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










Publication year range
1.
Physiol Res ; 72(5): 607-619, 2023 11 28.
Article in English | MEDLINE | ID: mdl-38015760

ABSTRACT

Recent data suggest that the orexigenic peptide ghrelin and liver-expressed antimicrobial peptide 2 (LEAP2) have opposing effects on food intake regulation. Although circulating ghrelin is decreased in obesity, peripheral ghrelin administration does not induce food intake in obese mice. Limited information is available on ghrelin resistance in relation to LEAP2. In this study, the interplay between ghrelin and LEAP2 in obesity induced by a high-fat (HF) diet in mice was studied. First, the progression of obesity and intolerance to glucose together with plasma levels of active and total ghrelin, leptin, as well as liver LEAP2 mRNA expression at different time points of HF diet feeding was examined. In addition, the impact of switch from a HF diet to a standard diet on plasma ghrelin and LEAP2 production was studied. Second, sensitivity to the stable ghrelin analogue [Dpr3]Ghrelin or our novel LEAP2 analogue palm-LEAP2(1-14) during the progression of HF diet-induced obesity and after the switch for standard diet was investigated. Food intake was monitored after acute subcutaneous administration. HF diet feeding decreased both active and total plasma ghrelin and increased liver LEAP2 mRNA expression along with intolerance to glucose and the switch to a standard diet normalized liver LEAP2 mRNA expression and plasma level of active ghrelin, but not of total ghrelin. Additionally, our study demonstrates that a HF diet causes resistance to [Dpr3]Ghrelin, reversible by switch to St diet, followed by resistance to palm-LEAP2(1-14). Further studies are needed to determine the long-term effects of LEAP2 analogues on obesity-related ghrelin resistance.


Subject(s)
Diet, High-Fat , Ghrelin , Animals , Mice , Ghrelin/pharmacology , Glucose , Obesity/drug therapy , Receptors, Ghrelin , RNA, Messenger
2.
Physiol Res ; 70(4): 579-590, 2021 08 31.
Article in English | MEDLINE | ID: mdl-34062082

ABSTRACT

Prolactin-releasing peptide (PrRP) has been proposed to mediate the central satiating effects of cholecystokinin (CCK) through the vagal CCK1 receptor. PrRP acts as an endogenous ligand of G protein-coupled receptor 10 (GPR10), which is expressed at the highest levels in brain areas related to food intake regulation, e.g., the paraventricular hypothalamic nucleus (PVN) and nucleus of the solitary tract (NTS). The NTS and PVN are also significantly activated after peripheral CCK administration. The aim of this study was to determine whether the endogenous PrRP neuronal system in the brain is involved in the central anorexigenic effect of the peripherally administered CCK agonist JMV236 or the CCK1 antagonist devazepide and whether the CCK system is involved in the central anorexigenic effect of the peripherally applied lipidized PrRP analog palm-PrRP31 in fasted lean mice. The effect of devazepide and JMV236 on the anorexigenic effects of palm-PrRP31 as well as devazepide combined with JMV236 and palm-PrRP31 on food intake and Fos cell activation in the PVN and caudal NTS was examined. Our results suggest that the anorexigenic effect of JMV236 is accompanied by activation of PrRP neurons of the NTS in a CCK1 receptor-dependent manner. Moreover, while the anorexigenic effect of palm-PrRP31 was not affected by JMV236, it was partially attenuated by devazepide in fasted mice. The present findings indicate that the exogenously influenced CCK system may be involved in the central anorexigenic effect of peripherally applied palm-PrRP31, which possibly indicates some interaction between the CCK and PrRP neuronal systems.


Subject(s)
Appetite Depressants/administration & dosage , Cholecystokinin/metabolism , Eating/drug effects , Feeding Behavior/drug effects , Paraventricular Hypothalamic Nucleus/drug effects , Prolactin-Releasing Hormone/analogs & derivatives , Solitary Nucleus/drug effects , Animals , Chemokines, CC/drug effects , Chemokines, CC/metabolism , Devazepide/administration & dosage , Fasting , Hormone Antagonists/administration & dosage , Injections, Intraperitoneal , Injections, Subcutaneous , Male , Mice, Inbred C57BL , Paraventricular Hypothalamic Nucleus/metabolism , Peptide Fragments/administration & dosage , Prolactin-Releasing Hormone/administration & dosage , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction , Sincalide/administration & dosage , Sincalide/analogs & derivatives , Solitary Nucleus/metabolism
3.
Front Endocrinol (Lausanne) ; 11: 597583, 2020.
Article in English | MEDLINE | ID: mdl-33324348

ABSTRACT

Obesity, diabetes, insulin resistance, sedentary lifestyle, and Western diet are the key factors underlying non-alcoholic fatty liver disease (NAFLD), one of the most common liver diseases in developed countries. In many cases, NAFLD further progresses to non-alcoholic steatohepatitis (NASH), fibrosis, cirrhosis, and to hepatocellular carcinoma. The hepatic lipotoxicity and non-liver factors, such as adipose tissue inflammation and gastrointestinal imbalances were linked to evolution of NAFLD. Nowadays, the degree of adipose tissue inflammation was shown to directly correlate with the severity of NAFLD. Consumption of higher caloric intake is increasingly emerging as a fuel of metabolic inflammation not only in obesity-related disorders but also NAFLD. However, multiple causes of NAFLD are the reason why the mechanisms of NAFLD progression to NASH are still not well understood. In this review, we explore the role of food intake regulating peptides in NAFLD and NASH mouse models. Leptin, an anorexigenic peptide, is involved in hepatic metabolism, and has an effect on NAFLD experimental models. Glucagon-like peptide-1 (GLP-1), another anorexigenic peptide, and GLP-1 receptor agonists (GLP-1R), represent potential therapeutic agents to prevent NAFLD progression to NASH. On the other hand, the deletion of ghrelin, an orexigenic peptide, prevents age-associated hepatic steatosis in mice. Because of the increasing incidence of NAFLD and NASH worldwide, the selection of appropriate animal models is important to clarify aspects of pathogenesis and progression in this field.


Subject(s)
Appetite Regulation/drug effects , Disease Models, Animal , Eating , Hypoglycemic Agents/pharmacology , Inflammation/complications , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/physiopathology , Peptide Fragments/pharmacology , Animals , Disease Progression , Humans , Non-alcoholic Fatty Liver Disease/etiology
4.
Physiol Res ; 67(2): 339-346, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29303606

ABSTRACT

Neurodegenerative disorders, such as Alzheimer's disease (AD) and Parkinson's disease (PD), are increasing in prevalence. Currently, there are no effective and specific treatments for these disorders. Recently, positive effects of the orexigenic hormone ghrelin on memory and learning were demonstrated in mouse models of AD and PD. In this study, we tested the potential neuroprotective properties of a stable and long-lasting ghrelin analog, Dpr(3)ghrelin (Dpr(3)ghr), in SH-SY5Y neuroblastoma cells stressed with 1.2 mM methylglyoxal (MG), a toxic endogenous by-product of glycolysis, and we examined the impact of Dpr(3)ghr on apoptosis. Pre-treatment with both 10(-5) and 10(-7) M Dpr(3)ghr resulted in increased viability in SH-SY5Y cells (determined by MTT staining), as well as reduced cytotoxicity of MG in these cells (determined by LDH assay). Dpr(3)ghr increased viability by altering pro-apoptotic and viability markers: Bax was decreased, Bcl-2 was increased, and the Bax/Bcl-2 ratio was attenuated. The ghrelin receptor GHS-R1 and Dpr(3)ghr-induced activation of PBK/Akt were immuno-detected in SH-SY5Y cells to demonstrate the presence of GHS-R1 and GHS-R1 activation, respectively. We demonstrated that Dpr(3)ghr protected SH-SY5Y cells against MG-induced neurotoxicity and apoptosis. Our data suggest that stable ghrelin analogs may be candidates for the effective treatment of neurodegenerative disorders.


Subject(s)
Apoptosis/drug effects , Ghrelin/analogs & derivatives , Ghrelin/pharmacology , Neuroprotective Agents/pharmacology , Apoptosis Regulatory Proteins/biosynthesis , Apoptosis Regulatory Proteins/genetics , Cell Line, Tumor , Glycolysis/drug effects , Humans , L-Lactate Dehydrogenase/metabolism , MAP Kinase Signaling System/drug effects , Membrane Potential, Mitochondrial/drug effects , Neurotoxicity Syndromes/prevention & control , Pyruvaldehyde/toxicity , Receptors, Ghrelin/biosynthesis
5.
J Physiol Pharmacol ; 67(1): 121-8, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27010901

ABSTRACT

Prolactin-releasing peptide (PrRP) is an anorexigenic neuropeptide expressed in the brain where it regulates food intake and energy expenditure. The C-terminal Arg-Phe-NH2 of PrRP is crucial for its biological activity. In our previous study, we showed that PrRP analogs myristoylated or palmitoylated at the N- terminus seem to cross the blood-brain barrier and lower food intake following peripheral administration. In this study, myristoylated and palmitoylated PrRP31 analogs with a modified C-terminal Phe were designed and tested. Lipidized analogs containing Phe(31) replaced by aromatic non-coded amino acids or tyrosine revealed high binding affinity to rat pituitary RC-4B/C cells with endogenous PrRP and neuropeptide FF 2 receptors and to CHO-K1 cells overexpressing either PrRP or neuropeptide FF 2 receptors. The analogs also showed strong agonistic properties at the GPR10 receptor using the beta-lactamase reporter gene assay. Moreover, lipidized PrRP analogs, especially those that were palmitoylated, demonstrated strong and long-lasting anorexigenic effects in fasted mice after subcutaneous administration. The most efficient PrRP31 analogs with PheCl2(31), either palmitoylated or myristoylated at the N-terminus, are promising candidates for the study of food disorders, possibly for anti-obesity treatment. Despite the therapeutic potential in targeting central GPR10, the endogenous ligand PrRP cannot cross the blood-brain barrier. Understanding biological activity and transport of novel structural analogs of PrRP with a potential central anorexigenic effect is of key therapeutic significance.


Subject(s)
Neuropeptides/pharmacology , Prolactin-Releasing Hormone/analogs & derivatives , Prolactin-Releasing Hormone/pharmacology , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , CHO Cells , Cell Line , Cricetulus , Eating/drug effects , Humans , Lipids , Male , Mice , Mice, Inbred C57BL , Neuropeptides/metabolism , Obesity/drug therapy , Obesity/metabolism , Prolactin-Releasing Hormone/metabolism , Rats , Receptors, Neuropeptide/metabolism
6.
Int J Obes (Lond) ; 39(6): 986-93, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25771926

ABSTRACT

OBJECTIVES: Obesity is a frequent metabolic disorder but an effective therapy is still scarce. Anorexigenic neuropeptides produced and acting in the brain have the potential to decrease food intake and ameliorate obesity but are ineffective after peripheral application. We have designed lipidized analogs of prolactin-releasing peptide (PrRP), which is involved in energy balance regulation as demonstrated by obesity phenotypes of both PrRP- and PrRP-receptor-knockout mice. RESULTS: Lipidized PrRP analogs showed binding affinity and signaling in PrRP receptor-expressing cells similar to natural PrRP. Moreover, these analogs showed high binding affinity also to anorexigenic neuropeptide FF-2 receptor. Peripheral administration of myristoylated and palmitoylated PrRP analogs to fasted mice induced strong and long-lasting anorexigenic effects and neuronal activation in the brain areas involved in food intake regulation. Two-week-long subcutaneous administration of palmitoylated PrRP31 and myristoylated PrRP20 lowered food intake, body weight and improved metabolic parameters, and attenuated lipogenesis in mice with diet-induced obesity. CONCLUSIONS: Our data suggest that the lipidization of PrRP enhances stability and mediates its effect in central nervous system. Strong anorexigenic and body-weight-reducing effects make lipidized PrRP an attractive candidate for anti-obesity treatment.


Subject(s)
Anti-Obesity Agents/pharmacology , Lipids/chemistry , Obesity/prevention & control , Prolactin-Releasing Hormone/pharmacology , Animals , Appetite Regulation , Eating , Energy Metabolism , Half-Life , Male , Mice , Mice, Inbred C57BL , Prolactin-Releasing Hormone/analogs & derivatives , Signal Transduction
7.
J Physiol Pharmacol ; 65(4): 477-86, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25179080

ABSTRACT

Catecholamines participate in the food intake regulation, however, there are no literature data available, dealing with the activity of tyrosine hydroxylase (TH) neurons in response to stimulation or inhibition of GHS-R (growth hormone secretagogue receptor) in the hypothalamic arcuate nucleus (ARC). The present study was focused to reveal whether [Dpr(N-octanoyl) 3ghrelin], a stable GHS-R agonist, itself in doses of 5 or 10 mg/kg (s.c.) or in combination with GHS-R receptor antagonist ([DLys3]GHRP-6) in dose of 10 mg/kg (s.c.), may affect the activity of ARC TH-containing neurons in C57BL/6 male mice fed either with standard (SD) or high fat diet (HFD) that developed a diet-induced obesity (DIO). The data of the present study clearly indicate that both doses of GHS-R agonist stimulated food intake in SD mice and GHS-R antagonist significantly reduced GHS-R agonist orexinergic effect in SD mice and suppressed the voluntary food intake in HFD mice. Both doses of the GHS-R agonist stimulated Fos expression in ARC neurons in both diet groups of mice which was not abolished by GHS-R antagonist pretreatment. Moreover, both doses of the GHS-R agonist significantly influenced the activation of TH neurons in the ARC of SD mice. The GHS-R antagonist also significantly increased TH neurons activation after GHS-R agonist although this effect was less powerful in HFD mice. This is the first study demonstrating response of local ARC TH neurons to peripherally applied GHS-R agonist and antagonist. The present data point out that the response of TH neurons to GHS-R agonist and antagonist is different in normal and DIO mice and extend our knowledge about the further ARC neuronal phenotype responding to peripheral ghrelin. To bring insight into the understanding of the functional significance of the activated TH neurons in ARC, in the context of the ghrelin peripheral increase, further studies are required.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Diet, High-Fat , Receptors, Ghrelin/agonists , Receptors, Ghrelin/antagonists & inhibitors , Tyrosine 3-Monooxygenase/metabolism , Animals , Arcuate Nucleus of Hypothalamus/drug effects , Male , Mice, Inbred C57BL , Neurons/drug effects , Peptides/pharmacology , Proto-Oncogene Proteins c-fos/metabolism
8.
Physiol Res ; 63(4): 483-90, 2014.
Article in English | MEDLINE | ID: mdl-24908095

ABSTRACT

Patients with obesity and type 2 diabetes often display high levels of the anti-diabetic factor fibroblast growth factor-21 (FGF21), suggesting that the overproduction of FGF21 may result from increased adiposity in an attempt by white adipose tissue (WAT) to counteract insulin resistance. However, the production of FGF21 diabetes in the absence of WAT has not been examined. In this study, we investigated the effects of lipodystrophy in A-ZIP F-1 mice on FGF21 production in relation to diabetes. A-ZIP F-1 mice displayed high FGF21 plasma levels resulting from enhanced FGF21 mRNA expression in the liver. Concomitant enhancement of FGF21 receptor (FGFR1) and glucose transporter 1 (GLUT-1) mRNA expression was observed in the muscles of A-ZIP F-1 mice. Furthermore, the activation of hypothalamic NPY and AgRP mRNA expression positively correlated with plasma levels of FGF21 but not active ghrelin. Our study demonstrates that an increased FGF21 plasma level in lipodystrophic A-ZIP F-1 mice results mainly from up-regulated liver production but does not suffice to overcome the lipodystrophy-induced severe type 2-diabetes and insulin resistance in the liver linked to the augmented liver fat deposition.


Subject(s)
Diabetes Mellitus, Experimental/metabolism , Fibroblast Growth Factors/metabolism , Insulin Resistance , Lipodystrophy/metabolism , Liver/metabolism , RNA, Messenger/biosynthesis , Adipose Tissue, Brown/metabolism , Animals , Fibroblast Growth Factors/biosynthesis , Fibroblast Growth Factors/blood , Hypothalamus/metabolism , Mice , Neuropeptides/metabolism , Pancreas/metabolism , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Spleen/metabolism
9.
Mol Cell Endocrinol ; 393(1-2): 120-8, 2014 Aug 05.
Article in English | MEDLINE | ID: mdl-24953973

ABSTRACT

The only peripherally released orexigenic hormone, ghrelin, plays a key role in food intake and body weight regulation. Antagonizing the ghrelin receptor, GHS-R1a, represents a promising approach for anti-obesity therapy. In our study, two novel GHS-R1a antagonists JMV4208 and JMV3002, which are trisubstituted 1,2,4-triazoles, decreased food intake in fasted lean mice in a dose-dependent manner, with ED50 values of 5.25 and 2.05 mg/kg, respectively. Both compounds were stable in mouse blood, with half-lives of 90 min (JMV4208) and 60 min (JMV3002), and disappeared from the blood 8h after administration. Fourteen days of treatment with the ghrelin antagonists (20 mg/kg twice a day) decreased food intake, body weight and adipose tissue mass in mice with diet-induced obesity (DIO). These results are likely attributable to an impact on food intake reduction and an attenuated expression of the lipogenesis-promoting enzymes (acetyl-CoA carboxylase 1 in subcutaneous fat and fatty acid synthase in subcutaneous and intraperitoneal fat). The decrease in fat mass negatively impacted circulating leptin levels. These data suggest that JMV4208 and JMV3002 could be useful therapeutic agents for the treatment of obesity.


Subject(s)
Adipose Tissue/drug effects , Body Weight/drug effects , Eating/drug effects , Picolinic Acids/pharmacology , Receptors, Ghrelin/antagonists & inhibitors , Triazoles/pharmacology , Animals , Dose-Response Relationship, Drug , Mice , Mice, Inbred C57BL , Picolinic Acids/chemistry , Triazoles/chemistry
10.
Physiol Res ; 62(4): 435-44, 2013.
Article in English | MEDLINE | ID: mdl-23590608

ABSTRACT

Ghrelin and agonists of its receptor GHS-R1a are potential substances for the treatment of cachexia. In the present study, we investigated the acute and long term effects of the GHS R1a agonist JMV 1843 (H Aib-DTrp-D-gTrp-CHO) on food intake, body weight and metabolic parameters in lean C57BL/6 male mice. Additionally, we examined stability of JMV 1843 in mouse blood serum. A single subcutaneous injection of JMV 1843 (0.01-10 mg/kg) increased food intake in fed mice in a dose-dependent manner, up to 5-times relative to the saline-treated group (ED(50)=1.94 mg/kg at 250 min). JMV 1843 was stable in mouse serum in vitro for 24 h, but was mostly eliminated from mouse blood after 2 h in vivo. Ten days of treatment with JMV 1843 (subcutaneous administration, 10 or 20 mg/kg/day) significantly increased food intake, body weight and mRNA expression of the orexigenic neuropeptide Y and agouti-related peptide in the medial basal hypothalamus and decreased the expression of uncoupling protein 1 in brown adipose tissue. Our data suggest that JMV 1843 could have possible future uses in the treatment of cachexia.


Subject(s)
Agouti-Related Protein/metabolism , Appetite Stimulants/pharmacology , Eating/drug effects , Ghrelin/agonists , Hypothalamus/drug effects , Neuropeptide Y/metabolism , Oligopeptides/pharmacology , Weight Gain/drug effects , Adipose Tissue, Brown/drug effects , Adipose Tissue, Brown/metabolism , Agouti-Related Protein/genetics , Animals , Appetite Stimulants/administration & dosage , Appetite Stimulants/pharmacokinetics , Dose-Response Relationship, Drug , Ghrelin/metabolism , Hypothalamus/metabolism , Indoles , Injections, Subcutaneous , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/metabolism , Neuropeptide Y/genetics , Oligopeptides/administration & dosage , Oligopeptides/pharmacokinetics , RNA, Messenger/metabolism , Receptors, Ghrelin/agonists , Receptors, Ghrelin/metabolism , Signal Transduction/drug effects , Tryptophan/analogs & derivatives , Uncoupling Protein 1 , Up-Regulation
11.
Neurochem Int ; 59(6): 889-95, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21843570

ABSTRACT

Many peripheral substances, including ghrelin, induce neuronal activation in the brain. In the present study, we compared the effect of subcutaneously administered ghrelin and its three stable agonists: Dpr(3)ghr ([Dpr(N-octanoyl)(3)] ghrelin) (Dpr - diaminopropionic acid), YA GHRP-6 (H-Tyr-Ala-His-DTrp-Ala-Trp-DPhe-Lys-NH(2)), and JMV1843 (H-Aib-DTrp-D-gTrp-CHO) on the Fos expression in food intake-responsive brain areas such as the hypothalamic paraventricular (PVN) and arcuate (ARC) nuclei, the nucleus of the solitary tract (NTS), and area postrema (AP) in male C57BL/6 mice. Immunohistochemical analysis showed that acute subcutaneous dose of each substance (5mg/kg b.w.), which induced a significant food intake increase, elevated Fos protein expression in all brain areas studied. Likewise ghrelin, each agonist tested induced distinct Fos expression overall the PVN. In the ARC, ghrelin and its agonists specifically activated similarly distributed neurons. Fos occurrence extended from the anterior (aARC) to middle (mARC) ARC region. In the latter part of the ARC, the Fos profiles were localized bilaterally, especially in the ventromedial portions of the nucleus. In the NTS, all substances tested also significantly increased the number of Fos profiles in neurons, which also revealed specific location, i.e., in the NTS dorsomedial subnucleus (dmNTS) and the area subpostrema (AsP). In addition, cells located nearby the NTS, in the AP, also revealed a significant increase in number of Fos-activated cells. These results demonstrate for the first time that ghrelin agonists, regardless of their different chemical nature, have a significant and similar activating impact on specific groups of neurons that can be a part of the circuits involved in the food intake regulation. Therefore there is a real potency for ghrelin agonists to treat cachexia and food intake disorders. Thus, likewise JMV1843, the other ghrelin agonists represent substances that might be involved in trials for clinical purposes.


Subject(s)
Appetite Regulation/drug effects , Appetite Regulation/physiology , Brain Chemistry/drug effects , Ghrelin/agonists , Ghrelin/physiology , Neurons/drug effects , Proto-Oncogene Proteins c-fos/biosynthesis , Up-Regulation/drug effects , Animals , Biomarkers/metabolism , Brain Chemistry/physiology , Feeding and Eating Disorders/drug therapy , Feeding and Eating Disorders/physiopathology , Ghrelin/analogs & derivatives , Male , Mice , Mice, Inbred C57BL , Neurons/physiology , Up-Regulation/physiology
12.
Mol Cell Endocrinol ; 343(1-2): 55-62, 2011 Aug 22.
Article in English | MEDLINE | ID: mdl-21704671

ABSTRACT

It was demonstrated that estrogen deficiency and consuming high fat (HF) diet enhanced orexigenic activity of ghrelin. Therefore, we hypothesized that antagonizing of ghrelin action would attenuate food intake and body weight in mice obese both from ovariectomy (OVX) and feeding a HF diet. Ghrelin receptor antagonist [D-Lys(3)]GHRP-6 after seven days of subcutaneous treatment markedly decreased food intake in OVX mice fed both HF and standard diets; furthermore, it reduced body weight and blood glucose, insulin and leptin, and increased ß-hydroxybutyrate level and uncoupling-protein-1 mRNA in brown adipose tissue. Pair-feeding revealed that effect of [D-Lys(3)]GHRP-6 was primary anorexigenic. Estrogen supplementation reduced anorexigenic effects of [D-Lys(3)]GHRP-6. OVX [D-Lys(3)]GHRP-6 treatment in mice on HF diet resulted in markedly increased circulating level and liver expression of a major metabolic regulator, fibroblast growth factor 21. Our data suggest that ghrelin antagonists could be especially beneficial in individuals with common obesity combined with estrogen deficiency.


Subject(s)
Adiposity/drug effects , Diet, High-Fat , Models, Animal , Obesity/physiopathology , Oligopeptides/pharmacology , Postmenopause/metabolism , Receptors, Ghrelin/antagonists & inhibitors , Adipose Tissue, Brown/metabolism , Animals , Behavior, Animal/drug effects , Body Weight/drug effects , Eating/drug effects , Estrogens/administration & dosage , Estrogens/deficiency , Female , Ghrelin/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Ion Channels/genetics , Ion Channels/metabolism , Male , Mice , Mice, Inbred C57BL , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Motor Activity/drug effects , Ovariectomy , PPAR alpha/genetics , PPAR alpha/metabolism , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Uncoupling Protein 1
13.
Horm Metab Res ; 42(3): 182-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20049675

ABSTRACT

Ovariectomized mice on a high fat diet represent a model of diet-induced obesity during estrogen deficiency. Here, we tested the hypothesis that sensitivity to centrally administered leptin in ovariectomized mice with diet-induced obesity could be restored by estrogen supplementation. Ovariectomized C57BL/6 female mice were fed either a standard or high fat diet until they were 27 weeks old. Ovariectomized mice on a high fat diet developed extreme obesity and hyperleptinemia and moderate hyperinsulinemia compared to those on a standard diet. For the last 4 weeks, 17beta-estradiol-3-benzoate or its vehicle was administered subcutaneously in a 4-day cyclic regimen. Finally, leptin or saline was injected into the third ventricle, and food intake and body weight were measured for 36 h. In ovariectomized mice fed a standard diet, the decrease in food intake and body weight was significant and was pronounced in 17beta-estradiol-3-benzoate-supplemented mice. The response to centrally injected leptin in ovariectomized mice on a high fat diet was insignificant, whereas in 17beta-estradiol-3-benzoate-supplemented mice, the effect was significant, particularly with respect to body weight. We showed for the first time that central insensitivity to leptin in ovariectomized diet-induced obese mice was restored with 17beta-estradiol-3-benzoate supplementation, which also attenuated most of the parameters of metabolic syndrome. Only circulating adiponectin, a peripheral insulin sensitivity marker, was lowered following 17beta-estradiol-3-benzoate administration in both high fat and standard diet-fed ovariectomized mice, despite of decreased or unchanged glycemia, respectively.


Subject(s)
Diet , Dietary Fats/pharmacology , Dietary Supplements , Estradiol/pharmacology , Leptin/pharmacology , Ovariectomy , Animals , Body Weight/drug effects , Dietary Fats/administration & dosage , Estradiol/administration & dosage , Feeding Behavior/drug effects , Female , Leptin/administration & dosage , Mice , Mice, Inbred C57BL , Obesity/metabolism , Weight Gain/drug effects
14.
Endocr Regul ; 43(3): 99-106, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19817504

ABSTRACT

OBJECTIVE: Obesity represents a low-grade inflammatory disease and appears a risk factor for insulin resistance, but little is known on whether this may contribute to the development of autoimmune inflammatory diseases. The aim of this work was to study the early-life diet-induced obesity in Lewis rats which are known to be highly susceptible to autoimmunity. METHODS: Obesity was induced by reduced litter size (4 pups per litter) followed by high-fat diet (SHF rats). Control rats (8 pups per litter) were fed with standard diet (CN rats). Oral glucose tolerance test (3 g glucose per kg b.w.) was performed by intra-gastric tube in conscious rats after 12 h fast. Adipocyte size was assessed by light microscope after collagenase digestion. Hypothalamic arcuate (ARC) and paraventricular nuclei (PVN) were isolated by the punching technique. Target mRNAs were quantified by real-time PCR with the use of TaqMan probes and primers. Serum hormones (leptin, ghrelin, adiponectin, visfatin and insulin) were assayed by specific RIAs . RESULTS: During the experimental period SHF rats had the same body weight gain and caloric intake as CN rats. At the age of 8 weeks SHF rats showed increased epididymal fat mass and adipocyte volume, impaired glucose tolerance, normal basal fasting insulin, visfatin, and ghrelin level, but decreased adiponectin and high leptin level. In the ARC, the SHF rats showed increased expression of mRNA for orexigenic neuropeptide Y (NPY), agouti-related protein (AgRP) and anorexigenic pro-inflammatory cytokine IL-6. In the PVN, the SHF rats showed increased expression of mRNA for anorexigenic melanocortin 4 receptor (MC4R) and IL-6. CONCLUSION: Overexpression of orexigenic NPY and AgRP in the ARC indicates leptin resistance in SHF rats. The increased expression of MC4R in PVN points to the activation of melanocortin anorexigenic system which, along with increased hypothalamic IL-6, might prevent the animals from overfeeding. Higher adiposity in these rats results from the high fat-diet composition and not from increased caloric intake. Furthermore, enhanced leptin production appears the main factor indicating the predisposition to autoimmunity in these overfed rats.


Subject(s)
Agouti-Related Protein/genetics , Arcuate Nucleus of Hypothalamus/metabolism , Interleukin-6/genetics , Neuropeptide Y/genetics , Obesity/genetics , Paraventricular Hypothalamic Nucleus/metabolism , Receptor, Melanocortin, Type 4/genetics , Adipocytes, White , Adiponectin/blood , Adipose Tissue, White/cytology , Adiposity , Aging , Agouti-Related Protein/metabolism , Analysis of Variance , Animals , Appetite Regulation/physiology , Area Under Curve , Body Weight , Cell Size , Dietary Fats/administration & dosage , Energy Intake , Feeding Behavior , Gene Expression , Ghrelin/blood , Glucose Intolerance , Insulin/blood , Interleukin-6/metabolism , Leptin/blood , Litter Size , Male , Neuropeptide Y/metabolism , Nicotinamide Phosphoribosyltransferase/blood , Obesity/metabolism , RNA, Messenger/metabolism , Rats , Rats, Inbred Lew , Receptor, Melanocortin, Type 4/metabolism
15.
Physiol Res ; 58(5): 717-723, 2009.
Article in English | MEDLINE | ID: mdl-19093718

ABSTRACT

Monosodium glutamate (MSG) treatment of neonatal mice results in a selective damage to the arcuate nucleus (ARC) and development of obesity with increased adiposity at sustained body weight in the adulthood. Feeding pattern of the MSG obese mice is unusual. Our previous results showed that after 24-h fasting, MSG mice consumed negligible amount of food in several hours and therefore, it was impossible to register the effect of peptides attenuating food intake such as cholecystokinin (CCK) or cocaine- and amphetamine-regulated transcript (CART) peptide. To overcome this problem, two findings were used: firstly, orexigenic effect of neuropeptide Y (NPY) was attenuated both by CCK or CART peptide in lean fed mice and secondly, orexigenic effect of NPY was preserved in fed rats with MSG obesity. In this study, short-term food intake in fed lean and MSG obese C57BL/6 male mice was measured after simultaneous central administration of orexigenic NPY with either CART peptide or peripherally administered CCK. Anorexigenic action of exogenous CART peptide was preserved in MSG obese mice. On the other hand, satiety effect of exogenous CCK was completely lost in MSG obese mice. In conclusion, effective leptin signaling in ARC is necessary for satiety effect of CCK.


Subject(s)
Appetite Regulation , Arcuate Nucleus of Hypothalamus/physiology , Cholecystokinin/physiology , Nerve Tissue Proteins/physiology , Obesity/physiopathology , Animals , Disease Models, Animal , Male , Mice , Mice, Inbred C57BL , Neuropeptide Y/physiology , Obesity/chemically induced , Sodium Glutamate
16.
Physiol Res ; 57(5): 727-734, 2008.
Article in English | MEDLINE | ID: mdl-17949248

ABSTRACT

In this study, susceptibility of inbred C57BL/6 and outbred NMRI mice to monosodium glutamate (MSG) obesity or diet-induced obesity (DIO) was compared in terms of food intake, body weight, adiposity as well as leptin, insulin and glucose levels. MSG obesity is an early-onset obesity resulting from MSG-induced lesions in arcuate nucleus to neonatal mice. Both male and female C57BL/6 and NMRI mice with MSG obesity did not differ in body weight from their lean controls, but had dramatically increased fat to body weight ratio. All MSG obese mice developed severe hyperleptinemia, more remarkable in females, but only NMRI male mice showed massive hyperinsulinemia and an extremely high HOMA index that pointed to development of insulin resistance. Diet-induced obesity is a late-onset obesity; it developed during 16-week-long feeding with high-fat diet containing 60 % calories as fat. Inbred C57BL/6 mice, which are frequently used in DIO studies, both male and female, had significantly increased fat to body weight ratio and leptin and glucose levels compared with their appropriate lean controls, but only female C57BL/6 mice had also significantly elevated body weight and insulin level. NMRI mice were less prone to DIO than C57BL/6 ones and did not show significant changes in metabolic parameters after feeding with high-fat diet.


Subject(s)
Arcuate Nucleus of Hypothalamus/physiopathology , Obesity/physiopathology , Adiposity , Age Factors , Animals , Animals, Newborn , Arcuate Nucleus of Hypothalamus/metabolism , Blood Glucose/metabolism , Body Weight , Dietary Fats , Disease Models, Animal , Disease Susceptibility , Eating , Female , Insulin/blood , Insulin Resistance , Leptin/blood , Male , Mice , Mice, Inbred C57BL , Obesity/etiology , Obesity/metabolism , Phenotype , Sodium Glutamate , Species Specificity
17.
Gen Physiol Biophys ; 25(1): 43-51, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16714774

ABSTRACT

This study was designed to determine particular changes in the renin gene expression and activity in renal cortex and medulla after AT(1) receptor blockade. It was found that two-week-treatment with AT(1) blocker losartan induced an increase in tissue renin activity in both parts of kidney causing subsequent elevation of plasma renin activity. Renin mRNA in losartan-treated rats was increased only in cortex, suggesting cortex origin of elevated renin activity in medulla. Medullary renin mRNA indicated local synthesis of renin within the whole kidney and supported the idea of the presence of tissue renin-angiotensin system. Our results show that gene expression of renin in kidney medulla is insensitive to AT(1) receptor blockade and this points out that the regulation of kidney renin-angiotensin system probably differs from that in cortex.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Gene Expression Regulation , Kidney Medulla/drug effects , Losartan/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Renin/genetics , Animals , Blood Pressure , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Kidney Medulla/metabolism , Male , Polymerase Chain Reaction , RNA, Messenger/metabolism , Radioimmunoassay , Rats , Rats, Inbred WKY , Renin/blood
18.
Physiol Res ; 53(6): 603-7, 2004.
Article in English | MEDLINE | ID: mdl-15588127

ABSTRACT

Cardiovascular effects of LVV-hemorphin-7, a member of the family of fragments from beta-chain of human or bovine hemoglobin, were studied in conscious spontaneously hypertensive (SHR) and Wistar-Kyoto (WKY) rats by radiotelemetry. Intraperitoneal injection of hemorphin in a dose of 100 microg/kg significantly decreased blood pressure in SHR, whereas negligible effect was seen in normotensive WKY rats. Blood pressure changes were accompanied by reduction of heart rate. In conclusion, a direct effect of LVV-hemorphin-7 on blood pressure was demonstrated in SHR. These biologically active peptides could be involved in blood pressure regulation especially in hypertensive rats, but the precise mechanism should be elucidated.


Subject(s)
Blood Pressure/drug effects , Heart Rate/drug effects , Hemoglobins/administration & dosage , Hypertension/physiopathology , Peptide Fragments/administration & dosage , Animals , Male , Rats , Rats, Inbred WKY , Rats, Wistar , Telemetry/methods
19.
Reproduction ; 127(5): 581-5, 2004 May.
Article in English | MEDLINE | ID: mdl-15129013

ABSTRACT

The immunosuppressive fraction (ISF) of boar seminal vesicle fluid was recently demonstrated to inhibit production of T helper (Th)1 cytokines and enhance production of Th2 cytokines. The present study shows the effect of the ISF on leptin concentrations in blood plasma and adipose tissue in mice during pregnancy. The ISF effect on thymus weight during pregnancy is also demonstrated. The leptin concentration in blood plasma and adipose tissue increased and remained high in the latter half of pregnancy. ISF treatment at the beginning of pregnancy significantly lowered the leptin concentration both in blood plasma and adipose tissue of pregnant mice. Thymus involution has been described previously in pregnant mice. ISF treatment compensated for the loss of thymus mass during the whole pregnancy in the ISF-treated mice. The treatment of pregnant mice with ISF did not affect pregnancy and litter size.


Subject(s)
Leptin/metabolism , Pregnancy, Animal/metabolism , Seminal Plasma Proteins/pharmacology , Thymus Gland/anatomy & histology , Adipose Tissue/chemistry , Animals , Female , Leptin/analysis , Leptin/blood , Male , Mice , Mice, Inbred BALB C , Organ Size/drug effects , Pregnancy , Semen/chemistry , Seminal Plasma Proteins/isolation & purification , Seminal Vesicles/metabolism , Swine
20.
Am J Reprod Immunol ; 49(4): 249-54, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12852499

ABSTRACT

PROBLEM: The immunosuppressive fraction (ISF) of boar seminal vesicle fluid has recently been demonstrated to inhibit mitogen-stimulated proliferation of lymphocytes and antibody response to corpuscular and soluble antigens. The effects of ISF on in vitro and in vivo production of cytokines as well as its possible inhibitory effect on proliferation of B lymphoma cells remain to be elucidated. METHODS: The effect of ISF on proliferation of normal mouse spleen cells stimulated by Concanavalin A (Con A) and on mouse B lymphoma cells was measured by 3H-thymidine incorporation. Cytokines were determined in the supernatants of mouse spleen cells stimulated with Con A in the presence or absence of ISF by enzyme-linked immunosorbent assay (ELISA). In vivo cytokine production in the sera samples of mice treated with ISF and immunized with keyhole limpet hemocyanin (KLH) was followed by ELISA, too. RESULTS: We confirmed the inhibitory effect of ISF on Con A-stimulated lymphocyte proliferation. ISF affected cytokine production in the Con A-stimulated spleen cells: production of interleukin-2 (IL-2) and interferon-gamma (IFN-gamma) was lowered, but production of IL-4, IL-6, and IL-10 was enhanced. Similarly, in the sera samples of mice immunized with keyhole limpet hemocyanin (KLH), IL-2 and IFN-gamma levels were decreased by ISF. ISF inhibited proliferation of Ag 8 and X 63-IL-2 B lymphoma cells as well. CONCLUSIONS: ISF inhibited production of T helper1 (Th1) cytokines (IL-2 and IFN-gamma) and enhanced production of Th2 cytokines (IL-4, IL-6, and IL-10). ISF seems to shift the Th1/Th2 pattern in favor of Th2. ISF exhibited an antiproliferative activity on mouse B lymphoma cells.


Subject(s)
Cytokines/biosynthesis , Growth Substances/pharmacology , Lymphocytes/metabolism , Proton Pumps , Semen/chemistry , Adjuvants, Immunologic/pharmacology , Animals , Cell Division/drug effects , Cell Line, Tumor , Concanavalin A/pharmacology , Cytokines/blood , Cytokines/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Growth Substances/immunology , Hemocyanins/pharmacology , Lymphocytes/drug effects , Lymphoma, B-Cell/metabolism , Mice , Proton-Translocating ATPases , Sus scrofa
SELECTION OF CITATIONS
SEARCH DETAIL
...