Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 337
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Eur J Pharmacol ; 879: 173141, 2020 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-32353360

RESUMO

Dapagliflozin (DAPA) or canagliflozin (CANA), Na+-dependent glucose co-transporter type 2 (SGLT2) inhibitors, were used for treatment of type II diabetes mellitus. Addition of DAPA or CANA suppressed M-type K+ current (IK(M)) in pituitary tumor (GH3) and pheochromocytoma PC12 cells. The IC50 value for DAPA- or CANA-mediated inhibition of IK(M) in GH3 cells was 0.11 or 0.42 µM, respectively. The presence of DAPA (0.1 µM) shifted the steady-state activation of IK(M) to less depolarized potential without changing the gating charge of the current. During high-frequency depolarizing pulses, IK(M) magnitude was reduced by DAPA; however, DAPA-induced block of IK(M) remained effective. The amplitude of neither erg-mediated K+ current nor hyperpolarization-activated cation current in GH3 cells was modified in the presence of 1 µM DAPA. Alternatively, addition of DAPA, CANA, phlorizin or chlorotoxin effectively suppressed α-methylglucoside-(αMG-) induced current (IαMG) in GH3 cells, albeit inability of tefluthrin (activator of INa) to suppress this current. DAPA shifted the charge-voltage relation of presteady-state IαMG in a rightward and downward direction with no change in the gating charge of the IαMG. Under current-clamp recordings, subsequent additions of DAPA, but still in the continued presence of αMG, increased the firing rate of spontaneous action potentials stimulated by αMG. Our results suggested that activity of SGLT was expressed functionally in GH3 and PC12 cells. Therefore, inhibitory actions of DAPA or CANA on the amplitude and gating of IK(M) might provide a yet unidentified mechanism through which the SGLT1 or SGLT2 activity were attenuated in unclamped cells occurring in vivo.


Assuntos
Neoplasias das Glândulas Suprarrenais/fisiopatologia , Compostos Benzidrílicos/farmacologia , Canagliflozina/farmacologia , Glucosídeos/farmacologia , Feocromocitoma/fisiopatologia , Neoplasias Hipofisárias/fisiopatologia , Canais de Potássio/fisiologia , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Neoplasias das Glândulas Suprarrenais/metabolismo , Animais , Linhagem Celular Tumoral , Metilglucosídeos/farmacologia , Feocromocitoma/metabolismo , Neoplasias Hipofisárias/metabolismo , Ratos
2.
Histol Histopathol ; 34(4): 431-443, 2019 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-30394506

RESUMO

Following an injury or resection, the mammalian liver has the capacity to regain its former volume and functioning by restoring itself. Studies have demonstrated that antioxidants play a role in hepatic regeneration. This study investigated the effect of 4-(3,4-dihydroxybenzoyloxymethyl) phenyl-O-ß-D-glucopyranoside (PG) obtained from Origanum micranthum on liver regeneration. Sixty Wistar Albino rats were used. In the sham-operated group, a midline abdominal laparotomy was performed without hepatectomy. In the partial hepatectomy (PHx) group, the median and left lateral lobes were removed. Rats in the PHx group received 20 mg/kg/day PG intraperitoneally before being sacrificed at 24, 48, and 72 hrs, and 7 days later. Liver tissues were collected for immunohistochemical analysis and electron microscopic evaluation. We found an increase in mitotic index, and the numbers of Ki-67 stained hepatocytes in all PHx early stage groups (24 hr, 48hr, 72 hr), but not in 7-day groups. The regeneration mediators eNOS, iNOS, TNF-α and NF-κB were shown to increase in PHx groups. This increase was more prominent dependening on time. In the PHx treatment (PHx+PG) groups, while eNOS was still high, iNOS, TNF-α and NF-κB had decreased. The apoptotic index was markedly high in the PHx groups; this was prevented by PG treatment. These findings were supported by the ultrastructural results. Our findings indicate that PG supports liver regeneration, hepatocyte proliferation, reduced liver damage, and inflammatory mediators following PHx.


Assuntos
Proliferação de Células/efeitos dos fármacos , Regeneração Hepática/efeitos dos fármacos , Metilglucosídeos/farmacologia , Animais , Hepatectomia , Hepatócitos/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar
3.
Eur J Pharmacol ; 799: 103-110, 2017 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-28174043

RESUMO

We recently reported that cerebral sodium-glucose transporter type 1 (SGLT-1) plays a role in exacerbation of cerebral ischemia. However, the mechanism by which cerebral SGLT-1 acts remains unclear. Here we demonstrated that sodium influx through cerebral SGLT-1 exacerbates cerebral ischemic neuronal damage. SGLT-specific sodium ion influx was induced using α-methyl-D-glucopyranoside (α-MG). Intracellular sodium concentrations in primary cortical neurons were estimated using sodium-binding benzofuran isophthalate fluorescence. SGLT-1 knockdown in primary cortical neurons and mice was achieved using SGLT-1 siRNA. The survival rates of primary cultured cortical neurons were assessed using biochemical assays 1 day after treatment. Middle cerebral artery occlusion (MCAO) was used to generate a focal cerebral ischemic model in SGLT-1 knockdown mice. The change in fasting blood glucose levels, infarction development, and behavioral abnormalities were assessed 1 day after MCAO. Treatment with 200mM α-MG induced a continuous increase in the intracellular sodium concentration, and this increase was normalized after α-MG removal. Neuronal SGLT-1 knockdown had no effect on 100µM H2O2-induced neuronal cell death; however, the knockdown prevented the neuronal cell death induced by 17.5mM glucose and the co-treatment of 100µM H2O2/8.75mM glucose. Neuronal SGLT-1 knockdown also suppressed the cell death induced by α-MG alone and the co-treatment of 100µM H2O2/0.01mM α-MG. Our in vivo results showed that the exacerbation of cerebral ischemic neuronal damage induced by the intracerebroventricular administration of 5.0µg α-MG/mouse was ameliorated in cerebral SGLT-1 knockdown mice. Thus, sodium influx through cerebral SGLT-1 may exacerbate cerebral ischemia-induced neuronal damage.


Assuntos
Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Neurônios/patologia , Transportador 1 de Glucose-Sódio/metabolismo , Sódio/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Transporte Biológico/genética , Encéfalo/efeitos dos fármacos , Encéfalo/metabolismo , Encéfalo/patologia , Isquemia Encefálica/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Técnicas de Silenciamento de Genes , Hiperglicemia/complicações , Espaço Intracelular/efeitos dos fármacos , Espaço Intracelular/metabolismo , Masculino , Metilglucosídeos/farmacologia , Camundongos , Neurônios/efeitos dos fármacos , RNA Interferente Pequeno/genética , Proteínas de Transporte de Sódio-Glucose/metabolismo , Transportador 1 de Glucose-Sódio/deficiência , Transportador 1 de Glucose-Sódio/genética
4.
PLoS One ; 11(11): e0165592, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27806092

RESUMO

BACKGROUND: The global epidemic of Type-2-Diabetes (T2D) highlights the need for novel therapeutic targets and agents. Roux-en-Y-Gastric-Bypass (RYGB) is the most effective treatment. Studies investigating the mechanisms of RYGB suggest a role for post-operative changes in portal glucose levels. We investigate the impact of stimulating portal glucose sensors on systemic glucose levels in health and T2D, and evaluated the role of sodium-glucose-cotransporter-3 (SGLT3) as the possible sensor. METHODS: Systemic glucose and hormone responses to portal stimulation were measured. In Sprague-Dawley (SD) rats, post-prandial state was simulated by infusing glucose into the portal vein. The SGLT3 agonist, alpha-methyl-glucopyranoside (αMG), was then added to further stimulate the portal sensor. To elucidate the neural pathway, vagotomy or portal denervation was followed by αMG+glucose co-infusion. The therapeutic potential of portal glucose sensor stimulation was investigated by αMG-only infusion (vs. saline) in SD and Zucker-Diabetic-Fatty (ZDF) rats. Hepatic mRNA expression was also measured. RESULTS: αMG+glucose co-infusion reduced peak systemic glucose (vs. glucose alone), and lowered hepatic G6Pase expression. Portal denervation, but not vagotomy, abolished this effect. αMG-only infusion lowered systemic glucose levels. This glucose-lowering effect was more pronounced in ZDF rats, where portal αMG infusion increased insulin, C-peptide and GIP levels compared to saline infusions. CONCLUSIONS: The portal vein is capable of sensing its glucose levels, and responds by altering hepatic glucose handling. The enhanced effect in T2D, mediated through increased GIP and insulin, highlights a therapeutic target that could be amenable to pharmacological modulation or minimally-invasive surgery.


Assuntos
Glicemia/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Metilglucosídeos/administração & dosagem , Obesidade/metabolismo , Veia Porta/metabolismo , Proteínas de Transporte de Sódio-Glucose/metabolismo , Animais , Diabetes Mellitus Experimental/cirurgia , Diabetes Mellitus Tipo 2/cirurgia , Derivação Gástrica , Masculino , Metilglucosídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Ratos Zucker , Transdução de Sinais , Vagotomia
5.
Mol Pharmacol ; 90(5): 508-521, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27555600

RESUMO

Na+-d-glucose cotransporter 1 (SGLT1) is rate-limiting for glucose absorption in the small intestine. Shortly after intake of glucose-rich food, SGLT1 abundance in the luminal membrane of the small intestine is increased. This upregulation occurs via glucose-induced acceleration of the release of SGLT1-containing vesicles from the trans-Golgi network (TGN), which is regulated by a domain of protein RS1 (RSC1A1) named RS1-Reg. Dependent on phosphorylation, RS1-Reg blocks release of vesicles containing SGLT1 or concentrative nucleoside transporter 1. The hypothesis has been raised that RS1-Reg binds to different receptor proteins at the TGN, which trigger release of vesicles with different transporters. To identify the presumed receptor proteins, two-hybrid screening was performed. Interaction with ornithine decarboxylase 1 (ODC1), the rate-limiting enzyme of polyamine synthesis, was observed and verified by immunoprecipitation. Binding of RS1-Reg mutants to ODC1 was characterized using surface plasmon resonance. Inhibition of ODC1 activity by RS1-Reg mutants and the ODC1 inhibitor difluoromethylornithine (DFMO) was measured in the absence and presence of glucose. In addition, short-term effects of DFMO, RS1-Reg mutants, the ODC1 product putrescine, and/or glucose on SGLT1 expressed in oocytes of Xenopus laevis were investigated. High-affinity binding of RS1-Reg to ODC1 was demonstrated, and evidence for a glucose binding site in ODC1 was provided. Binding of RS1-Reg to ODC1 inhibits the enzymatic activity at low intracellular glucose, which is blunted at high intracellular glucose. The data suggest that generation of putrescine by ODC1 at the TGN stimulates release of SGLT1-containing vesicles. This indicates a biomedically important role of ODC1 in regulation of glucose homeostasis.


Assuntos
Regulação para Baixo/efeitos dos fármacos , Exocitose/efeitos dos fármacos , Glucose/farmacologia , Proteínas de Transporte de Monossacarídeos/metabolismo , Ornitina Descarboxilase/metabolismo , Transportador 1 de Glucose-Sódio/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Células CACO-2 , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Eflornitina/farmacologia , Eletroforese em Gel de Poliacrilamida , Células HEK293 , Humanos , Imunoprecipitação , Espaço Intracelular/metabolismo , Cinética , Metilglucosídeos/farmacologia , Modelos Biológicos , Proteínas de Transporte de Monossacarídeos/química , Oócitos/efeitos dos fármacos , Oócitos/metabolismo , Florizina/farmacologia , Ligação Proteica/efeitos dos fármacos , Domínios Proteicos , Proteínas Recombinantes/metabolismo , Frações Subcelulares/efeitos dos fármacos , Frações Subcelulares/metabolismo , Ressonância de Plasmônio de Superfície , Xenopus laevis
6.
J Pharm Pharmacol ; 68(7): 922-31, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27139580

RESUMO

OBJECTIVES: We recently demonstrated that the cerebral sodium-glucose transporter (SGLT) is involved in postischaemic hyperglycaemia-induced exacerbation of cerebral ischaemia. However, the associated SGLT-mediated mechanisms remain unclear. Thus, we examined the involvement of cerebral SGLT-induced excessive sodium ion influx in the development of cerebral ischaemic neuronal damage. METHODS: [Na+]i was estimated according to sodium-binding benzofuran isophthalate fluorescence. In the in vitro study, primary cortical neurons were prepared from fetuses of ddY mice. Primary cortical neurons were cultured for 5 days before each treatment with reagents, and these survival rates were assessed using biochemical assays. In in vivo study, a mouse model of focal ischaemia was generated using middle cerebral artery occlusion (MCAO). KEY FINDINGS: In these experiments, treatment with high concentrations of glucose induced increment in [Na+]i, and this phenomenon was suppressed by the SGLT-specific inhibitor phlorizin. SGLT-specific sodium ion influx was induced using a-methyl-D-glucopyranoside (a-MG) treatments, which led to significant concentration-dependent declines in neuronal survival rates and exacerbated hydrogen peroxide-induced neuronal cell death. Moreover, phlorizin ameliorated these effects. Finally, intracerebroventricular administration of a-MG exacerbated the development of neuronal damage induced by MCAO, and these effects were ameliorated by the administration of phlorizin. CONCLUSIONS: Hence, excessive influx of sodium ions into neuronal cells through cerebral SGLT may exacerbate the development of cerebral ischaemic neuronal damage.


Assuntos
Córtex Cerebral/metabolismo , Infarto da Artéria Cerebral Média/fisiopatologia , Transportador 1 de Glucose-Sódio/metabolismo , Sódio/metabolismo , Animais , Glicemia , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Isquemia Encefálica/fisiopatologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Córtex Cerebral/patologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Glucose/antagonistas & inibidores , Glucose/farmacologia , Peróxido de Hidrogênio/efeitos adversos , Peróxido de Hidrogênio/antagonistas & inibidores , Infarto da Artéria Cerebral Média/patologia , Infusões Intraventriculares , Metilglucosídeos/administração & dosagem , Metilglucosídeos/antagonistas & inibidores , Metilglucosídeos/farmacologia , Camundongos , Neurônios/metabolismo , Neurônios/patologia , Florizina/administração & dosagem , Florizina/farmacologia , Cultura Primária de Células
7.
Microbiology (Reading) ; 162(6): 1037-1046, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27045200

RESUMO

Escherichia coli strain 15 (ATCC 9723), which forms robust biofilms, was grown under optimal biofilm conditions in NaCl-free Luria-Bertani broth (LB*) or in LB* supplemented with one of the non-metabolizable analogues 2-deoxy-d-glucose (2DG), methyl α-d-mannopyranoside (αMM), or methyl α-d-glucopyranoside (αMG). Biofilm growth was inhibited by mannose analogue 2DG even at very low concentration in unbuffered medium, and the maximal inhibition was enhanced in the presence of either 100 mM KPO4 or 100 mM MOPS, pH 7.5; in buffered medium, concentrations of 0.02 % (1.2 mM) or more inhibited growth nearly completely. In contrast, mannose analogue αMM, which should not be able to enter the cells but has been reported to inhibit biofilm growth by binding to FimH, did not exhibit strong inhibition even at concentrations up to 1.8 % (108 mM). The glucose analogue αMG inhibited biofilm growth, but much less strongly than did 2DG. None of the analogues inhibited planktonic growth or caused a change in pH of the unbuffered medium. Similar inhibitory effects of the analogues were observed in minimal medium. The effects were not strain-specific, as 2DG and αMG also inhibited the weak biofilm growth of E. coli K12.


Assuntos
Antimetabólitos/farmacologia , Biofilmes/crescimento & desenvolvimento , Desoxiglucose/farmacologia , Escherichia coli/crescimento & desenvolvimento , AMP Cíclico/farmacologia , Escherichia coli/efeitos dos fármacos , Glucose-6-Fosfato/farmacologia , Metilglucosídeos/farmacologia , Metilmanosídeos/farmacologia
8.
Diabetes ; 64(2): 370-82, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25157092

RESUMO

Glucose is an important stimulus for glucagon-like peptide 1 (GLP-1) secretion, but the mechanisms of secretion have not been investigated in integrated physiological models. We studied glucose-stimulated GLP-1 secretion from isolated perfused rat small intestine. Luminal glucose (5% and 20% w/v) stimulated the secretion dose dependently, but vascular glucose was without significant effect at 5, 10, 15, and 25 mmol/L. GLP-1 stimulation by luminal glucose (20%) secretion was blocked by the voltage-gated Ca channel inhibitor, nifedipine, or by hyperpolarization with diazoxide. Luminal administration (20%) of the nonmetabolizable sodium-glucose transporter 1 (SGLT1) substrate, methyl-α-D-glucopyranoside (α-MGP), stimulated release, whereas the SGLT1 inhibitor phloridzin (luminally) abolished responses to α-MGP and glucose. Furthermore, in the absence of luminal NaCl, luminal glucose (20%) did not stimulate a response. Luminal glucose-stimulated GLP-1 secretion was also sensitive to luminal GLUT2 inhibition (phloretin), but in contrast to SGLT1 inhibition, phloretin did not eliminate the response, and luminal glucose (20%) stimulated larger GLP-1 responses than luminal α-MGP in matched concentrations. Glucose transported by GLUT2 may act after metabolization, closing KATP channels similar to sulfonylureas, which also stimulated secretion. Our data indicate that SGLT1 activity is the driving force for glucose-stimulated GLP-1 secretion and that KATP-channel closure is required to stimulate a full-blown glucose-induced response.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Glucose/farmacologia , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/metabolismo , Animais , Canais de Cálcio/genética , Canais de Cálcio/metabolismo , Peptídeo 1 Semelhante ao Glucagon/genética , Ativação do Canal Iônico , Masculino , Manitol/farmacologia , Metilglucosídeos/farmacologia , Ratos , Ratos Wistar , Sódio , Transportador 1 de Glucose-Sódio/genética , Transportador 1 de Glucose-Sódio/metabolismo
9.
Am J Physiol Heart Circ Physiol ; 307(8): H1120-33, 2014 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-25128166

RESUMO

Exposure of cardiomyocytes to high glucose concentrations (HG) stimulates reactive oxygen species (ROS) production by NADPH oxidase (NOX2). NOX2 activation is triggered by enhanced glucose transport through a sodium-glucose cotransporter (SGLT) but not by a stimulation of glucose metabolism. The aim of this work was to identify potential therapeutic approaches to counteract this glucotoxicity. In cultured adult rat cardiomyocytes incubated with 21 mM glucose (HG), AMP-activated protein kinase (AMPK) activation by A769662 or phenformin nearly suppressed ROS production. Interestingly, glucagon-like peptide 1 (GLP-1), a new antidiabetic drug, concomitantly induced AMPK activation and prevented the HG-mediated ROS production (maximal effect at 100 nM). α2-AMPK, the major isoform expressed in cardiomyocytes (but not α1-AMPK), was activated in response to GLP-1. Anti-ROS properties of AMPK activators were not related to changes in glucose uptake or glycolysis. Using in situ proximity ligation assay, we demonstrated that AMPK activation prevented the HG-induced p47phox translocation to caveolae, whatever the AMPK activators used. NOX2 activation by either α-methyl-d-glucopyranoside, a glucose analog transported through SGLT, or angiotensin II was also counteracted by GLP-1. The crucial role of AMPK in limiting HG-mediated NOX2 activation was demonstrated by overexpressing a constitutively active form of α2-AMPK using adenoviral infection. This overexpression prevented NOX2 activation in response to HG, whereas GLP-1 lost its protective action in α2-AMPK-deficient mouse cardiomyocytes. Under HG, the GLP-1/AMPK pathway inhibited PKC-ß2 phosphorylation, a key element mediating p47phox translocation. In conclusion, GLP-1 induces α2-AMPK activation and blocks HG-induced p47phox translocation to the plasma membrane, thereby preventing glucotoxicity.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Peptídeo 1 Semelhante ao Glucagon/farmacologia , Glucose/farmacologia , Hipoglicemiantes/farmacologia , Miócitos Cardíacos/metabolismo , NADPH Oxidases/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Compostos de Bifenilo , Células Cultivadas , Masculino , Glicoproteínas de Membrana/metabolismo , Metilglucosídeos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , NADPH Oxidase 2 , NADPH Oxidases/genética , Fenformin/farmacologia , Proteína Quinase C/metabolismo , Transporte Proteico , Pironas/farmacologia , Ratos , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Tiofenos/farmacologia
10.
Physiol Behav ; 133: 92-8, 2014 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-24811140

RESUMO

Mice adapted to drink a flavored saccharin solution (CS-) paired with intragastric (IG) self-infusions of water rapidly increase their intake of a new flavored solution (CS+) that is paired with IG glucose self-infusions. The present study extends this method to examine post-oral glucose appetition in rats. Food-restricted rats were trained to consume a CS- flavor (e.g., grape saccharin) paired with IG water in 5 daily 1-h tests. In the next 3 tests, they drank a CS+ (e.g., cherry saccharin) paired with IG glucose. Rats infused with 8% glucose increased intake significantly on CS+ Test 1, but those infused with 16% glucose showed only a small increase in intake, which may reflect a counteracting satiating effect. Both groups further increased CS+ intakes in Tests 2 and 3, and preferred (81%) the CS+ to the CS- in a two-bottle test without infusions. A second experiment investigated rats' responses to IG alpha-methyl-d-glucopyranoside (MDG), a non-metabolizable sugar analog which stimulates CS+ intake and preference in mice. The rats reduced their intake of the MDG-paired CS+ flavor over sessions, and preferred the CS- to the CS+ in the choice test. The glucose data show that rats, like mice, rapidly detect the sugar's positive post-oral effects that can stimulate intake within the first hour of exposure. The MDG avoidance may indicate a greater sensitivity to its post-oral inhibitory effects in rats than in mice, or perhaps slower clearance of MDG in rats. The test protocol described here can be used to investigate the peripheral and central processes involved in stimulation of intake by post-oral nutrients in rats.


Assuntos
Condicionamento Clássico/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Preferências Alimentares/efeitos dos fármacos , Glucose/administração & dosagem , Edulcorantes/administração & dosagem , Administração Oral , Análise de Variância , Animais , Apetite/efeitos dos fármacos , Relação Dose-Resposta a Droga , Comportamento de Ingestão de Líquido/efeitos dos fármacos , Masculino , Metilglucosídeos/farmacologia , Ratos , Ratos Sprague-Dawley , Sacarina/administração & dosagem , Fatores de Tempo
11.
Carbohydr Res ; 362: 21-9, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23069484

RESUMO

Increased activity of the main carbohydrate pathways (glycolysis, pentose phosphate, and hexosamine biosynthetic pathways) is one of the hallmarks of metabolic diseases such as cancer. Sulfoquinovosyl diacylglycerol is a sulfoglycolipid found in the human diet that possesses anticancer activity that is absent when its carbohydrate moiety (glucose 6-sulfonate or sulfoquinovose) is removed. This work used bacterial systems to further understand the metabolism of this sugar through three main carbohydrate processing pathways and how this could influence its biological activity. Using (13)C NMR spectroscopy and enzyme assays, we showed that glucose 6-sulfonate cannot enter the pentose phosphate pathway, hence decreasing pentose and nucleotide biosyntheses. In glycolysis, glucose 6-sulfonate only provides one pyruvate per monosaccharide molecule, decreasing the flux of this pathway by half when compared to glucose 6-phosphate. Glucose 6-sulfonate can enter the hexosamine biosynthetic pathway by producing glucosamine 6-sulfonate, which is a reported antibacterial agent that competitively inhibits hexosamine production. All these interactions with carbohydrate routes might help explain the observed anticancer activity that glucose 6-sulfonate has in vitro. This adds to our knowledge of how vegetables rich in glucose 6-sulfonate can also act as metabolic inhibitors of pathways that are increased in metabolic diseases.


Assuntos
Antineoplásicos Fitogênicos/metabolismo , Escherichia coli/química , Glicólise/efeitos dos fármacos , Hexosaminas/biossíntese , Metilglucosídeos/metabolismo , Via de Pentose Fosfato/efeitos dos fármacos , Antineoplásicos Fitogênicos/farmacologia , Isótopos de Carbono , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Escherichia coli/metabolismo , Glucosamina/análogos & derivados , Glucosamina/metabolismo , Glucose-6-Fosfato/metabolismo , Humanos , Espectroscopia de Ressonância Magnética , Metilglucosídeos/farmacologia , Nucleotídeos/metabolismo , Pentoses/metabolismo , Ácido Pirúvico/metabolismo
12.
J Agric Food Chem ; 60(31): 7690-6, 2012 Aug 08.
Artigo em Inglês | MEDLINE | ID: mdl-22800339

RESUMO

4-(3,4-Dihydroxybenzoyloxymethyl)phenyl-O-ß-d-glucopyranoside (DBPG), a polyphenolic glycoside, isolated from Origanum vulgare has shown 1,1-diphenyl-2-picrylhydrazyl (DPPH(•))-scavenging capacity in previous work. This study demonstrated that DBPG exhibits antioxidant activity by a series of DPPH(•), 2,2'-azinobis(3-ethylbenzothiazoline-6-sulfonic acid) diammonium salt (ABTS(•+)), and superoxide anion radical (O(2)(•-)) radical-scavenging assays. The inhibition of lipid peroxidation (LP) by DBPG exceeded that by l-ascorbic acid (AA) in a liposome model system. Adding DBPG to mouse liver and brain tissue inhibited the formation of thiobarbituric acid reactive substances (TBARS) to a greater extent than did trolox. In the oxygen stress test, BNLCL2 and HaCaT cells pretreated with DBPG showed increased activities of glutathione peroxidase (GPx), perhaps as a result of reduction of the production of reactive oxygen species (ROS). These findings proved that DBPG had antioxidant activities and a cytoprotective effect in hepatocytes and keratinocytes, suggesting that DBPG may be a useful food and cosmetic additive.


Assuntos
Sequestradores de Radicais Livres/farmacologia , Metilglucosídeos/farmacologia , Origanum/química , Estresse Oxidativo/efeitos dos fármacos , Extratos Vegetais/farmacologia , Animais , Linhagem Celular , Sequestradores de Radicais Livres/análise , Humanos , Metilglucosídeos/análise , Camundongos , Extratos Vegetais/análise
13.
Biochim Biophys Acta ; 1823(4): 971-82, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22230192

RESUMO

This study demonstrated that exchange proteins directly activated by cAMP (Epac) and protein kinase A (PKA) by 8-bromo (8-Br)-adenosine 3',5'-cyclic monophosphate (cAMP) stimulated [(14)C]-α-methyl-D-glucopyranoside (α-MG) uptake through increased sodium-glucose cotransporters (SGLTs) expression and translocation to lipid rafts in renal proximal tubule cells (PTCs). In PTCs, SGLTs were colocalized with lipid raft caveolin-1 (cav-1), disrupted by methyl-ß-cyclodextrin (MßCD). Selective activators of Epac or PKA, 8-Br-cAMP, and forskolin stimulated expressions of SGLTs and α-MG uptake in PTCs. In addition, 8-Br-cAMP-induced PKA and Epac activation increased phosphorylation of extracellular signal-regulated kinase (ERK), p38 mitogen-activated protein kinase (MAPK), and nuclear factor kappa B (NF-κB), which were involved in expressions of SGLTs. Furthermore, 8-Br-cAMP stimulated SGLTs translocation to lipid rafts via filamentous actin (F-actin) organization, which was blocked by cytochalasin D. In addition, cav-1 and SGLTs stimulated by 8-Br-cAMP were detected in lipid rafts, which were blocked by cytochalasin D. Furthermore, 8-Br-cAMP-induced SGLTs translocation and α-MG uptake were attenuated by inhibition of cav-1 activation with cav-1 small interfering RNA (siRNA) and inhibition of F-actin organization with TRIO and F-actin binding protein (TRIOBP). In conclusion, 8-Br-cAMP stimulated α-MG uptake via Epac and PKA-dependent SGLTs expression and trafficking through cav-1 and F-actin in PTCs.


Assuntos
Actinas/metabolismo , Caveolina 1/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Túbulos Renais Proximais/citologia , Túbulos Renais Proximais/metabolismo , Proteínas de Transporte de Sódio-Glucose/metabolismo , 8-Bromo Monofosfato de Adenosina Cíclica/farmacologia , Animais , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/enzimologia , Masculino , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Metilglucosídeos/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Transporte Proteico/efeitos dos fármacos , Coelhos
14.
Exp Dermatol ; 20(9): 743-8, 2011 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-21672031

RESUMO

Ov-16 (4-(3,4-dihydroxybenzoyloxymethyl)phenyl-O-ß-D-glucopyranoside), a polyphenolic glycoside that is isolated from oregano (Origanum vulgare L.), can scavenge 1,1-diphenyl-2-picrylhydrazyl (DPPH) free radicals. This investigation is the first to study in detail the hypopigmentary properties of Ov-16. It demonstrates that 0-1000 µg/ml Ov-16 inhibits the activity of mushroom tyrosinase (Tyr) in a concentration-dependent manner. The inhibitionary Tyr kinetics of Ov-16 towards the oxidation of L-DOPA was found to be uncompetitive. Following the treatment of human skin premalignant kerationcyte HaCaT cells, human skin fibroblast Hs68 cells and mice melanoma B16 cells with Ov-16 (0-100 µg/ml), cell viability was >98%, suggesting that Ov-16 is non-toxic. Ov-16 can reduce cellular Tyr activity, DOPA oxidase activity and melanin synthesis in B16 cells that are stimulated by the α-melanocyte-stimulating hormone (α-MSH). Moreover, Ov-16 inhibited the production of melanin in Streptomyces bikiniensis without affecting the growth of the microorganism. The treatment of B16 cells with Ov-16 considerably reduced the gene expressions of melanocortin-1 receptor (Mc1r), microphthalmia-associated transcription factor (Mitf), Tyr, tyrosinase-related proteins-2 (Trp-2) and Trp-1, as determined by RT-PCR. The expressions of Mc1r, Mift, Tyr, Trp-2 and TrpP-1 protein in Ov-16-treated B16 cells were also significantly reduced, as determined by western blotting and fluorescent staining analysis. These results suggest that Ov-16 exhibits hypopigmentary performance.


Assuntos
Melaninas/antagonistas & inibidores , Melaninas/biossíntese , Metilglucosídeos/farmacologia , Agaricales/enzimologia , Animais , Antioxidantes/farmacologia , Sequência de Bases , Linhagem Celular , Primers do DNA/genética , Medicamentos de Ervas Chinesas/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Oxirredutases Intramoleculares/genética , Melanoma Experimental/tratamento farmacológico , Melanoma Experimental/genética , Melanoma Experimental/metabolismo , Glicoproteínas de Membrana/genética , Camundongos , Fator de Transcrição Associado à Microftalmia/genética , Monofenol Mono-Oxigenase/antagonistas & inibidores , Monofenol Mono-Oxigenase/genética , Oxirredutases/genética , Receptor Tipo 1 de Melanocortina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Pigmentação da Pele/efeitos dos fármacos , Pigmentação da Pele/genética , Pigmentação da Pele/fisiologia , Streptomyces/efeitos dos fármacos , Streptomyces/metabolismo
15.
J Biosci Bioeng ; 112(2): 194-201, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21596618

RESUMO

We studied the effects of d-glucose on transgene expression in mammalian cells by a reporter gene assay using CV-1 cells and a CMV promoter-controlled EGFP gene. Treatment of CV-1 cells with 5% D-glucose unchanged the number of fluorescent cells in fluorescence microscopic observation but significantly intensified fluorescence in the fluorometric assay. Furthermore, EGFP itself and mRNA became more abundant in Western blot and quantitative RT-PCR analyses of 5% D-glucose-treated cells, respectively. These results indicate that elevated D-glucose can activate transgene expression via transcriptional stimulation, at least in part. The same concentrations of L-glucose led to only negligible increases in transgene expression, indicating that D-glucose's effect is different from its osmotic effect. The D-glucose-induced augmentation of fluorescence was observed not only in the experiment using the CMV promoter-controlled EGFP gene but also in experiments using the SV40 and RSV promoter-controlled ones, suggesting that elevated D-glucose can enhance transgene expression regulated by various promoters commonly used in transgene expression. The assessment of D-glucose analogs for their enhancive effects on transgene expression revealed that 1,6-anhydro-D-glucose and ß-methyl-D-glucoside had stronger effects than D-glucose. From this result, we can expect to find more effective carbohydrates to enhance transgene expression. The α- and ß-M-D-glucosides, which are slightly different from each other in three-dimensional structure, exerted largely distinct stimulative effects on transgene expression, suggesting that fundamental rules determine the enhancive effects of saccharides and that the modification of the saccharide by applying such rules will enable us to develop more powerful substances for transgene expression.


Assuntos
Glucose/farmacologia , Transgenes , Animais , Linhagem Celular , Corantes Fluorescentes/análise , Expressão Gênica/efeitos dos fármacos , Genes Reporter , Vetores Genéticos , Glucose/análogos & derivados , Proteínas de Fluorescência Verde/análise , Proteínas de Fluorescência Verde/genética , Metilglucosídeos/farmacologia , Regiões Promotoras Genéticas , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Vírus 40 dos Símios/genética , Transfecção
16.
Diabetes Technol Ther ; 13(7): 743-51, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21510766

RESUMO

BACKGROUND: Diabetes may alter renal glucose reabsorption by sodium (Na(+))-dependent glucose transporters (SGLTs). Radiolabeled substrates are commonly used for in vitro measurements of SGLT activity in kidney cells. We optimized a method to measure glucose uptake using a fluorescent substrate, 2-(N-(7-nitrobenz-2-oxa-1,3-diazol-4-yl)amino)-2-deoxyglucose (2-NBDG). METHODS: Uptake buffers for 2-NBDG were the same as for (14)C-labeled α-methyl-d-glucopyranoside ([(14)C]AMG). Cell lysis buffer was optimized for fluorescence of 2-NBDG and Hoechst DNA stain. Uptake was performed on cultures of primary mouse kidney cells (PMKCs), the LLC-PK(1) proximal tubule cell line, or COS-7 cells transiently overexpressing mouse SGLT1 or SGLT2 by incubating cells at 37°C in buffer containing 50-200 µM 2-NBDG. Microscopy was performed to visualize uptake in intact cells, while a fluorescence microplate reader was used to measure intracellular concentration of 2-NBDG ([2-NBDG](i)) in cell homogenates. RESULTS: Fluorescent cells were observed in cultures of PMKCs and LLC-PK(1) cells exposed to 2-NBDG in the presence or absence of Na(+). In LLC-PK(1) cells, 2-NBDG transport in the presence of Na(+) had a maximum rate of 0.05 nmol/min/µg of DNA. In these cells, Na(+)-independent uptake of 2-NBDG was blocked with the GLUT inhibitor, cytochalasin B. The Na(+)-dependent uptake of 2-NBDG decreased in response to co-exposure to the SGLT substrate, AMG, and it could be blocked with the SGLT inhibitor, phlorizin. Immunocytochemistry showed overexpression of SGLT1 and SGLT2 in COS-7 cells, in which, in the presence of Na(+), [2-NBDG](i) was fivefold higher than in controls. CONCLUSION: Glucose transport in cultured kidney cells can be measured with the fluorescence method described in this study.


Assuntos
4-Cloro-7-nitrobenzofurazano/análogos & derivados , Desoxiglucose/análogos & derivados , Corantes Fluorescentes/farmacocinética , Glucose/metabolismo , Rim/metabolismo , 4-Cloro-7-nitrobenzofurazano/farmacocinética , Animais , Transporte Biológico/efeitos dos fármacos , Células COS , Linhagem Celular , Células Cultivadas , Chlorocebus aethiops , Citocalasina B/farmacologia , Desoxiglucose/farmacocinética , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Humanos , Rim/citologia , Rim/efeitos dos fármacos , Rim/ultraestrutura , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/ultraestrutura , Metilglucosídeos/farmacologia , Camundongos , Microscopia de Fluorescência , Florizina/farmacologia , Sódio/metabolismo , Transportador 1 de Glucose-Sódio/antagonistas & inibidores , Transportador 1 de Glucose-Sódio/genética , Transportador 1 de Glucose-Sódio/metabolismo , Transportador 2 de Glucose-Sódio/genética , Transportador 2 de Glucose-Sódio/metabolismo , Inibidores do Transportador 2 de Sódio-Glicose , Espectrometria de Fluorescência
17.
J Appl Microbiol ; 108(6): 2152-61, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-19961546

RESUMO

AIMS: This study investigates the antimicrobial activity and mode of action of novel carbohydrate fatty acid (CFA) derivatives against Staphylococcus aureus and methicillin-resistant Staph. aureus (MRSA). METHODS AND RESULTS: Minimum inhibitory concentrations (MICs) and the effect of CFA derivatives on lag phase were determined using a broth microdilution method. Lauric acid carbohydrate esters and corresponding ether analogues showed the greatest antimicrobial activity with MIC values between 0.04 and 0.16 mmol l(-1). Leakage studies at 260 nm following exposure to CFA derivatives at 4x MIC showed a significant increase in membrane permeability for all compounds, after c. 15 min exposure except for the lauric beta ether CFA derivative. Further assessment using both BacLight and luminescence ATP assays confirmed that an increase in membrane permeability and reduced metabolic activity was associated with CFA treatment. CONCLUSIONS: All strains were significantly inhibited by the novel compounds studied, and efficacy was related to specific structural features. Cell-membrane permeabilization was associated with CFA treatment and may account for at least a component of the mode of action of these compounds. SIGNIFICANCE AND IMPACT OF THE STUDY: This study reports the antimicrobial action of CFA compounds against a range of Staph. aureus and MRSA strains, and provides insights into their mode of action.


Assuntos
Antibacterianos/farmacologia , Ésteres/farmacologia , Ácidos Láuricos/farmacologia , Staphylococcus aureus Resistente à Meticilina/efeitos dos fármacos , Metilglucosídeos/farmacologia , Permeabilidade da Membrana Celular/efeitos dos fármacos , Staphylococcus aureus Resistente à Meticilina/crescimento & desenvolvimento , Testes de Sensibilidade Microbiana
18.
J Membr Biol ; 223(2): 87-106, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18592293

RESUMO

Drugs are transported by cotransporters with widely different turnover rates. We have examined the underlying mechanism using, as a model system, glucose and indican (indoxyl-beta-D-glucopyranoside) transport by human Na+/glucose cotransporter (hSGLT1). Indican is transported by hSGLT1 at 10% of the rate for glucose but with a fivefold higher apparent affinity. We expressed wild-type hSGLT1 and mutant G507C in Xenopus oocytes and used electrical and optical methods to measure the kinetics of glucose (using nonmetabolized glucose analogue alpha-methyl-D-glucopyranoside, alphaMDG) and indican transport, alone and together. Indican behaved as a competitive inhibitor of alphaMDG transport. To examine protein conformations, we recorded SGLT1 capacitive currents (charge movements) and fluorescence changes in response to step jumps in membrane voltage, in the presence and absence of indican and/or alphaMDG. In the absence of sugar, voltage jumps elicited capacitive SGLT currents that decayed to steady state with time constants (tau) of 3-20 ms. These transient currents were abolished in saturating alphaMDG but only slightly reduced (10%) in saturating indican. SGLT1 G507C rhodamine fluorescence intensity increased with depolarizing and decreased with hyperpolarizing voltages. Maximal fluorescence increased approximately 150% in saturating indican but decreased approximately 50% in saturating alphaMDG. Modeling indicated that the rate-limiting step for indican transport is sugar translocation, whereas for alphaMDG it is dissociation of Na+ from the internal binding sites. The inhibitory effects of indican on alphaMDG transport are due to its higher affinity and a 100-fold lower translocation rate. Our results indicate that competition between substrates and drugs should be taken into consideration when targeting transporters as drug delivery systems.


Assuntos
Transporte Biológico Ativo/fisiologia , Glucose/metabolismo , Glucosídeos/metabolismo , Indicã/análogos & derivados , Transportador 1 de Glucose-Sódio/fisiologia , Animais , Fluorescência , Glucosídeos/farmacologia , Humanos , Indicã/metabolismo , Indicã/farmacologia , Cinética , Metilglucosídeos/metabolismo , Metilglucosídeos/farmacologia , Modelos Biológicos , Oócitos , Conformação Proteica , Xenopus laevis
19.
Diabetes ; 57(10): 2569-76, 2008 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-18591392

RESUMO

OBJECTIVE: Glucose sensing by specialized neurons of the hypothalamus is vital for normal energy balance. In many glucose-activated neurons, glucose metabolism is considered a critical step in glucose sensing, but whether glucose-inhibited neurons follow the same strategy is unclear. Orexin/hypocretin neurons of the lateral hypothalamus are widely projecting glucose-inhibited cells essential for normal cognitive arousal and feeding behavior. Here, we used different sugars, energy metabolites, and pharmacological tools to explore the glucose-sensing strategy of orexin cells. RESEARCH DESIGN AND METHODS: We carried out patch-clamp recordings of the electrical activity of individual orexin neurons unambiguously identified by transgenic expression of green fluorescent protein in mouse brain slices. RESULTS- We show that 1) 2-deoxyglucose, a nonmetabolizable glucose analog, mimics the effects of glucose; 2) increasing intracellular energy fuel production with lactate does not reproduce glucose responses; 3) orexin cell glucose sensing is unaffected by glucokinase inhibitors alloxan, d-glucosamine, and N-acetyl-d-glucosamine; and 4) orexin glucosensors detect mannose, d-glucose, and 2-deoxyglucose but not galactose, l-glucose, alpha-methyl-d-glucoside, or fructose. CONCLUSIONS: Our new data suggest that behaviorally critical neurocircuits of the lateral hypothalamus contain glucose detectors that exhibit novel sugar selectivity and can operate independently of glucose metabolism.


Assuntos
Carboidratos/farmacologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neurônios/efeitos dos fármacos , Neuropeptídeos/metabolismo , Acetilglucosamina/farmacologia , Aloxano/farmacologia , Animais , Desoxiglucose/farmacologia , Metabolismo Energético/efeitos dos fármacos , Frutose/farmacologia , Galactose/farmacologia , Glucosamina/farmacologia , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Região Hipotalâmica Lateral/citologia , Região Hipotalâmica Lateral/efeitos dos fármacos , Região Hipotalâmica Lateral/metabolismo , Ácido Láctico/farmacologia , Manose/farmacologia , Potenciais da Membrana/efeitos dos fármacos , Metilglucosídeos/farmacologia , Camundongos , Camundongos Transgênicos , Neurônios/citologia , Neurônios/metabolismo , Orexinas , Técnicas de Patch-Clamp , Tolbutamida/farmacologia
20.
J Ind Microbiol Biotechnol ; 35(7): 695-701, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18317827

RESUMO

In this study, we utilized a catabolite repressor to improve the enzymatic activity of recombinant beta-galactosidase inclusion bodies (IBs) produced in Escherichia coli under the araBAD promoter system. Specifically, we employed methyl alpha-D: -glucopyranoside (alpha-MG) to lower the transcription rate of the beta-galactosidase structural gene. In deepwell microtiter plate and lab-scale fermentor culture systems, we demonstrated that the addition of alpha-MG after induction improved the specific beta-galactosidase production, even though beta-galactosidase was still produced as an IB. Particularly, the addition of 0.0025% alpha-MG led to the most significant increase in the specific activity of the beta-galactosidase. Interestingly, the beta-galactosidase IBs obtained in the presence of 0.0025% alpha-MG were more loosely packed, as determined by IB solubilization in guanidine hydrochloride solution. We propose that the reduced gene transcription rate was responsible for the increased specific beta-galactosidase activity and the loose packing that characterized the IBs produced in the presence of alpha-MG. This principle could be applied throughout the enzyme bioprocessing industry in order to enhance the activity of aggregate-prone enzymes within IBs.


Assuntos
Ativadores de Enzimas/farmacologia , Escherichia coli/metabolismo , Corpos de Inclusão/enzimologia , Metilglucosídeos/farmacologia , beta-Galactosidase/metabolismo , Regulação para Baixo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Expressão Gênica/efeitos dos fármacos , Regiões Promotoras Genéticas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , beta-Galactosidase/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...