Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
J Physiol Pharmacol ; 74(4)2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37865959

RESUMO

Overweight and obesity are associated with severe metabolic disorders and an increased risk of cardiovascular diseases. It is a known fact that physical activity has a positive effect on metabolic parameters, and also reduces the risk of diseases such as diabetes. Some products can enhance the rate of lipolysis and help in improving fat loss. One of these are selective androgen receptor modulators (SARMs) which act as anabolic agents and are also believed to aid in fat-burning. In this study, we investigated whether 30 days of ostarine administration could potentially improve metabolic parameters using the rat model of obesity combined with exercise. We assessed the levels of biochemical and hormonal parameters in serum samples as well as insulin sensitivity indices of tissues. There were significant changes in the metabolic parameters with exercise. However, we did not find any additive effects of ostarine and exercise on most of the parameters tested. Similar results were obtained from the analysis of gene expression and the concentration of leptin and adiponectin. Our results indicated that ostarine had a lowering effect on cholesterol concentration in the serum (P<0.05). Moreover, when combining ostarine and exercise, additive changes were only observed in the levels of total and HDL cholesterol. No significant change was observed in the metabolic parameters of obese rats with the use of ostarine at the dose of 0.4 mg/kg body weight. Since ostarine is known to enhance performance, further research on its effects is needed.


Assuntos
Leptina , Obesidade , Ratos , Animais , Obesidade/metabolismo , Anilidas/farmacologia , Sobrepeso , Adiponectina
2.
Mol Cell Endocrinol ; 562: 111850, 2023 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-36623583

RESUMO

Neuropeptide B (NPB) modulates energy homeostasis and metabolism through activation of NPBWR1 and NPBWR2 in humans and NPBWR1 in rodents. Recently, we reported that NPB promotes adipogenesis in rat brown preadipocytes. In the present study, we evaluated the effects of NPB on proliferation and differentiation into mature adipocytes of white rat preadipocytes and 3T3-L1 cells. We found the expression of NPBWR1 and NPB on mRNA and protein level in rat white preadipocytes and 3T3-L1 cells. NPB increased expression of mRNA and protein production of adipogenic genes (PPARγ, C/EBPß, CEBPα and FABP4) in rat preadipocytes and 3T3-L1 cells during the differentiation process. Furthermore, NPB stimulated lipid accumulation in rat preadipocytes and 3T3-L1 cells. In addition, we found that NPB promotes phosphorylation of p38 kinase in rat preadipocytes and 3T3-L1 cells. NPB failed to stimulate expression of proadipogenic genes in the presence of p38 inhibitor. NPB failed to modulate viability and proliferation of rat preadipocytes and 3T3-L1 cells. Taken together, we report that NPB promotes differentiation of rodent preadipocytes via p38-dependent mechanism. NPB does not modulate viability and proliferation of rat preadipocytes and 3T3-L1 cells.


Assuntos
Adipócitos , Animais , Camundongos , Ratos , Células 3T3-L1 , Adipócitos/metabolismo , Adipogenia/genética , Diferenciação Celular , PPAR gama/metabolismo , RNA Mensageiro/metabolismo
3.
J Physiol Pharmacol ; 74(6)2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38345440

RESUMO

Orexins A (OXA) and B (OXB) (hypocretin 1 and 2) are neuropeptides produced in the brain and peripheral tissues. Biological activities of orexins are mediated through activation of two G-protein coupled receptors termed as orexin 1 receptor (OX1R) and orexin 2 receptor (OX1R). Orexin system (OXA, OXB, OX1R, OX2R) was implicated in controlling sleep, energy expenditure, appetite, reproduction as well as metabolism and energy homeostasis. In this review, we summarize the current knowledge regarding the role of the orexin system in controlling porcine physiology. Particularly, we review and discuss evidence indicating that in pig and other living organisms, orexins and their receptors modulate the energy homeostasis, reproduction as well as functions of peripheral tissues including the pancreas, adrenal glands, gastro-intestinal tract and adipose tissue.


Assuntos
Receptores Acoplados a Proteínas G , Reprodução , Animais , Suínos , Orexinas/metabolismo , Receptores de Orexina/metabolismo , Homeostase , Sistema Endócrino/metabolismo , Receptores de Neuropeptídeos
4.
Eur Rev Med Pharmacol Sci ; 25(19): 5993-6002, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34661259

RESUMO

OBJECTIVE: Kisspeptin (KP) is a major regulator of reproductive functions. It has also been shown to be involved in the metabolic changes associated with obesity. According to the well-established concept of prenatal programming, environmental factors can influence physiological and behavioral systems at the early stages of development. Thus, we hypothesized that in pregnant women, obesity can be associated with alterations in the levels of KP. We also assumed that the observed changes in obese mothers' blood (MB) would be reflected in the umbilical cord blood (CB). MATERIALS AND METHODS: We collected MB and CB from obese and nonobese women and analyzed the differences in metabolic and hormonal profiles, including KP concentration, using commercially available assays. RESULTS: We found that the level of KP was increased in the MB and CB of obese patients compared to nonobese subjects (p<0.05). A strong correlation was observed between the concentration of KP in MB and CB (r=0.8343; p<0.01). Moreover, we detected that the differences in the adipokine profile observed in the MB were not reflected in CB. CONCLUSIONS: Our results indicate that blood KP concentration can serve as a valuable marker in pregnant women. However, further studies are needed to understand the alterations of this peptide in obese pregnant woman and their potential effects on offspring.


Assuntos
Sangue Fetal/metabolismo , Kisspeptinas/sangue , Obesidade/epidemiologia , Adulto , Feminino , Humanos , Recém-Nascido , Masculino , Mães , Obesidade/sangue , Projetos Piloto , Gravidez
5.
J Physiol Pharmacol ; 71(1)2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32554844

RESUMO

Adropin is a peptide hormone which is produced in brain and peripheral tissues such as liver. It was found that adropin modulates lipid and glucose homeostasis by interacting with hepatocytes and myocytes. Adropin enhances insulin sensitivity and alleviates hyperinsulinemia in animal models with high-fat diet-induced insulin resistance. However, it is unknown whether adropin regulates insulin secretion and proliferation of beta cells. Therefore, we studied the effects of adropin on insulin secretion in INS-1E cells as well as isolated pancreatic islets. Furthermore, we assessed the influence of adropin on insulin mRNA expression, cell viability and proliferation in INS-1E cells. Pancreatic islets were isolated from male Wistar rats. mRNA expression was evaluated using real-time PCR and cell viability by MTT assay. Cell replication was measured by BrdU incorporation and insulin secretion by RIA. We found that adropin suppresses insulin mRNA expression in INS-1E cells. Moreover, adropin attenuates glucose-induced insulin secretion in INS-1E cells as well as in isolated pancreatic islets. In addition, using INS-1E cells we found that adropin suppresses glucose-induced cAMP production. However, adropin fails to modulate INS-1E cell viability and proliferation. In summary, we found adropin suppresses insulin mRNA expression and secretion, without affecting beta cell viability or proliferation.


Assuntos
Proteínas Sanguíneas/farmacologia , Antagonistas da Insulina/farmacologia , Secreção de Insulina/fisiologia , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Peptídeos/farmacologia , Animais , Linhagem Celular , Relação Dose-Resposta a Droga , Humanos , Secreção de Insulina/efeitos dos fármacos , Ilhotas Pancreáticas/efeitos dos fármacos , Masculino , Camundongos , Ratos , Ratos Wistar
6.
J Endocrinol ; 238(2): R95-R108, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-29848609

RESUMO

Orexin A and B are two neuropeptides, which regulate a variety of physiological functions by interacting with central nervous system and peripheral tissues. Biological effects of orexins are mediated through two G-protein-coupled receptors (OXR1 and OXR2). In addition to their strong influence on the sleep-wake cycle, there is growing evidence that orexins regulate body weight, glucose homeostasis and insulin sensitivity. Furthermore, orexins promote energy expenditure and protect against obesity by interacting with brown adipocytes. Fat tissue and the endocrine pancreas play pivotal roles in maintaining energy homeostasis. Since both organs are crucially important in the context of pathophysiology of obesity and diabetes, we summarize the current knowledge regarding the role of orexins and their receptors in controlling adipocytes as well as the endocrine pancreatic functions. Particularly, we discuss studies evaluating the effects of orexins in controlling brown and white adipocytes as well as pancreatic alpha and beta cell functions.


Assuntos
Tecido Adiposo/fisiologia , Ilhotas Pancreáticas/fisiologia , Orexinas/fisiologia , Adipócitos/metabolismo , Tecido Adiposo/metabolismo , Animais , Peso Corporal/genética , Metabolismo Energético/genética , Humanos , Obesidade/genética , Obesidade/metabolismo , Pâncreas/metabolismo , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/genética
7.
Domest Anim Endocrinol ; 63: 59-68, 2018 04.
Artigo em Inglês | MEDLINE | ID: mdl-29413903

RESUMO

It is well known that orexins are involved in the metabolism and endocrine function of rodent adipocytes, but there are no data on other animal species, including pigs. Therefore, in this study, we tested the hypothesis that orexin A (OxA) and orexin B (OxB) modulate the metabolism and endocrine functions of isolated porcine adipocytes and adipose tissue explants. Moreover, we characterized the possible mechanism of OxA action in porcine adipocytes. According to the results, both orexin receptor 1 and orexin receptor 2 were expressed in the porcine adipose tissue. We found that OxA suppressed the release of glycerol from porcine adipocytes both in the absence (basal lipolysis; P < 0.05) and in the presence (stimulated lipolysis; P < 0.05) of isoproterenol. Orexin A increased basal and insulin-stimulated glucose uptake (P < 0.05), as well as it enhanced the rate of glucose incorporation into lipids with insulin (stimulated lipogenesis; P < 0.01) or without insulin (basal; P < 0.05). We have also shown that OxA stimulated the mRNA expression of glucose transporter 4 (P < 0.05) and its translocation into the plasma membrane (P < 0.01). Moreover, OxA upregulated the mRNA expression of leptin in isolated porcine adipocytes (P < 0.05) and increased the secretion of leptin (P < 0.05). We have also demonstrated one of the possible mechanisms of OxA action in adipocytes. In the presence of extracellular-signal-regulated kinase 1 and 2 (ERK1/2) inhibitor, the effect of OxA was not detectable in porcine adipocytes, which indicates that this peptide increased cell viability via ERK1/2 pathway (P < 0.05). However, OxB did not show any effect on the metabolism and endocrine functions of porcine adipocytes. In summary, we have shown for the first time that OxA has a significant impact on the intensity of lipolysis, glucose uptake, lipogenesis, as well as on the expression and secretion of leptin. Therefore, we conclude that OxA but not OxB regulates lipid metabolism in porcine adipose tissue and that this regulation is partly mediated via ERK1/2 pathway. The action of orexins should be further explored to better understand their role in the regulation of adiposity in pigs.


Assuntos
Adipócitos/efeitos dos fármacos , Leptina/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Orexinas/farmacologia , Adipócitos/metabolismo , Animais , Transporte Biológico , Sobrevivência Celular , Células Cultivadas , Glucose/metabolismo , Lipogênese/efeitos dos fármacos , Masculino , Suínos
8.
Physiol Res ; 67(1): 45-56, 2018 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-29137471

RESUMO

Spexin (SPX) and kisspeptin (KISS) are novel peptides relevant in the context of regulation of metabolism, food intake, puberty and reproduction. Here, we studied changes of serum SPX and KISS levels in female non-obese volunteers (BMI<25 kg/m(2)) and obese patients (BMI>35 kg/m(2)). Correlations between SPX or KISS with BMI, McAuley index, QUICKI, HOMA IR, serum levels of insulin, glucagon, leptin, adiponectin, orexin-A, obestatin, ghrelin and GLP-1 were assessed. Obese patients had lower SPX and KISS levels as compared to non-obese volunteers (SPX: 4.48+/-0.19 ng/ml vs. 6.63+/-0.29 ng/ml; p<0.001, KISS: 1.357+/-0.15 nmol/l vs. 2.165+/-0.174 nmol/l; p<0.01). SPX negatively correlated with BMI, HOMA-IR, insulin, glucagon, active ghrelin and leptin. Positive correlations were found between SPX and QUICKI index, McAuley index, serum levels of obestatin, GLP-1 and adiponectin and orexin-A Serum KISS negatively correlated with BMI, HOMA-IR, serum levels of insulin, glucagon, active ghrelin and leptin. KISS positively correlated with QUICKI index, McAuley index and adiponectin. In summary, SPX and KISS show negative correlations with obesity, insulin resistance indices, and hormones known to affect insulin sensitivity in females. Both, SPX and KISS could be therefore relevant in the pathophysiology of obesity and insulin resistance.


Assuntos
Resistência à Insulina/fisiologia , Kisspeptinas/sangue , Obesidade/sangue , Hormônios Peptídicos/sangue , Adulto , Biomarcadores/sangue , Feminino , Humanos , Pessoa de Meia-Idade , Obesidade/diagnóstico
9.
Cell Signal ; 35: 242-249, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28359774

RESUMO

TRPV4 is a Ca2+-permeable, nonselective cation channel. Recently, TRPV4 was implicated in controlling peripheral insulin sensitivity, insulin secretion and apoptosis of pancreatic beta cells. Here, we characterize the role and potential mechanisms of TRPV4 in regulating insulin mRNA expression and cell death in insulin producing INS-1E cells and rat pancreatic islets. TRPV4 protein production was downregulated by siRNA. Intracellular calcium level was measured using Fluo-3 AM. Gene expression was studied by real-time PCR. Phosphorylation of extracellular signal-regulated kinase (ERK1 and ERK2) was detected by Western blot. Nitric oxide (NO) production was assessed by chemiluminescent reaction. Reactive oxygen species (ROS) level was analysed using a fluorogenic dye (DCFDA). Cell death was evaluated by determination of cytoplasmic histone-associated DNA fragments. Downregulation of TRPV4 neither affected insulin mRNA expression nor INS-1E cell growth. By contrast, pharmacological TRPV4 activation by 100nmol/l GSK1016790A increased Ca2+ levels in INS-1E cells and enhanced insulin mRNA expression after 1 and 3h, whereas a suppression of insulin mRNA expression was detected after 24h incubation. GSK1016790A increased ERK1/2 phosphorylation and NO production but not ROS production. Pharmacological blockade of ERK1/2 attenuated GSK1016790A-induced insulin mRNA expression. Inhibition of NO synthesis by l-NAME failed to affect insulin mRNA expression in GSK1016790A treated INS-1E cells. Furthermore, inhibition of NO production attenuated GSK1016790A-induced INS-1E cell death. In pancreatic islets, 100nmol/l GSK1016790A increased insulin mRNA levels after 3h without inducing cytotoxicity after 24h. In conclusion, TRPV4 differently regulates insulin mRNA expression in INS-1E cells via ERK1/2 and NO-dependent mechanisms.


Assuntos
Cálcio/metabolismo , Insulina/genética , Óxido Nítrico/metabolismo , Canais de Cátion TRPV/genética , Animais , Proliferação de Células/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Insulina/metabolismo , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/patologia , Leucina/administração & dosagem , Leucina/análogos & derivados , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosforilação , RNA Mensageiro/genética , Ratos , Espécies Reativas de Oxigênio/metabolismo , Sulfonamidas/administração & dosagem , Canais de Cátion TRPV/metabolismo
10.
J Physiol Pharmacol ; 68(5): 669-681, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29375041

RESUMO

Orexin regulates food intake and energy expenditure. Here, we test the ability of orexin-A (OXA, hypocretin-1) at improving metabolic control in type 2 diabetic animals and elaborate potential mechanisms of action. Rats with experimentally induced type 2 diabetes by a combination of streptozotocin injection and high-fat diet feeding were chronically infused with OXA. In vitro experiments were conducted on isolated pancreatic islets, primary adipocytes and insulin secreting INS-1E cells. OXA improved glucose control, enhanced insulin sensitivity and attenuated pancreatic ß-cell loss in type 2 diabetic rats. Ex vivo, apoptotic death of pancreatic islets isolated from OXA-treated type 2 diabetic animals as well as the impairment of glucose-stimulated insulin secretion were attenuated, as compared to islets derived from vehicle-treated rats. OXA reduced plasma tumor necrosis factor-α (TNF-α) and non-esterified fatty acids (NEFA) levels in type 2 diabetic rats. OXA decreased palmitate- and TNF-α-induced apoptosis of INS-1E cells. OXA improves glucose control by enhancing insulin sensitivity and protecting ß-cells from apoptotic cell death in type 2 diabetic animals.


Assuntos
Glicemia/efeitos dos fármacos , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Células Secretoras de Insulina/efeitos dos fármacos , Orexinas/uso terapêutico , Animais , Glicemia/metabolismo , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Tipo 2/sangue , Células Secretoras de Insulina/metabolismo , Masculino , Orexinas/farmacologia , Ratos , Resultado do Tratamento
11.
J Physiol Pharmacol ; 68(6): 815-821, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29550793

RESUMO

The physiology of porcine pancreatic islets is poorly understood. Orexin A is one of important agents regulating the physiology of porcine pancreatic islets. This study aimed to determine the potential effect of orexin A on the functioning of porcine pancreatic islets. Orexin receptor localization was done by PCR (polymerase chain reaction) and Western Blot, both in pancreatic isolated islets and whole pancreas. Secretion of insulin and glucagon from islets after orexin-A treatment was assayed. The viability of pig pancreatic islet cells and level of cleaved/total caspase 3 protein were measured by MTT test (3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide) and Western blotting, respectively. Orexin receptors were detected in pancreatic isolated islets, and orexin-A stimulated insulin secretion and decreased glucagon secretion from isolated porcine islets. Moreover, we detected a protective effect of orexin A on pancreatic islet cells, which manifested as higher cell viability and lower caspase 3 activation. These findings generate a better understanding of pancreatic cells functions and perhaps provide a novel tool to prevent or alleviate negative consequences of disorders in pancreatic islets.


Assuntos
Ilhotas Pancreáticas/metabolismo , Orexinas/metabolismo , Animais , Caspase 3/metabolismo , Sobrevivência Celular , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Glucagon/metabolismo , Insulina/metabolismo , Secreção de Insulina , Receptores de Orexina/genética , Receptores de Orexina/metabolismo , Suínos
12.
J Physiol Pharmacol ; 67(5): 643-652, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28011945

RESUMO

Orexins A (OXA) and B (OXB) control energy homeostasis by regulating food intake, energy expenditure and sleep-wake cycle. Several studies showed that OXA stimulates insulin secretion and proliferation of beta cells. However, mechanisms of action are still not well understood. Here, we investigated whether ERK and transient receptor potential channels (TRPs) play a role in mediating the effect of OXA on cell growth, insulin production, and secretion using the established INS-1E cell line. Cell proliferation was measured using BrdU assay. Insulin mRNA expression was detected by real-time PCR. Insulin secretion was assessed using ELISA. Intracellular calcium levels were measured using fluorescence calcium imaging (fura-2/AM). Extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation was detected by Western blot. TRP channel activity was blocked by lanthanum (III) chloride (La3+; 100 - 300 µM) or ruthenium red (RuR; 10 µM). OXA (100 nM) stimulated INS-1E cell proliferation, insulin secretion, intracellular Ca2+ concentration and ERK1/2 phosphorylation, without changing insulin mRNA expression. Inhibition of ERK1/2 by 10 µM U0126 attenuated OXA-stimulated INS-1E cell proliferation. Blockade of TRP channel activity by La3+ or RuR rendered OXA ineffective at modulating Ca2+ regulation and insulin release. In contrast, the L-type channel blocker nifedipine (10 µM) failed to affect OXA-stimulated insulin release. Taken together, OXA increases INS-1E cell proliferation via ERK1/2-dependent mechanism. Furthermore, OXA stimulates insulin secretion from INS-1E cells. TRPs are relevant for OXA-stimulated insulin secretion and intracellular calcium regulation.


Assuntos
MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Orexinas/farmacologia , Canais de Potencial de Receptor Transitório/metabolismo , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Insulina/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Receptores de Orexina/genética , RNA Mensageiro/metabolismo , Ratos
13.
Neuropeptides ; 56: 41-9, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26853724

RESUMO

Recent data indicates that kisspeptin, encoded by the KISS1 gene, could play a role in transducing metabolic information into the hypothalamic-pituitary-gonadal (HPG) axis, the mechanism that controls reproductive functions. Numerous studies have shown that in a state of negative energy balance, the hypothalamic kisspeptin system is impaired. However, data concerning positive energy balance (e.g. diabetes and obesity) and the role of kisspeptin in the peripheral tissues is scant. We hypothesized that: 1) in diet-induced obese (DIO) male rats and/or rats with diabetes type 1 (DM1) and type 2 (DM2), altered reproductive functions are related to an imbalance in Kiss1 and GPR54 mRNA in the HPG axis; and 2) in DIO and/or DM1 and/or DM2 rats, Kiss1 and GPR 54 expression are altered in the peripheral tissues involved in metabolic functions (fat, pancreas and liver). Animals were fed a high-fat or control diets and STZ (streptozotocin - toxin, which destroys the pancreas) was injected in high or low doses to induce diabetes type 1 (DM1) or diabetes type 2 (DM2), respectively. RT-PCR and Western blot techniques were used to assess the expression of Kiss1 and GRP54 in tissues. At the level of mRNA, we found that diabetic but not obese rats have alterations in Kiss1 and/or GPR54 mRNA levels in the HPG axis as well as in peripheral tissues involved in metabolic functions (fat, pancreas and liver). The most severe changes were seen in DM1 rats. However, in the case of protein levels in the peripheral tissues (fat, pancreas and liver), changes in Kiss1/GPR54 expression were noticed in DIO, DM1 and DM2 animals and were tissue-specific. Our data support the hypothesis that alterations in Kiss1/GPR54 balance may account for both reproductive and metabolic abnormalities reported in obese and diabetic rats.


Assuntos
Diabetes Mellitus Experimental/metabolismo , Gônadas/metabolismo , Hipotálamo/metabolismo , Kisspeptinas/metabolismo , Obesidade/metabolismo , Hipófise/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animais , Dieta Hiperlipídica , Masculino , RNA Mensageiro/metabolismo , Ratos , Ratos Wistar , Receptores de Kisspeptina-1
14.
Biochim Biophys Acta ; 1853(12): 3202-10, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26384871

RESUMO

Transient receptor potential channel vanilloid type 6 (TRPV6) is a non-selective cation channel with high permeability for Ca²âº ions. So far, the role of TRPV6 in pancreatic beta cells is unknown. In the present study, we characterized the role of TRPV6 in controlling calcium signaling, cell proliferation as well as insulin expression, and secretion in experimental INS-1E beta cell model. TRPV6 protein production was downregulated using siRNA by approx. 70%, as detected by Western blot. Intracellular free Ca²âº ([Ca²âº]i) was measured by fluorescence Ca²âº imaging using fura-2. Calcineurin/NFAT signaling was analyzed using a NFAT reporter assay as well as a calcineurin activity assay. TRPV6 downregulation resulted in impaired cellular calcium influx. Its downregulation also reduced cell proliferation and decreased insulin mRNA expression. These changes were companied by the inhibition of the calcineurin/NFAT signaling. In contrast, insulin exocytosis was not affected by TRPV6 downregulation. In conclusion, this study demonstrates for the first time the expression of TRPV6 in INS-1E cells and rat pancreatic beta cells and describes its role in modulating calcium signaling, beta cell proliferation and insulin mRNA expression. In contrast, TRPV6 fails to influence insulin secretion.


Assuntos
Proliferação de Células/fisiologia , Insulinoma/metabolismo , Canais de Cátion TRPV/fisiologia , Animais , Cálcio/metabolismo , Linhagem Celular Tumoral , Homeostase , Insulina/metabolismo , Secreção de Insulina , Insulinoma/patologia , Fosforilação , Ratos , Canais de Cátion TRPV/genética , Canais de Cátion TRPV/metabolismo
15.
Cell Signal ; 26(1): 41-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24075930

RESUMO

Capsaicin (CAP), the pungent ingredient of chili peppers, inhibits growth of various solid cancers via TRPV1 as well as TRPV1-independent mechanisms. Recently, we showed that TRPV1 regulates intracellular calcium level and chromogranin A secretion in pancreatic neuroendocrine tumor (NET) cells. In the present study, we characterize the role of the TRPV1 agonist - CAP - in controlling proliferation and apoptosis of pancreatic BON and QGP-1 NET cells. We demonstrate that CAP reduces viability and proliferation, and stimulates apoptotic death of NET cells. CAP causes mitochondrial membrane potential loss, inhibits ATP synthesis and reduces mitochondrial Bcl-2 protein production. In addition, CAP increases cytochrome c and cleaved caspase 3 levels in cytoplasm. CAP reduces reactive oxygen species (ROS) generation. The antioxidant N-acetyl-l-cysteine (NAC) acts synergistically with CAP to reduce ROS generation, without affecting CAP-induced toxicity. TRPV1 protein reduction by 75% reduction fails to attenuate CAP-induced cytotoxicity. In summary, these results suggest that CAP induces cytotoxicity by disturbing mitochondrial potential, and inhibits ATP synthesis in NET cells. Stimulation of ROS generation by CAP appears to be a secondary effect, not related to CAP-induced cytotoxicity. These results justify further evaluation of CAP in modulating pancreatic NETs in vivo.


Assuntos
Capsaicina/farmacologia , Mitocôndrias/metabolismo , Tumores Neuroendócrinos/patologia , Neoplasias Pancreáticas/patologia , Trifosfato de Adenosina/metabolismo , Apoptose/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Humanos , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Tumores Neuroendócrinos/metabolismo , Neoplasias Pancreáticas/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo
16.
FEBS Lett ; 587(19): 3281-7, 2013 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-23999312

RESUMO

Transient receptor potential channel vanilloid type 4 (TRPV4) is a Ca(2+)- and Mg(2+)-permeable cation channel that influences oxidative metabolism and insulin sensitivity. The role of TRPV4 in pancreatic beta cells is largely unknown. Here, we characterize the role of TRPV4 in controlling intracellular Ca(2+) and insulin secretion in INS-1E beta cells. Osmotic, thermal or pharmacological activation of TRPV4 caused a rapid rise of intracellular Ca(2+) and enhanced glucose-stimulated insulin secretion. In the presence of the TRPV channel blocker ruthenium red (RuR) or after suppression of TRPV4 protein production, TRPV4 activators failed to increase [Ca(2+)]i and insulin secretion in INS-1E cells.


Assuntos
Cálcio/metabolismo , Glucose/metabolismo , Insulina/metabolismo , Ilhotas Pancreáticas/metabolismo , Canais de Cátion TRPV/metabolismo , Linhagem Celular , Humanos , Secreção de Insulina
17.
J Biol Regul Homeost Agents ; 27(1): 23-33, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23489684

RESUMO

Ghrelin and obestatin are encoded by the preproghrelin gene and originate from post-translational processing of the preproghrelin peptide. Obestatin is mainly present in the stomach, but its action is focused on appetite inhibition in opposition to ghrelin function. Recently, it has been presented that obestatin may regulate adipocyte metabolism and influence fat content. However, obestatin action is still poorly understood. Therefore, we aimed to investigate obestatin function on adipocyte metabolism in the rat. We studied changes in the mRNA expression of active and inactive isoforms of obestatin receptors. In addition, we analyzed influence of obestatin on lipogenesis, lipolysis and glucose transport in isolated adipocytes. Moreover, we also performed analysis of obestatin action on lipolysis in differentiated rat preadipocytes with silenced obestatin receptor. We found significantly higher expression of the obestatin receptor Gpr39-1a active form at an mRNA level following adipocytes incubation with obestatin. We did not observe expression changes in the inactive form of obestatin receptor Gpr39-1b. Additionally, we found significant changes in Gpr39-1a expression following obestatin receptor silencing in cells incubated with obestatin in comparison to control. Obestatin inhibited both, basal and insulin-stimulated lipogenesis and glucose transport in adipocytes. Furthermore, obestatin potentiated adrenalin-stimulated lipolysis. We also found reduced glycerol release following obestatin incubation in adipocytes with silenced Gpr39 gene. Our results indicate that obestatin acts via the GPR39 receptor in isolated adipocytes, and that through this mechanism obestatin influences lipid accumulation, glucose uptake and lipolysis.


Assuntos
Adipócitos/metabolismo , Grelina/farmacologia , Glucose/metabolismo , Lipogênese/efeitos dos fármacos , Adipócitos/efeitos dos fármacos , Animais , Transporte Biológico/efeitos dos fármacos , Separação Celular , Células Cultivadas , Epinefrina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Inativação Gênica/efeitos dos fármacos , Insulina/farmacologia , Lipogênese/genética , Lipólise/efeitos dos fármacos , Masculino , Ratos , Ratos Wistar , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo
18.
Diabetologia ; 56(3): 588-97, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23262585

RESUMO

AIMS/HYPOTHESIS: Glucagon reduces body weight by modifying food intake, glucose/lipid metabolism and energy expenditure. All these physiological processes are also controlled by fibroblast growth factor 21 (FGF-21), a circulating hepatokine that improves the metabolic profile in obesity and type 2 diabetes. Animal experiments have suggested a possible interaction between glucagon and FGF-21 however, the metabolic consequences of this crosstalk are not understood. METHODS: The effects of exogenous glucagon on plasma FGF-21 levels and lipolysis were evaluated in healthy volunteers and humans with type 1 diabetes, as well as in rodents with streptozotocin (STZ)-induced insulinopenic diabetes. In vitro, the role of glucagon on FGF-21 secretion and lipolysis was studied using isolated primary rat hepatocytes and adipocytes. Fgf-21 expression in differentiated rat pre-adipocytes was suppressed by small interfering RNA and released FGF-21 was immunoneutralised by polyclonal antibodies. RESULTS: Glucagon induced lipolysis in healthy human volunteers, patients with type 1 diabetes, mice and rats with STZ-induced insulinopenic diabetes, and in adipocytes isolated from diabetic and non-diabetic animals. In addition, glucagon increased circulating FGF-21 in healthy humans and rodents, as well as in patients with type 1 diabetes, and insulinopenic rodents. Glucagon stimulated FGF-21 secretion from isolated primary hepatocytes and adipocytes derived from animals with insulinopenic diabetes. Furthermore, FGF-21 stimulated lipolysis in primary adipocytes isolated from non-diabetic and diabetic rats. Reduction of Fgf-21 expression (by approximately 66%) or immunoneutralisation of released FGF-21 markedly attenuated glucagon-stimulated lipolysis in adipocytes. CONCLUSIONS/INTERPRETATION: These results indicate that glucagon increases circulating FGF-21 independently of endogenous insulin levels. FGF-21 participates in glucagon-induced stimulation of lipolysis.


Assuntos
Diabetes Mellitus Tipo 1/sangue , Fatores de Crescimento de Fibroblastos/sangue , Glucagon/farmacologia , Insulina/sangue , Lipólise/efeitos dos fármacos , Células 3T3-L1 , Adulto , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Diabetes Mellitus Experimental/sangue , Feminino , Humanos , Masculino , Camundongos , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase em Tempo Real
19.
FEBS Lett ; 586(23): 4157-64, 2012 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-23123090

RESUMO

Metabolic activities of orexin A (OXA) in mature adipocytes are mediated via PI3K/PKB and PPARγ. However, the effects of OXA on preadipocytes are largely unknown. We report here that OXA stimulates the proliferation and viability of 3T3-L1 preadipocytes and protects them from apoptosis via ERK1/2, but not through PKB. OXA reduces proapoptotic activity of caspase-3 via ERK1/2. Inhibition of ERK1/2 prevents the differentiation of preadipocytes into adipocytes. Unlike insulin, neither short-term nor prolonged exposure of 3T3-L1 preadipocytes to OXA induces preadipocyte differentiation to adipocytes, despite increased ERK1/2 phosphorylation. Unlike insulin, OXA fails to activate PKB, which explains its inability to induce the differentiation of preadipocytes.


Assuntos
Adipócitos/citologia , Adipócitos/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Peptídeos e Proteínas de Sinalização Intracelular/farmacologia , Neuropeptídeos/farmacologia , Células 3T3-L1 , Adipócitos/metabolismo , Animais , Apoptose/genética , Caspase 3/metabolismo , Diferenciação Celular/genética , Proliferação de Células/efeitos dos fármacos , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Orexinas , Reação em Cadeia da Polimerase em Tempo Real
20.
Transplant Proc ; 44(9): 2754-6, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23146514

RESUMO

BACKGROUND: With an increasing number of patients with hepatocellular carcinoma (HCC) undergoing liver transplantation (OLT), HCC recurrence remains the main limiting factor for long-term survival. We herein report our experience with sorafenib treatment for HCC recurrence post-OLT. PATIENTS AND METHODS: We reviewed data on transplanted HCC patients receiving sorafenib for HCC recurrence. RESULTS: Fourteen patients were included for the period November 2006 to February 2011. There were 9 men and 5 women of median age of 57 years. Twelve patients (86%) received rescue grafts through Eurotransplant allocation. Median values for alpha fetoprotein levels, Model for End-Stage Liver Disease score, sorafenib daily dose, and length of treatment were 97 ng/mL, 10, 400 mg, and 6.5 months, respectively. Sorafenib side effects led to discontinuation (n = 4) or reduction (n = 2) of the daily dose. Four patients experienced tumor progression during treatment. Seven patients are currently alive, 3 patients died of tumor progression, and 4 patients of non-tumor-related causes of death. Median survival was 25 months. CONCLUSION: Sorafenib treatment for HCC recurrence in transplant recipients represents a challenging oncologic approach that requires further validation in prospective, multicenter studies.


Assuntos
Antineoplásicos/uso terapêutico , Carcinoma Hepatocelular/tratamento farmacológico , Carcinoma Hepatocelular/cirurgia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/cirurgia , Transplante de Fígado/efeitos adversos , Recidiva Local de Neoplasia/tratamento farmacológico , Niacinamida/análogos & derivados , Compostos de Fenilureia/uso terapêutico , Inibidores de Proteínas Quinases/uso terapêutico , Antineoplásicos/efeitos adversos , Carcinoma Hepatocelular/mortalidade , Feminino , Humanos , Neoplasias Hepáticas/mortalidade , Transplante de Fígado/mortalidade , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/mortalidade , Niacinamida/efeitos adversos , Niacinamida/uso terapêutico , Compostos de Fenilureia/efeitos adversos , Inibidores de Proteínas Quinases/efeitos adversos , Estudos Retrospectivos , Sorafenibe , Fatores de Tempo , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA