Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
J Physiol Pharmacol ; 74(4)2023 Aug.
Article in English | MEDLINE | ID: mdl-37865959

ABSTRACT

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.


Subject(s)
Leptin , Obesity , Rats , Animals , Obesity/metabolism , Anilides/pharmacology , Overweight , Adiponectin
2.
J Physiol Pharmacol ; 73(5)2022 Oct.
Article in English | MEDLINE | ID: mdl-36942807

ABSTRACT

Hepcidin is a primary regulator of iron metabolism in the human body. By promoting ferroportin degradation, hepcidin reduces intestinal iron absorption and its release from intracellular stores. In the course of pregnancy, gradually declining hepcidin concentrations encourage placental iron transfer, thereby providing the appropriate amount of iron for fetal development. Hence, we aimed to investigate changes in maternal and cord blood hepcidin and iron metabolism parameters in normal-weight (n=17) and obese (n=17) gestating women, as well as gravid women with a history of hypothyroidism following the restoration of euthyroidism (n=17). All blood samples were taken on the day of delivery, and ELISA kits were used for measurements. A significant increase in maternal hepcidin concentration was observed in obese pregnant women, compared to normal-weight controls (29.53±4.20 ng/mL vs. 25.69±5.70 ng/mL; P<0.05). However, only a slight, insignificant tendency for lower hepcidin was noted in the hypothyroid group, compared to the healthy controls (23.10±6.00 ng/mL vs. 25.69±5.70 ng/mL; P=NS). Moreover, decreased maternal free triiodothyronine, triiodothyronine, free thyroxine, and ferritin levels were revealed in the hypothyroid group, compared to the normal-weight individuals (P<0.05). Furthermore, positive correlations between maternal hepcidin and the majority of maternal thyroid hormones were found, with a most potent relation to FT3 (r=0.40; P<0.01). Interestingly, no alterations of thyroid hormones and iron metabolism parameters were noticed in cord blood in any of the subgroups. In summary, pre-pregnancy obesity is associated with elevated maternal hepcidin, albeit no signs of lowered cord blood iron status were shown. Medical history of hypothyroidism following the restoration of euthyroidism does not substantially influence maternal nor cord blood hepcidin concentration, as well as fetal iron homeostasis, even though free thyroid hormone levels correlate with maternal hepcidin.


Subject(s)
Fetal Blood , Hypothyroidism , Female , Pregnancy , Humans , Fetal Blood/metabolism , Pilot Projects , Triiodothyronine/metabolism , Placenta/metabolism , Iron/metabolism , Obesity/metabolism , Hypothyroidism/metabolism
3.
Eur Rev Med Pharmacol Sci ; 25(19): 5993-6002, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34661259

ABSTRACT

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.


Subject(s)
Fetal Blood/metabolism , Kisspeptins/blood , Obesity/epidemiology , Adult , Female , Humans , Infant, Newborn , Male , Mothers , Obesity/blood , Pilot Projects , Pregnancy
4.
Br Poult Sci ; 61(3): 294-302, 2020 Jun.
Article in English | MEDLINE | ID: mdl-31955595

ABSTRACT

1. The aim of the present study was to investigate the effect of partial (50%) or total replacement of soybean oil (SO) by black soldier fly larvae (BSFL) fat on the growth performance, coefficients of apparent nutrient digestibility, selected internal organ weights and length, pancreatic enzyme activity and gastrointestinal tract (GIT) microecology modulation, as well as microbiota activity, physiological and immunological responses in young turkey poults. 2. A total of 216, seven day old female turkeys (B.U.T 6) were randomly distributed to three dietary treatments using six replicate pens per group with 12 birds per pen. The following design of the trial was applied: SO 100% soybean oil; BSFL50 a 50/50 combination of SO and BSFL fat; or 100% BSFL fat (total replacement of SO). 3. The use of BSFL fat did not affect the growth performance, nutrient digestibility, GIT morphology, or quality of the breast and thigh muscles. However, reduced trypsin activity was noticed in the BSFL100 group, but this had no effect on digestibility. Total replacement of SO reduced proliferation of potentially pathogenic bacteria, i.e., Enterobacteriaceae spp., as well as decreasing levels of IL-6, while partial substitution lowered the TNF-α concentration. 4. The replacement of commonly used SO by BSFL fat can be successfully applied in young turkey poult nutrition. BSFL fat may be considered an antimicrobial agent and support immune responses.


Subject(s)
Microbiota , Simuliidae , Animal Feed , Animals , Chickens , Larva , Soybean Oil , Turkeys
5.
Domest Anim Endocrinol ; 63: 59-68, 2018 04.
Article in English | MEDLINE | ID: mdl-29413903

ABSTRACT

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.


Subject(s)
Adipocytes/drug effects , Leptin/metabolism , Lipid Metabolism/drug effects , Orexins/pharmacology , Adipocytes/metabolism , Animals , Biological Transport , Cell Survival , Cells, Cultured , Glucose/metabolism , Lipogenesis/drug effects , Male , Swine
6.
Physiol Res ; 67(1): 45-56, 2018 03 16.
Article in English | MEDLINE | ID: mdl-29137471

ABSTRACT

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.


Subject(s)
Insulin Resistance/physiology , Kisspeptins/blood , Obesity/blood , Peptide Hormones/blood , Adult , Biomarkers/blood , Female , Humans , Middle Aged , Obesity/diagnosis
7.
Poult Sci ; 96(11): 4026-4037, 2017 Sep 01.
Article in English | MEDLINE | ID: mdl-29050441

ABSTRACT

The aim of this study was to investigate the effect of dietary supplementation with nisin alone or in combination with salinomycin or monensin on broiler chickens in terms of growth performance, selected blood parameters, digestive enzyme activity, apparent nutrient digestibility, and tibiotarsus mineralization, as well as selected gastrointestinal tract (GIT) organ weights, intestinal length, and central immune organ weights. Two independent experiments, each including 400 one-day-old female Ross 308 chicks differing in ionophore coccidiostats, i.e., salinomycin and monensin supplementation, were conducted. The following treatments were applied: experiment 1: NA-no additives, SAL-salinomycin (60 mg/kg diet), NIS-nisin (2,700 IU/kg diet), SAL+NIS-salinomycin (60 mg/kg diet) and nisin (2,700 IU/kg diet); experiment 2: NA-no additives, MON-monensin (100 mg/kg diet), NIS-nisin (2,700 IU/kg diet) and MON+NIS-monensin (100 mg/kg diet) and nisin (2,700 IU/kg diet). The addition of nisin with or without ionophores to the birds' diet improved broiler growth performance in terms of BWG and FCR (days 1 to 14) and BWG and FI (15 to 35 d; 1 to 35 d). Salinomycin showed effects similar to those of nisin influence on growth performance (1 to 35 d), while monensin supplementation resulted in lower BWG. Moreover, no additive effect between nisin and ionophores was observed. Nisin and salinomycin had no influence on the serum concentration of selected hormones and other blood biochemical parameters except glucose, which was reduced by nisin. A decrease in lipase activity was observed during nisin and salinomycin supplementation, while the apparent ileal digestibility of fat was not affected. However, the digestibility of crude protein increased with nisin administration. Additionally, the effects of nisin on decreasing the weight and length of GIT segments were observed. Supplementation with nisin and monensin was not associated with a negative impact on tibiotarsus mineralization and the immune organ index. This study suggests that nisin may be used in broiler nutrition as a growth promotor, with no negative influence on the bird's metabolism or immune status.


Subject(s)
Chickens/physiology , Coccidiostats/adverse effects , Digestion/drug effects , Monensin/adverse effects , Nisin/adverse effects , Pyrans/adverse effects , Animal Feed/analysis , Animal Nutritional Physiological Phenomena/drug effects , Animals , Chickens/growth & development , Diet/veterinary , Dietary Supplements/analysis , Female , Random Allocation
8.
Cell Signal ; 35: 242-249, 2017 07.
Article in English | MEDLINE | ID: mdl-28359774

ABSTRACT

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.


Subject(s)
Calcium/metabolism , Insulin/genetics , Nitric Oxide/metabolism , TRPV Cation Channels/genetics , Animals , Cell Proliferation/genetics , Gene Expression Regulation/drug effects , Humans , Insulin/metabolism , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Leucine/administration & dosage , Leucine/analogs & derivatives , MAP Kinase Signaling System/drug effects , Phosphorylation , RNA, Messenger/genetics , Rats , Reactive Oxygen Species/metabolism , Sulfonamides/administration & dosage , TRPV Cation Channels/metabolism
9.
J Physiol Pharmacol ; 68(5): 669-681, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29375041

ABSTRACT

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.


Subject(s)
Blood Glucose/drug effects , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Type 2/drug therapy , Insulin-Secreting Cells/drug effects , Orexins/therapeutic use , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Experimental/blood , Diabetes Mellitus, Type 2/blood , Insulin-Secreting Cells/metabolism , Male , Orexins/pharmacology , Rats , Treatment Outcome
10.
J Physiol Pharmacol ; 68(6): 815-821, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29550793

ABSTRACT

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.


Subject(s)
Islets of Langerhans/metabolism , Orexins/metabolism , Animals , Caspase 3/metabolism , Cell Survival , Extracellular Signal-Regulated MAP Kinases/metabolism , Glucagon/metabolism , Insulin/metabolism , Insulin Secretion , Orexin Receptors/genetics , Orexin Receptors/metabolism , Swine
11.
Neuropeptides ; 56: 41-9, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26853724

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Gonads/metabolism , Hypothalamus/metabolism , Kisspeptins/metabolism , Obesity/metabolism , Pituitary Gland/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Diet, High-Fat , Male , RNA, Messenger/metabolism , Rats , Rats, Wistar , Receptors, Kisspeptin-1
12.
Poult Sci ; 94(8): 1909-16, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26112038

ABSTRACT

The purpose of the study was to examine the effect of 2 prebiotics and 2 synbiotics on the digestive potency of pancreas in 1-, 3-, 7-, 14-, 21-, and 34-day-old cockerels. Prebiotics (inulin and Bi²tos) and synbiotics (inulin + Lactococcus lactis subsp. lactis and Bi²tos + Lactococcus lactis subsp. cremoris) were injected in ovo into the air cell on the 12th d embryonic development. Their application increased the activity of amylase, lipase, and trypsin in the pancreas. The most pronounced changes were observed at the end of the investigated rearing period (d 34). The strongest stimulative effects on amylase were shown by both synbiotics, on lipase synbiotic Bi²tos + Lactococcus lactis subsp. cremoris, and on trypsin all the used prebiotics and synbiotics. Simultaneously, neither the absolute nor the relative mass of the pancreas in comparison to control group were changed. Also, the injected in ovo compounds did not cause a deterioration in the posthatching condition of the chicken liver, as determined by measurement of the activity of marker enzymes in the blood (alanine aminotransferase and aspartate aminotransferase). Treatment with the prebiotics and synbiotics did not change the feed conversion ratio but Bi²tos (galacto-oligosaccharide) and inulin (fructan) + Lactococcus lactis subsp. lactis significantly increased final BW.


Subject(s)
Amylases/metabolism , Chickens , Pancreas/drug effects , Prebiotics , Synbiotics , Animals , Chick Embryo , Male , Pancreas/enzymology , Pancreas/growth & development
13.
Cell Signal ; 26(1): 41-8, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24075930

ABSTRACT

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.


Subject(s)
Capsaicin/pharmacology , Mitochondria/metabolism , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/pathology , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Neuroendocrine Tumors/metabolism , Pancreatic Neoplasms/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Time Factors
14.
J Biol Regul Homeost Agents ; 27(3): 647-54, 2013.
Article in English | MEDLINE | ID: mdl-24152833

ABSTRACT

Adipocyte-originated hormonal factors, playing a role of signaling particles, are widely engaged in energy control, feeding behavior and general glucose or lipid metabolism. One of them – resistin – has been suspected to initiate or develop insulin resistance and diabetes. From the moment of discovery of resistin, during last 13 years, numerous investigations put some light on a potential role of this hormone in mammals. In this review knowledge on resistin, including its structure, physiological role related to obesity and diabetes, as well as, gene sequence and phenotypic effects of the identified polymorphisms in human and domestic mammals is discussed.


Subject(s)
Diabetes Mellitus/etiology , Resistin/physiology , Diabetes Mellitus/genetics , Gene Expression Regulation , Humans , Polymorphism, Genetic , Resistin/chemistry , Resistin/genetics
15.
FEBS Lett ; 587(19): 3281-7, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23999312

ABSTRACT

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.


Subject(s)
Calcium/metabolism , Glucose/metabolism , Insulin/metabolism , Islets of Langerhans/metabolism , TRPV Cation Channels/metabolism , Cell Line , Humans , Insulin Secretion
16.
Poult Sci ; 92(8): 2124-34, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23873561

ABSTRACT

A total of 1,200 Ross broiler chickens were used in 2 separate feeding studies to explore the effect of myo-inositol (MYO) and phytase on performance and blood biochemistry of broilers fed diets formulated to be either adequate or insufficient in Ca and digestible P (dP). Supplementation of diets that were formulated to be insufficient in Ca and dP with MYO resulted in improved BW gain and feed conversion ratio in both experiments. However, these effects were most pronounced in the finisher phase, and moderate negative effects were observed during the starter period. Supplementation of the diet with microbial phytase improved BW gain and feed conversion ratio to a similar extent as was observed with MYO, and there was a degree of subadditivity between the 2 additives. Blood glucose concentrations were increased by both MYO and phytase, though possibly by different mechanisms, because insulin concentrations were not directly relatable to circulating glucose levels, especially when both MYO and phytase were applied simultaneously. The increase in blood glucose concentrations with MYO and phytase was most pronounced in the diet with a lower Ca and dP concentration. It can be concluded that dietary supplementation with MYO or phytase was effective in improving performance of commercial broiler chickens. However, further work is required to explore complex ontogenetic effects of MYO and possible involvement of both MYO and phytase in Na-dependent transport mechanisms.


Subject(s)
6-Phytase/pharmacology , Chickens/blood , Chickens/growth & development , Inositol/pharmacology , Triticum/chemistry , Zea mays/chemistry , 6-Phytase/metabolism , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Calcium/blood , Calcium/metabolism , Diet/veterinary , Inositol/chemistry , Male , Phosphorus/blood , Phosphorus/metabolism
17.
J Biol Regul Homeost Agents ; 27(1): 23-33, 2013.
Article in English | MEDLINE | ID: mdl-23489684

ABSTRACT

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.


Subject(s)
Adipocytes/metabolism , Ghrelin/pharmacology , Glucose/metabolism , Lipogenesis/drug effects , Adipocytes/drug effects , Animals , Biological Transport/drug effects , Cell Separation , Cells, Cultured , Epinephrine/pharmacology , Gene Expression Regulation/drug effects , Gene Silencing/drug effects , Insulin/pharmacology , Lipogenesis/genetics , Lipolysis/drug effects , Male , Rats , Rats, Wistar , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism
18.
Diabetologia ; 56(3): 588-97, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23262585

ABSTRACT

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.


Subject(s)
Diabetes Mellitus, Type 1/blood , Fibroblast Growth Factors/blood , Glucagon/pharmacology , Insulin/blood , Lipolysis/drug effects , 3T3-L1 Cells , Adult , Animals , Blotting, Western , Cell Differentiation/drug effects , Cells, Cultured , Diabetes Mellitus, Experimental/blood , Female , Humans , Male , Mice , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction
19.
FEBS Lett ; 586(23): 4157-64, 2012 Nov 30.
Article in English | MEDLINE | ID: mdl-23123090

ABSTRACT

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.


Subject(s)
Adipocytes/cytology , Adipocytes/drug effects , Apoptosis/drug effects , Cell Differentiation/drug effects , Intracellular Signaling Peptides and Proteins/pharmacology , Neuropeptides/pharmacology , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Apoptosis/genetics , Caspase 3/metabolism , Cell Differentiation/genetics , Cell Proliferation/drug effects , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Orexins , Real-Time Polymerase Chain Reaction
20.
Diabetologia ; 54(7): 1841-52, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21505958

ABSTRACT

AIMS/HYPOTHESIS: Orexin A (OXA) modulates body weight, food intake and energy expenditure. In vitro, OXA increases PPARγ (also known as PPARG) expression and inhibits lipolysis, suggesting direct regulation of lipid metabolism. Here, we characterise the metabolic effects and mechanisms of OXA action in adipocytes. METHODS: Isolated rat adipocytes and differentiated murine 3T3-L1 adipocytes were exposed to OXA in the presence or absence of phosphoinositide 3-kinase (PI3K) inhibitors. Pparγ expression was silenced using small interfering RNA. Glucose uptake, GLUT4 translocation, phosphatidylinositol (3,4,5)-trisphosphate production, lipogenesis, lipolysis, and adiponectin secretion were measured. Adiponectin plasma levels were determined in rats treated with OXA for 4 weeks. RESULTS: OXA PI3K-dependently stimulated active glucose uptake by translocating the glucose transporter GLUT4 from cytoplasm into the plasma membrane. OXA increased cellular triacylglycerol content via PI3K. Cellular triacylglycerol accumulation resulted from increased lipogenesis as well as from a decrease of lipolysis. Adiponectin levels in chow- and high-fat diet-fed rats treated chronically with OXA were increased. OXA stimulated adiponectin expression and secretion in adipocytes. Both pharmacological blockade of peroxisome proliferator-activated receptor γ (PPARγ) activity or silencing Pparγ expression prevented OXA from stimulating triacylglycerol accumulation and adiponectin production. CONCLUSIONS/INTERPRETATION: Our study demonstrates that OXA stimulates glucose uptake in adipocytes and that the evolved energy is stored as lipids. OXA increases lipogenesis, inhibits lipolysis and stimulates the secretion of adiponectin. These effects are conferred via PI3K and PPARγ2. Overall, OXA's effects on lipids and adiponectin secretion resemble that of insulin sensitisers, suggesting a potential relevance of this peptide in metabolic disorders.


Subject(s)
Adipocytes/drug effects , Adipocytes/metabolism , Glucose/metabolism , Intracellular Signaling Peptides and Proteins/pharmacology , Lipid Metabolism/drug effects , Neuropeptides/pharmacology , Neurotransmitter Agents/pharmacology , 3T3-L1 Cells , Animals , Biological Transport/drug effects , Blotting, Western , Cells, Cultured , Male , Mice , Orexins , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL
...