membrane of the Caco-2 cells. Analysis of the expression of the MG and fructose transporters at the brush border membrane of the cells, after 24h pre-incubation with the cytokine, revealed decrease on the amount of SGLT1 and increase on the amount of GLUT5 proteins. Short-term inhibition of MG transport by TNFα was not modified by H-89 but was blocked by chelerythrine. CONCLUSIONS: SGLT1 and GLUT5 expression in the plasma membrane is regulated by TNFα in the human epithelial cell line Caco-2 cells, leading to alteration on sugars transport, suggesting that TNFα could be considered as a physiological local regulator of nutrients absorption in response to an intestinal inflammatory status.


Assuntos
Transportador de Glucose Tipo 5/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Benzofenantridinas/farmacologia , Transporte Biológico/efeitos dos fármacos , Células CACO-2 , Linhagem Celular , Frutose/metabolismo , Galactose/metabolismo , Transportador de Glucose Tipo 5/biossíntese , Humanos , Inflamação/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Isoquinolinas/farmacologia , Metilglucosídeos/metabolismo , Proteínas de Transporte de Monossacarídeos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Transportador 1 de Glucose-Sódio/biossíntese , Sulfonamidas/farmacologia
7.
Pflugers Arch ; 460(3): 617-32, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20495821

RESUMO

Human concentrative nucleoside transporter 3 (hCNT3) uses the electrochemical gradient of Na(+) and H(+) to drive the transport of nucleosides and therapeutic nucleoside analogs into the cells. We employed the two-electrode voltage clamp technique to compare the steady-state and presteady-state kinetics of hCNT3 in the presence of Na(+) and H(+). We found that H(+) supported a higher maximal rate of uridine transport than Na(+), but the efficiency of transport was lower. For both cations, maximal uridine-induced current increased with hyperpolarizing potentials and did not saturate within the voltage range tested. Apparent affinity of hCNT3 for uridine in H(+) was insensitive to membrane voltage at negative potentials, and decreased with depolarization. In contrast, apparent affinity for uridine in Na(+) decreased with hyperpolarization and was independent of voltage at depolarizing potentials. H(+)-coupled hCNT3 exhibited lower affinity for all natural nucleosides and different substrate selectivity compared to Na(+)-coupled hCNT3. In H(+), lack of the hydroxyl groups at 2' and 5' decreased the affinity, while lack of the nitrogen N-7 or inversion of the configuration of the hydroxyl group at 2' prevented transport. Presteady-state charge movements of hCNT3 did not decrease when extracellular cation concentration (Na(+) or H(+)) was reduced, but the tau(ON)-V and Q-V curves were shifted to more negative potentials. The different effects of uridine and inosine on presteady-state currents in H(+) indicated a change in rate-limiting step for the transport of these substrates by H(+)-coupled hCNT3.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Prótons , Sódio/metabolismo , Uridina/metabolismo , Adenosina/metabolismo , Animais , Feminino , Humanos , Cinética , Técnicas de Patch-Clamp , Xenopus laevis
8.
Am J Physiol Gastrointest Liver Physiol ; 299(1): G179-85, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20448142

RESUMO

L-glutamine is the primary metabolic fuel for enterocytes. Glutamine from the diet is transported into the absorptive cells by two sodium-dependent neutral amino acid transporters present at the apical membrane: ASCT2/SLC1A5 and B(0)AT1/SLC6A19. We have demonstrated that leptin is secreted into the stomach lumen after a meal and modulates the transport of sugars after binding to its receptors located at the brush border of the enterocytes. The present study was designed to address the effect of luminal leptin on Na(+)-dependent glutamine (Gln) transport in rat intestine and identify the transporters involved. We found that 0.2 nM leptin inhibited uptake of Gln and phenylalanine (Phe) (substrate of B(0)AT1) using everted intestinal rings. In Ussing chambers, 10 mM Gln absorption followed as Na(+)-induced short-circuit current was inhibited by leptin in a dose-dependent manner (maximum inhibition at 10 nM; I(C50) = approximately 0.1 nM). Phe absorption was also decreased by leptin. Western blot analysis after 3-min incubation of the intestinal loops with 10 mM Gln, showed marked increase of ASCT2 and B(0)AT1 protein in the brush-border membrane that was reduced by rapid preincubation of the intestinal lumen with 1 nM leptin. Similarly, the increase in ASCT2 and B(0)AT1 gene expression induced by 60-min incubation of the intestine with 10 mM Gln was strongly reduced after a short preincubation period with leptin. Altogether these data demonstrate that, in rat, leptin controls the active Gln entry through reduction of both B(0)AT1 and ASCT2 proteins traffic to the apical plasma membrane and modulation of their gene expression.


Assuntos
Sistema ASC de Transporte de Aminoácidos/metabolismo , Sistemas de Transporte de Aminoácidos Neutros/metabolismo , Glutamina/metabolismo , Intestino Delgado/metabolismo , Leptina/metabolismo , Sistema ASC de Transporte de Aminoácidos/genética , Sistemas de Transporte de Aminoácidos Neutros/genética , Animais , Transporte Biológico , Regulação da Expressão Gênica , Técnicas In Vitro , Mucosa Intestinal/metabolismo , Intestino Delgado/efeitos dos fármacos , Masculino , Potenciais da Membrana , Antígenos de Histocompatibilidade Menor , Peptídeos/farmacologia , Fenilalanina/metabolismo , Transporte Proteico , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores para Leptina/antagonistas & inibidores , Receptores para Leptina/metabolismo , Proteínas Recombinantes/metabolismo , Membrana Serosa/metabolismo , Sódio/metabolismo , Fatores de Tempo
9.
Appl Physiol Nutr Metab ; 45(9): 957-967, 2020 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-32176854

RESUMO

Obesity is characterized by excessive fat accumulation and inflammation. Aging has also been characterized as an inflammatory condition, frequently accompanied by accumulation of visceral fat. Beneficial effects of exercise and n-3 long-chain polyunsaturated fatty acids in metabolic disorders have been described. Glucose transporter 12 (GLUT12) is one of the less investigated members of the GLUT family. Glucose, insulin, and tumor necrosis factor alpha (TNF-α) induce GLUT12 translocation to the membrane in muscle, adipose tissue, and intestine. We aimed to investigate GLUT12 expression in obesity and aging, and under diet supplementation with docosahexaenoic acid (DHA) alone or in combination with physical exercise in mice. Aging increased GLUT12 expression in intestine, kidney, and adipose tissue, whereas obesity reduced it. No changes on the transporter occurred in skeletal muscle. In obese 18-month-old mice, DHA further decreased GLUT12 in the 4 organs. Aerobic exercise alone did not modify GLUT12, but the changes triggered by exercise were able to prevent the DHA-diminishing effect, and almost restored GLUT12 basal levels. In conclusion, the downregulation of metabolism in aging would be a stimulus to upregulate GLUT12 expression. Contrary, obesity, an excessive energy condition, would induce GLUT12 downregulation. The combination of exercise and DHA would contribute to restore basal function of GLUT12. Novelty In small intestine, kidney and adipose tissue aging increases GLUT12 protein expression whereas obesity reduces it. Dietary DHA decreases GLUT12 in small intestine, kidney, adipose tissue and skeletal muscle. Exercise alone does not modify GLUT12 expression, nevertheless exercise prevents the DHA-diminishing effect on GLUT12.


Assuntos
Envelhecimento/metabolismo , Ácidos Docosa-Hexaenoicos/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Obesidade/metabolismo , Condicionamento Físico Animal , Tecido Adiposo/metabolismo , Animais , Células CACO-2 , Dieta , Feminino , Humanos , Intestino Delgado/metabolismo , Rim/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo
10.
Mol Neurobiol ; 57(2): 798-805, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31473905

RESUMO

The brain depends on glucose as a source of energy. This implies the presence of glucose transporters, being GLUT1 and GLUT3 the most relevant. Expression of GLUT12 is found in mouse and human brain at low levels. We previously demonstrated GLUT12 upregulation in the frontal cortex of aged subjects that was even higher in aged Alzheimer's disease (AD) patients. However, the cause and the mechanism through which this increase occurs are still unknown. Here, we aimed to investigate whether the upregulation of GLUT12 in AD is related with aging or Aß deposition in comparison with GLUT1, GLUT3, and GLUT4. In the frontal cortex of two amyloidogenic mouse models (Tg2576 and APP/PS1) GLUT12 levels were increased. Contrary, expression of GLUT1 and GLUT3 were decreased, while GLUT4 did not change. In aged mice and the senescence-accelerated model SAMP8, GLUT12 and GLUT4 were upregulated in comparison with young animals. GLUT1 and GLUT3 did not show significant changes with age. The effect of ß-amyloid (Aß) deposition was also evaluated in Aß peptide i.c.v. injected mice. In the hippocampus, GLUT12 expression increased whereas GLUT4 was not modified. Consistent with the results in the amyloidogenic models, GLUT3 and GLUT1 were downregulated. In summary, Aß increases GLUT12 protein expression in the brain pointing out a central role of the transporter in AD pathology and opening new perspectives for the treatment of this neurodegenerative disease.


Assuntos
Doença de Alzheimer/metabolismo , Encéfalo/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Doença de Alzheimer/patologia , Peptídeos beta-Amiloides/administração & dosagem , Animais , Encéfalo/patologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Modelos Animais de Doenças , Injeções Intraventriculares , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
11.
J Nutr Biochem ; 76: 108264, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31760230

RESUMO

Tumor necrosis factor-alfa (TNF-α) is a pro-inflammatory cytokine highly-involved in intestinal inflammation. Omega-3 polyunsaturated fatty acids (n3-PUFAs) show anti-inflammatory actions. We previously demonstrated that the n3-PUFA EPA prevents TNF-α inhibition of sugar uptake in Caco-2 cells. Here, we investigated whether the n3-PUFA DHA and its derived specialized pro-resolving lipid mediators (SPMs) MaR1, RvD1 and RvD2, could block TNF-α inhibition of intestinal sugar and glutamine uptake. DHA blocked TNF-α-induced inhibition of α-methyl-D-glucose (αMG) uptake and SGLT1 expression in the apical membrane of Caco-2 cells, through a pathway independent of GPR120. SPMs showed the same preventive effect but acting at concentrations 1000 times lower. In diet-induced obese (DIO) mice, oral gavage of MaR1 reversed the up-regulation of pro-inflammatory cytokines found in intestinal mucosa of these mice. However, MaR1 treatment was not able to counteract the reduced intestinal transport of αMG and SGLT1 expression in the DIO mice. In Caco-2 cells, TNF-α also inhibited glutamine uptake being this inhibition prevented by EPA, DHA and the DHA-derived SPMs. Interestingly, TNF-α increased the expression in the apical membrane of the glutamine transporter B0AT1. This increase was partially blocked by the n-3 PUFAs. These data reveal DHA and its SPMs as promising biomolecules to restore intestinal nutrients transport during intestinal inflammation.


Assuntos
Ácidos Docosa-Hexaenoicos/farmacologia , Glutamina/metabolismo , Lipídeos/química , Açúcares/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Animais , Transporte Biológico/efeitos dos fármacos , Biotinilação , Células CACO-2 , Dieta , Ácido Eicosapentaenoico/farmacologia , Ácidos Graxos Ômega-3/farmacologia , Humanos , Inflamação , Mucosa Intestinal/metabolismo , Intestinos/química , Intestinos/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Transportador 1 de Glucose-Sódio/metabolismo , Fator de Necrose Tumoral alfa/metabolismo
12.
Acta Physiol (Oxf) ; 226(4): e13283, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31002200

RESUMO

AIM: The facilitative glucose transporter GLUT12 was isolated from the breast cancer cell line MCF-7 by its homology with GLUT4. GLUT12 is expressed in insulin-sensitive tissues such as adipose tissue. The aim of this work was to investigate GLUT12 expression and hormonal regulation in 3T3-L1 adipocytes and in adipose tissue of lean and diet-induced obese mice. METHODS: Uptake studies were performed using radio-labelled sugars; α-methyl-d-glucose (αMG) was used as specific substrate of GLUT12. Expression and localization of GLUT12 in adipocytes were investigated by western blot and immunohistochemical methods. RESULTS: GLUT12 is expressed in the peri-nuclear region of mouse adipocytes. Insulin, by AKT activation, and TNF-α, by AMPK activation, increase αMG uptake by inducing GLUT12 translocation to the membrane. In contrast, leptin and adiponectin decrease GLUT12 activity through its internalization. Under hypoxia conditions GLUT12 expression is upregulated. The response of GLUT12 to TNF-α, leptin, adiponectin and hypoxia is the opposite to that of GLUT4. In diet-induced obese mice and obese subjects, GLUT12 protein is decreased. Intraperitoneal injection of insulin increases AKT phosphorylation and GLUT12 expression, but this effect is lost in obese animals. CONCLUSION: We hypothesize that GLUT12 would contribute to modulate sugar absorption in physiological and pathophysiological situations such as obesity.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Obesidade/metabolismo , Células 3T3-L1 , Animais , Humanos , Camundongos
14.
J Physiol Biochem ; 71(2): 311-7, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25935421

RESUMO

Leptin is secreted into the digestive tract and contributes to the absorption of dietary molecules by regulating transporters activity. Here, we studied the effect of luminal leptin on the intestinal transport of L-glutamate, an important component of human diet. We examined the effect of leptin on L-glutamate uptake in rat intestine in vitro measuring glutamate-induced short-circuit current (Isc) in Ussing chambers and L-[(3)H (U)]-glutamate uptake in jejunal everted rings. Glutamate-induced Isc was only observed in Na(+)-free conditions. This Isc was concentration (1-60 mmol L(-1)) and pH dependent. Luminal leptin increased glutamate Isc (∼100 %). Dose-response curve showed a biphasic pattern, with maximal stimulations observed at 10(-13) and 10(-10) mmol L(-1), that were sensitive to leptin receptor antagonist. In everted rings, two glutamate transport mechanisms were distinguished: a Na(+)-dependent, H(+)-independent, that was inhibited by leptin (∼20 %), and a Na(+)-independent but H(+)-dependent, that was enhanced by leptin (∼20 %), in line with data obtained in Ussing chambers. Altogether, these data reveal original non-monotonic effect of luminal leptin in the intestine and demonstrate a new role for this hormone in the modulation of L-glutamate transport, showing that luminal active gut peptides can influence absorption of amino acids.


Assuntos
Ácido Glutâmico/metabolismo , Intestino Delgado/metabolismo , Leptina/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Relação Dose-Resposta a Droga , Ácido Glutâmico/farmacocinética , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Intestino Delgado/efeitos dos fármacos , Leptina/antagonistas & inibidores , Leptina/farmacologia , Masculino , Técnicas de Cultura de Órgãos/instrumentação , Técnicas de Cultura de Órgãos/métodos , Ratos Wistar , Sódio/metabolismo
15.
J Endocrinol ; 224(1): 17-23, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25349247

RESUMO

Leptin is secreted by the gastric mucosa and is able to reach the intestinal lumen and bind to its receptors located in the apical membranes of enterocytes. We have previously demonstrated that apical leptin inhibits uptake of amino acids in rat intestine in vitro and in Caco-2 cells. The aim of the present work was to investigate the effect of leptin on absorption of amino acids using in vivo techniques, which generate situations closer to physiological conditions. In vivo intestinal absorption of amino acids in rats was measured by isolating a jejunal loop and using the single-pass perfusion system. Disappearance of glutamine (Gln), proline (Pro), and ß-alanine (ß-Ala) from the perfusate, in the absence or presence of leptin, was measured using a radioactivity method. Luminal leptin (25 nM) inhibited the absorption of 2 mM Pro, 5 mM ß-Ala, and 5 mM Gln by approximately 45% after 5-15 min; the effect remained constant until the end of the experiment (80 min) and was rapidly and completely reversed when leptin was removed from the perfusion medium. Moreover, leptin was able to regulate the absorption of galactose and Gln in the same animal, indicating a direct action of the hormone on the specific transporters implicated in the uptake of each nutrient. The results of the present work indicate that luminal leptin decreases absorption of amino acids in vivo in a short-term manner and in a reversible way. These results, together with our previous findings, make it evident that leptin can be considered as a hormone which provides the intestine with a control mechanism to handle absorption of nutrients.


Assuntos
Aminoácidos/metabolismo , Absorção Intestinal/fisiologia , Leptina/fisiologia , Alanina/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Glutamina/metabolismo , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Leptina/farmacologia , Masculino , Prolina/metabolismo , Ratos , Ratos Wistar , Fatores de Tempo
16.
J Cancer ; 6(2): 139-43, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25561978

RESUMO

BACKGROUND: Recent studies proposed GLUT12 to be a major glucose transporter involved in the glycolytic metabolism of cancer cells. METHODS: GLUT12 expression was determined by immunohistochemistry in a selection of cancer cell lines and a tumour spheroid model. RESULTS: GLUT12 expression was high in A549 and RH-36; low in HT29; and absent in NB-EB cancer cell lines. GLUT12 expression was located in the necrotic centre of HT29 spheroids, which is characterised by anaerobic metabolism. CONCLUSION: The data supports the involvement of GLUT12 in the glycolytic metabolism of cancer cells and therefore, its potential as a novel therapeutic target for cancer treatment.

17.
J Alzheimers Dis ; 42(1): 97-101, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24820014

RESUMO

Alzheimer's disease (AD) might be conceptualized as a metabolic disease with progressive impairment of the brain's capacity to utilize glucose. One of the last glucose transporters discovered is GLUT12. The aim of the present work was to investigate the expression of GLUT12 in frontal cortex from AD patients. Human samples from young control donors barely expressed GLUT12. The level of expression of GLUT12 was significantly higher in AD compare to aged controls. Expression of GLUT12 and Ox-42, a microglia marker, correlate in controls but not in AD. The implications of these findings in AD are discussed further.


Assuntos
Doença de Alzheimer/metabolismo , Lobo Frontal/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Envelhecimento/metabolismo , Western Blotting , Antígeno CD11b/metabolismo , Feminino , Humanos , Masculino , Microglia/metabolismo
18.
J Physiol Biochem ; 69(3): 507-12, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23359137

RESUMO

Leptin is secreted by gastric mucosa and is able to reach the intestinal lumen where its receptors are located in the apical membrane of the enterocytes. We have previously demonstrated that apical leptin inhibits sugar and amino acids uptake in vitro and glucose absorption in vivo. Since leptin receptors are also expressed in the basolateral membrane of the enterocytes, the aim of the present work was to investigate whether leptin acting from the basolateral side could also regulate amino acid uptake. Tritiated Gln and ß-Ala were used to measure uptake into Caco-2 cells grown on filters, in the presence of basal leptin at short incubation times (5 and 30 min) and after 6 h of preincubation with the hormone. In order to compare apical and basal leptin effect, Gln and ß-Ala uptake was measured in the presence of leptin acting from the apical membrane also in cells grown on filters. Basal leptin (8 mM) inhibited by ~15-30% the uptake of 0.1 mM Gln and 1 mM ß-Ala quickly, after 5 min exposure, and the effect was maintained after long preincubation periods. Apical leptin had the same effect. Moreover, the inhibition was rapidly and completely reversed when leptin was removed from the apical or basolateral medium. These results extend our previous findings and contribute to the vision of leptin as an important hormonal signal for the regulation of intestinal absorption of nutrients.


Assuntos
Glicina/metabolismo , Leptina/metabolismo , Receptores para Leptina/metabolismo , beta-Alanina/metabolismo , Transporte Biológico , Células CACO-2 , Polaridade Celular , Humanos , Cinética , Contagem de Cintilação , Trítio
19.
J Agric Food Chem ; 61(49): 12012-9, 2013 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-24261475

RESUMO

Several plant extracts rich in flavonoids have been reported to improve hyperglycemia by inhibiting digestive enzyme activities and SGLT1-mediated glucose uptake. In this study, helichrysum ( Helichrysum italicum ) and grapefruit ( Citrus × paradisi ) extracts inhibited in vitro enzyme activities. The helichrysum extract showed higher inhibitory activity of α-glucosidase (IC50 = 0.19 mg/mL) than α-amylase (IC50 = 0.83 mg/mL), whereas the grapefruit extract presented similar α-amylase and α-glucosidase inhibitory activities (IC50 = 0.42 mg/mL and IC50 = 0.41 mg/mL, respectively). Both extracts reduced maltose digestion in noneverted intestinal sacs (57% with helichrysum and 46% with grapefruit). Likewise, both extracts inhibited SGLT1-mediated methylglucoside uptake in Caco-2 cells in the presence of Na(+) (56% of inhibition with helichrysum and 54% with grapefruit). In vivo studies demonstrated that helichrysum decreased blood glucose levels after an oral maltose tolerance test (OMTT), and both extracts reduced postprandial glucose levels after the oral starch tolerance test (OSTT). Finally, both extracts improved hyperinsulinemia (31% with helichrysum and 50% with grapefruit) and HOMA index (47% with helichrysum and 54% with grapefruit) in a dietary model of insulin resistance in rats. In summary, helichrysum and grapefruit extracts improve postprandial glycemic control in rats, possibly by inhibiting α-glucosidase and α-amylase enzyme activities and decreasing SGLT1-mediated glucose uptake.


Assuntos
Glicemia/metabolismo , Citrus paradisi/química , Helichrysum/química , Hiperglicemia/tratamento farmacológico , Hipoglicemiantes/administração & dosagem , Extratos Vegetais/administração & dosagem , Animais , Digestão , Regulação para Baixo/efeitos dos fármacos , Inibidores de Glicosídeo Hidrolases , Humanos , Hiperglicemia/enzimologia , Hiperglicemia/metabolismo , Hiperglicemia/fisiopatologia , Absorção Intestinal/efeitos dos fármacos , Masculino , Período Pós-Prandial/efeitos dos fármacos , Ratos , Ratos Wistar , alfa-Amilases/antagonistas & inibidores , alfa-Amilases/metabolismo , alfa-Glucosidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA