Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Int J Mol Sci ; 20(20)2019 Oct 10.
Article in English | MEDLINE | ID: mdl-31658598

ABSTRACT

Bisphenol A (BPA), a known endocrine disrupting chemical (EDC), was administered by diet to gilthead sea bream (Sparus aurata) in order to study its effects on the endocannabinoid system (ECS) and gonadal steroidogenesis. 2-year-old male gilthead sea bream were fed with two different concentrations of BPA (LOW at 4 and HIGH at 4000 µg/kg body weight for 21 days during the reproductive season. Exposure to 4000 µg BPA/kg bw/day (BPA HIGH) reduced sperm motility and altered the straight-line velocity (VSL) and linearity (LIN). Effects on steroidogenesis were evident, with testosterone (T) being up-regulated by both treatments and 11-ketotestosterone (11-KT) down-regulated by BPA HIGH. Plasma levels of 17ß-estradiol (E2) were not affected. The Gonadosomatic Index (GSI) increased in the BPA HIGH group. Interestingly, the levels of endocannabinoids and endocannabinoid-like compounds were significantly reduced after both treatments. Unpredictably, a few changes were noticed in the expression of genes coding for ECS enzymes, while the receptors were up-regulated depending on the BPA dose. Reproductive markers in testis (leptin receptor (lepr), estrogen receptors (era, erb), progesterone receptors (pr) and the gonadotropin releasing hormone receptor (gnrhr)) were up-regulated. BPA induced the up-regulation of the hepatic genes involved in oogenesis (vitellogenin (vtg) and zona pellucida 1 (zp1)).


Subject(s)
Benzhydryl Compounds/pharmacology , Diet , Phenols/pharmacology , Reproduction/drug effects , Sea Bream/growth & development , Testis/drug effects , Testis/metabolism , Animal Feed , Animals , Body Weight , Endocannabinoids/genetics , Endocrine Disruptors/pharmacology , Estradiol/blood , Gene Expression Regulation , Gonadal Steroid Hormones/blood , Gonads/drug effects , Gonads/pathology , Lipogenesis/drug effects , Liver/metabolism , Male , Models, Animal , Receptors, Leptin/drug effects , Sperm Motility/drug effects , Spermatozoa/drug effects , Testis/pathology , Testosterone/analogs & derivatives , Testosterone/blood , Testosterone/metabolism , Testosterone/pharmacology , Transcriptome , Up-Regulation , Vitellogenins/genetics , Vitellogenins/metabolism , Zona Pellucida/metabolism
2.
Am J Physiol Endocrinol Metab ; 317(4): E586-E596, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31361549

ABSTRACT

Leptin administration into the hindbrain, and specifically the nucleus of the solitary tract, increases phosphorylated signal transducer and activator of transcription 3 (pSTAT3), a marker of leptin receptor activation, in hypothalamic nuclei known to express leptin receptors. The ventromedial nucleus of the hypothalamus (VMH) shows the greatest response, with a threefold increase in pSTAT3. This experiment tested the importance of VMH leptin receptor-expressing neurons in mediating weight loss caused by fourth ventricle (4V) leptin infusion. Male Sprague-Dawley rats received bilateral VMH 75-nL injections of 260 ng/µL of leptin-conjugated saporin (Lep-Sap) or blank-saporin (Blk-Sap). After 23 days they were fitted with 4V infusion cannulas and 1 wk later adapted to housing in a calorimeter before they were infused with 0.9 µg leptin/day for 14 days. There was no effect of VMH Lep-Sap on weight gain or glucose clearance before leptin infusion. Leptin inhibited food intake and respiratory exchange ratio in Blk-Sap but not Lep-Sap rats. Leptin had no effect on energy expenditure or brown adipose tissue temperature of either group. Inguinal and epididymal fat were significantly reduced in leptin-treated Blk-Sap rats, but the response was greatly attenuated in Lep-Sap rats. VMH pSTAT3 was increased in leptin-treated Blk-Sap but not Lep-Sap rats. These results support the concept that leptin-induced weight loss results from an integrated response across different brain areas. They also support previous reports that VMH leptin receptors do not play a significant role in maintaining energy balance in basal conditions but limit weight gain during positive energy balance.


Subject(s)
Fourth Ventricle , Leptin/administration & dosage , Leptin/pharmacology , Neurons/drug effects , Neurons/metabolism , Receptors, Leptin/drug effects , Receptors, Leptin/metabolism , Ventromedial Hypothalamic Nucleus/metabolism , Weight Loss/drug effects , Adipose Tissue/drug effects , Adipose Tissue/growth & development , Animals , Body Temperature/drug effects , Eating/drug effects , Glucose/metabolism , Infusions, Intraventricular , Male , Rats , Rats, Sprague-Dawley , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/genetics , Saporins/pharmacology , Ventromedial Hypothalamic Nucleus/drug effects
3.
Neuroendocrinology ; 108(4): 278-290, 2019.
Article in English | MEDLINE | ID: mdl-30572328

ABSTRACT

BACKGROUND: Increased fructose consumption and chronic exposure to stress have been associated with the development of obesity and insulin resistance. In the hypothalamus, a crossroad of stress responses and energy balance, insulin and glucocorticoids regulate the expression of orexigenic neuropeptides, neuropeptide Y (NPY) and agouti-related protein (AgRP), and anorexigenic neuropeptides, proopio-melanocortin (POMC) and cocaine- and amphetamine-regulated transcript (CART). OBJECTIVES: We investigated whether chronic stress and fructose diet disrupt these hormonal signaling pathways and appetite control in the hypothalamus, contributing to the development of insulin resistance and obesity. Potential roles of hypothalamic inflammation and oxidative stress in the development of insulin resistance were also analyzed. METHODS: Insulin, glucocorticoid, and leptin signaling, expression of orexigenic and anorexigenic neuropeptides, and antioxidative and inflammatory statuses in the whole hypothalamus of fructose-fed female rats exposed to unpredictable stress for 9 weeks were analyzed using quantitative PCR and Western blotting. RESULTS: Chronic stress combined with a fructose-enriched diet reduced protein content and stimulatory phosphorylation of Akt kinase, and elevated 11ß-hydroxysteroid dehydrogenase 1 and glucocorticoid receptor expression, while alterations in appetite regulation (NPY, AgRP, POMC, CART, leptin receptor, and SOCS3 expression) were not observed. The expression of antioxidative defense enzymes (mitochondrial manganese superoxide dismutase 2, glutathione reductase, and catalase) and proinflammatory cytokines (IL-1ß, IL-6, and TNFα) was reduced. CONCLUSIONS: Our results underline the combination of long-term stress exposure and fructose overconsumption as more detrimental for hypothalamic function than for either of the factors separately, as it enhanced glucocorticoid and impaired insulin signaling, antioxidative -defense, and inflammatory responses of this homeostasis- regulating center.


Subject(s)
Antioxidants/pharmacology , Energy Metabolism/drug effects , Fructose/metabolism , Hypothalamus/metabolism , Animal Feed , Animals , Antioxidants/metabolism , Appetite Regulation/drug effects , Appetite Regulation/physiology , Diet , Energy Metabolism/physiology , Female , Insulin/metabolism , Leptin/metabolism , Neuropeptide Y/metabolism , Neuropeptides/metabolism , Obesity/drug therapy , Obesity/metabolism , Rats, Wistar , Receptors, Leptin/drug effects , Receptors, Leptin/metabolism , Stress, Physiological
4.
Acta Physiol (Oxf) ; 226(2): e13244, 2019 06.
Article in English | MEDLINE | ID: mdl-30589509

ABSTRACT

AIM: We aimed to investigate whether a dysregulated maternal diet during gestation and lactation induces long-lasting changes in the hypothalamic control of feeding behavior in the offspring and whether this effect is sex specific. METHODS: The study included an analysis of appetite-regulating metabolic hormones and hypothalamic signaling in male and female offspring in adulthood after exposure to a free-choice high-calorie palatable low-protein (P) diet or standard chow (C) during (pre)gestation/lactation (maternal) and/or postweaning (offspring). RESULTS: Maternal exposure to the P diet resulted in decreased protein intake and body weight gain in dams and decreased body weight gain in offspring during lactation. The maternal P diet (PC) specifically increased feed efficacy and decreased body weight and cholesterol levels in the female offspring in adulthood, but no changes in adiposity or leptin levels were found. In contrast, P diet exposure after weaning (CP and PP) increased caloric intake, adiposity and circulating levels of leptin in the male and female offspring in adulthood. The hypothalami of the female offspring exposed to the maternal P diet (PC and PP) expressed high levels of the phospho-leptin receptor and low levels of SOCS3, phospho-IRS1 and phospho-AMPK, regardless of the postweaning diet. The hypothalami of the female rats in the PC group also showed increased levels of STAT3 and the orexigenic neuropeptide Agrp. CONCLUSIONS: Maternal exposure to a free-choice high-calorie low-protein diet induces a long-term feed efficacy associated with changes in leptin signaling through IRS-1 and AMPK dephosphorylation in the hypothalami of female offspring in adulthood.


Subject(s)
Behavior, Animal/drug effects , Diet, Protein-Restricted/adverse effects , Leptin/pharmacology , Prenatal Exposure Delayed Effects , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Animals , Behavior, Animal/physiology , Feeding Behavior/drug effects , Feeding Behavior/physiology , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Leptin/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/metabolism , Rats, Wistar , Receptors, Leptin/drug effects , Signal Transduction/drug effects
5.
Am J Physiol Regul Integr Comp Physiol ; 315(4): R856-R865, 2018 10 01.
Article in English | MEDLINE | ID: mdl-30133304

ABSTRACT

The pancreatic hormone amylin is released from beta cells following nutrient ingestion and contributes to the control of body weight and glucose homeostasis. Amylin reduces food intake by activating neurons in the area postrema (AP). Amylin was also shown to synergize with the adipokine leptin, with combination therapy producing greater weight loss and food intake reduction than either hormone alone. Although amylin and leptin were initially thought to interact downstream of the AP in the hypothalamus, recent findings show that the two hormones can act on the same AP neurons, suggesting a more direct relationship. The objective of this study was to determine whether amylin action depends on functional leptin signaling. We tested the ability of amylin to induce satiation and to activate its primary target neurons in the AP in two rodent models of LepR deficiency, the db/db mouse and the Zucker diabetic fatty (ZDF) rat. When compared with wild-type (WT) mice, db/db mice exhibited reduced amylin-induced satiation, reduced amylin-induced Fos in the AP, and a lower expression of calcitonin receptor (CTR) protein, the core component of all amylin receptors. ZDF rats also showed no reduction in food intake following amylin treatment; however, unlike the db/db mice, levels of amylin-induced Fos and CTR in the AP were no different than WT rats. Our results suggest that LepR expression is required for the full anorexic effect of amylin; however, the neuronal activation in the AP seems to depend on the type of LepR mutation.


Subject(s)
Amylin Receptor Agonists/pharmacology , Appetite Depressants/pharmacology , Area Postrema/drug effects , Feeding Behavior/drug effects , Islet Amyloid Polypeptide/pharmacology , Leptin/metabolism , Receptors, Leptin/metabolism , Satiety Response/drug effects , Animals , Area Postrema/metabolism , Genotype , Male , Mutation , Phenotype , Proto-Oncogene Proteins c-fos/metabolism , Rats, Zucker , Receptors, Calcitonin/agonists , Receptors, Calcitonin/metabolism , Receptors, Islet Amyloid Polypeptide/drug effects , Receptors, Islet Amyloid Polypeptide/metabolism , Receptors, Leptin/deficiency , Receptors, Leptin/drug effects , Receptors, Leptin/genetics , Signal Transduction/drug effects
6.
Mol Metab ; 6(10): 1113-1125, 2017 10.
Article in English | MEDLINE | ID: mdl-29031713

ABSTRACT

OBJECTIVE: In visceral obesity, an overactive endocannabinoid/CB1 receptor (CB1R) system promotes increased caloric intake and decreases energy expenditure, which are mitigated by global or peripheral CB1R blockade. In mice with diet-induced obesity (DIO), inhibition of food intake by the peripherally restricted CB1R antagonist JD5037 could be attributed to endogenous leptin due to the rapid reversal of hyperleptinemia that maintains leptin resistance, but the signaling pathway engaged by leptin has remained to be determined. METHODS: We analyzed the hypothalamic circuitry targeted by leptin following chronic treatment of DIO mice with JD5037. RESULTS: Leptin treatment or an increase in endogenous leptin following fasting/refeeding induced STAT3 phosphorylation in neurons in the arcuate nucleus (ARC) in lean and JD5037-treated DIO mice, but not in vehicle-treated DIO animals. Co-localization of pSTAT3 in leptin-treated mice was significantly less common with NPY+ than with POMC+ ARC neurons. The hypophagic effect of JD5037 was absent in melanocortin-4 receptor (MC4R) deficient obese mice or DIO mice treated with a MC4R antagonist, but was maintained in NPY-/- mice kept on a high-fat diet. CONCLUSIONS: Peripheral CB1R blockade in DIO restores sensitivity to endogenous leptin, which elicits hypophagia via the re-activation of melanocortin signaling in the ARC.


Subject(s)
Leptin/metabolism , Receptor, Cannabinoid, CB1/metabolism , Receptors, Leptin/drug effects , Animals , Arcuate Nucleus of Hypothalamus/metabolism , Body Weight/physiology , Cannabinoids/metabolism , Diet, High-Fat , Dietary Fats/metabolism , Eating/physiology , Hypothalamus/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Obese , Neuropeptide Y/metabolism , Obesity/metabolism , Pro-Opiomelanocortin/metabolism , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/physiology , Receptor, Melanocortin, Type 4/metabolism , Receptors, Cannabinoid/metabolism , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Sulfonamides/pharmacology
7.
Transl Psychiatry ; 5: e600, 2015 Jul 14.
Article in English | MEDLINE | ID: mdl-26171981

ABSTRACT

Epigenetics may have an important role in mood stabilizer action. Valproic acid (VPA) is a histone deacetylase inhibitor, and lithium (Li) may have downstream epigenetic actions. To identify genes commonly affected by both mood stabilizers and to assess potential epigenetic mechanisms that may be involved in their mechanism of action, we administered Li (N = 12), VPA (N = 12), and normal chow (N = 12) to Brown Norway rats for 30 days. Genomic DNA and mRNA were extracted from the hippocampus. We used the mRNA to perform gene expression analysis on Affymetrix microarray chips, and for genes commonly regulated by both Li and VPA, we validated expression levels using quantitative real-time PCR. To identify potential mechanisms underlying expression changes, genomic DNA was bisulfite treated for pyrosequencing of key CpG island 'shores' and promoter regions, and chromatin was prepared from both hippocampal tissue and a hippocampal-derived cell line to assess modifications of histones. For most genes, we found little evidence of DNA methylation changes in response to the medications. However, we detected histone H3 methylation and acetylation in the leptin receptor gene, Lepr, following treatment with both drugs. VPA-mediated effects on histones are well established, whereas the Li effects constitute a novel mechanism of transcriptional derepression for this drug. These data support several shared transcriptional targets of Li and VPA, and provide evidence suggesting leptin signaling as an epigenetic target of two mood stabilizers. Additional work could help clarify whether leptin signaling in the brain has a role in the therapeutic action of Li and VPA in bipolar disorder.


Subject(s)
Epigenesis, Genetic/drug effects , Lithium Compounds/pharmacology , Receptors, Leptin/drug effects , Valproic Acid/pharmacology , Animals , DNA Methylation/drug effects , Gene Expression Regulation/drug effects , Genes/drug effects , Male , Oligonucleotide Array Sequence Analysis , Rats , Rats, Inbred BN , Real-Time Polymerase Chain Reaction , Receptors, Leptin/genetics , Transcriptome
8.
Br J Pharmacol ; 172(12): 2974-90, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25653112

ABSTRACT

BACKGROUND AND PURPOSE: Leptin, an adipokine synthesized by the placenta during pregnancy, has been proposed for the management of preterm labour (PTL), as it is able to prevent in vitro uterine contractility and remodelling associated with labour onset. Another common feature of labour onset is the phenotypic switch of myometrial smooth muscle cells from a proliferative to a hypertrophic state. As proliferative effects have been demonstrated for leptin in other tissues, we aimed to investigate its ability to induce myometrial proliferation and thus to maintain uterine quiescence. EXPERIMENTAL APPROACH: We stimulated human primary myometrial smooth muscle cells with leptin in the presence or absence of receptor antagonists or signalling pathway inhibitors. KEY RESULTS: Leptin induced myometrial cell proliferation in a biphasic manner. At 6.25 ng · mL(-1), leptin-induced proliferation was mediated by the leptin receptor and required the early activation of ERK1/2. At a concentration above 25 ng · mL(-1), leptin induced direct non-specific stimulation of the IL-6 receptor, leading to NF-κB activation, and exerted anti-proliferative effects. However, at 50 ng · mL(-1), leptin re-induces proliferation via IL-6 receptor stimulation that requires STAT3 and delayed ERK1/2 activation. CONCLUSIONS AND IMPLICATIONS: These data bring new insights into leptin signalling-induced myometrial proliferation and its interrelationship with the IL-6/IL-6 receptor axis. In the light of our previous work, the present study emphasizes the potential value of leptin in the pharmacological management of PTL and it also strengthens the hypothesis that leptin might be a contributory factor in the parturition-related disorders observed in obese women.


Subject(s)
Cell Proliferation/drug effects , Leptin/pharmacology , Myometrium/drug effects , Receptors, Leptin/drug effects , Dose-Response Relationship, Drug , Female , Humans , Interleukin-6/metabolism , Leptin/administration & dosage , Leptin/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myometrium/cytology , NF-kappa B/metabolism , Pregnancy , Receptors, Leptin/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
9.
Rheumatology (Oxford) ; 54(6): 1087-92, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25389358

ABSTRACT

OBJECTIVE: Glucocorticoids are powerful anti-inflammatory compounds that also induce the expression of leptin and leptin receptor (Ob-R) in synovial fibroblasts through TGF-ßsignalling and Smad1/5 phosphorylation. Compound A (CpdA), a selective glucocorticoid receptor agonist, reduces inflammation in murine arthritis models and does not induce diabetes or osteoporosis, thus offering an improved risk:benefit ratio in comparison with glucocorticoids. Due to the detrimental role of leptin in OA pathogenesis, we sought to determine whether CpdA also induced leptin and Ob-R protein expression as observed with prednisolone. METHODS: Human synovial fibroblasts and chondrocytes were isolated from the synovium and cartilage of OA patients after joint surgery. The cells were treated with prednisolone, TGF-ß1, TNF-α and/or CpdA. Levels of leptin, IL-6, IL-8, MMP-1 and MMP-3 were measured by ELISA and expression levels of Ob-R phospho-Smad1/5, phospho-Smad2, α-tubulin and glyceraldehyde 3-phosphate dehydrogenase were analysed by western blotting. RESULTS: CpdA, unlike prednisolone, did not induce leptin secretion or Ob-R protein expression in OA synovial fibroblasts. Moreover, CpdA decreased endogenous Ob-R expression and down-regulated prednisolone-induced leptin secretion and Ob-R expression. Mechanistically, CpdA, unlike prednisolone, did not induce Smad1/5 phosphorylation. CpdA, similarly to prednisolone, down-regulated endogenous and TNF-α-induced IL-6, IL-8, MMP-1 and MMP-3 protein secretion. The dissociative effect of CpdA was confirmed using chondrocytes with no induction of leptin secretion, but with a significant decrease in IL-6, IL-8, MMP-1 and MMP-3 protein secretion. CONCLUSION: CpdA, unlike prednisolone, did not induce leptin or Ob-R in human OA synovial fibroblasts, thereby demonstrating an improved risk:benefit ratio.


Subject(s)
Chondrocytes/metabolism , Fibroblasts/metabolism , Osteoarthritis/metabolism , Prednisolone/pharmacology , Receptors, Glucocorticoid/agonists , Synovial Membrane/metabolism , Aged , Aged, 80 and over , Blotting, Western , Chondrocytes/drug effects , Enzyme-Linked Immunosorbent Assay , Female , Fibroblasts/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/drug effects , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , Humans , Interleukin-6/metabolism , Interleukin-8/drug effects , Interleukin-8/metabolism , Leptin/metabolism , Male , Matrix Metalloproteinase 1/drug effects , Matrix Metalloproteinase 1/metabolism , Matrix Metalloproteinase 3/drug effects , Matrix Metalloproteinase 3/metabolism , Middle Aged , Receptors, Leptin/drug effects , Receptors, Leptin/metabolism , Smad Proteins, Receptor-Regulated/drug effects , Smad Proteins, Receptor-Regulated/metabolism , Synovial Membrane/drug effects , Transforming Growth Factor beta1/drug effects , Transforming Growth Factor beta1/metabolism , Tubulin/drug effects , Tubulin/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
10.
World J Gastroenterol ; 20(42): 15727-35, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25400456

ABSTRACT

AIM: To investigate the effect of GW4064 on the expression of adipokines and their receptors during differentiation of 3T3-L1 preadipocytes and in HepG2 cells. METHODS: The mRNA expression of farnesoid X receptor (FXR), peroxisome proliferator-activated receptor-gamma 2 (PPAR-γ2), adiponectin, leptin, resistin, adiponectin receptor 1 (AdipoR1), adiponectin receptor 2 (AdipoR2), and the long isoform of leptin receptor (OB-Rb) and protein levels of adiponectin, leptin, and resistin were determined using fluorescent real-time PCR and enzyme linked immunosorbent assay, respectively, on days 0, 2, 4, 6, and 8 during the differentiation of 3T3-L1 preadipocytes exposed to GW4064. Moreover, mRNA expression of AdipoR2 and OB-Rb was also examined using fluorescent real-time PCR at 0, 12, 24, and 48 h in HepG2 cells treated with GW4064. RESULTS: The mRNA expression of FXR, PPAR-γ2, adiponectin, leptin, resistin, AdipoR1, AdipoR2, and OB-Rb and protein levels of adiponectin, leptin, and resistin increased along with differentiation of 3T3-L1 preadipocytes (P < 0.05 for all). The mRNA expression of FXR, PPAR-γ2, adiponectin, leptin, and AdipoR2 in 3T3-L1 preadipocytes, and AdipoR2 and OB-Rb in HepG2 cells was significantly increased after treatment with GW4064, when compared with the control group (P < 0.05 for all). A similar trend was observed for protein levels of adipokines (including adiponectin, leptin and resistin). However, the expression of resistin, AdipoR1, and OB-Rb in 3T3-L1 cells did not change after treatment with GW4064. CONCLUSION: The FXR agonist through regulating, at least partially, the expression of adipokines and their receptors could offer an innovative way for counteracting the progress of metabolic diseases such as nonalcoholic fatty liver disease.


Subject(s)
Adipocytes/drug effects , Adipokines/metabolism , Hepatocytes/drug effects , Isoxazoles/pharmacology , Receptors, Cytoplasmic and Nuclear/agonists , 3T3-L1 Cells , Adipocytes/metabolism , Adipokines/genetics , Animals , Hep G2 Cells , Hepatocytes/metabolism , Humans , Mice , PPAR gamma/drug effects , PPAR gamma/genetics , PPAR gamma/metabolism , RNA, Messenger/metabolism , Receptors, Adiponectin/drug effects , Receptors, Adiponectin/genetics , Receptors, Adiponectin/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Leptin/drug effects , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Signal Transduction/drug effects , Time Factors , Up-Regulation
11.
Am J Physiol Regul Integr Comp Physiol ; 307(11): R1338-44, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25298514

ABSTRACT

Pontine parabrachial nucleus (PBN) neurons integrate visceral, oral, and other sensory information, playing an integral role in the neural control of feeding. Current experiments probed whether lateral PBN (lPBN) leptin receptor (LepRb) signaling contributes to this function. Intra-lPBN leptin microinjection significantly reduced cumulative chow intake, average meal size, and body weight in rats, independent of effects on locomotor activity or gastric emptying. In contrast to the effects observed following LepRb activation in other nuclei, lPBN LepRb stimulation did not affect progressive ratio responding for sucrose reward or conditioned place preference for a palatable food. Collectively, results suggest that lPBN LepRb activation reduces food intake by modulating the neural processing of meal size/satiation signaling, and highlight the lPBN as a novel site of action for leptin-mediated food intake control.


Subject(s)
Eating/physiology , Parabrachial Nucleus/physiology , Receptors, Leptin/physiology , Signal Transduction/physiology , Animals , Body Weight/drug effects , Cerebral Aqueduct/drug effects , Diet, High-Fat , Eating/drug effects , Food Preferences/drug effects , Gastric Emptying/drug effects , Leptin/administration & dosage , Leptin/pharmacology , Male , Parabrachial Nucleus/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Leptin/drug effects , Satiation/drug effects , Signal Transduction/drug effects
12.
Toxicol Appl Pharmacol ; 279(3): 401-408, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-24978599

ABSTRACT

Non-dioxin-like polychlorinated biphenyls (NDL-PCBs) are highly lipophilic environmental contaminants that accumulate in lipid-rich tissues, such as adipose tissue. Here, we reported the effects induced by PCBs 101, 153 and 180, three of the six NDL-PCBs defined as indicators, on mature 3T3-L1 adipocytes. We observed an increase in lipid content, in leptin gene expression and a reduction of leptin receptor expression and signaling, when cells were exposed to PCBs, alone or in combination. These modifications were consistent with the occurrence of "leptin-resistance" in adipose tissue, a typical metabolic alteration related to obesity. Therefore, we investigated how PCBs affect the expression of pivotal proteins involved in the signaling of leptin receptor. We evaluated the PCB effect on the intracellular pathway JAK/STAT, determining the phosphorylation of STAT3, a downstream activator of the transcription of leptin gene targets, and the expression of SOCS3 and PTP1B, two important regulators of leptin resistance. In particular, PCBs 153 and 180 or all PCB combinations induced a significant reduction in pSTAT3/STAT3 ratio and an increase in PTP1B and SOCS3, evidencing an additive effect. The impairment of leptin signaling was associated with the reduction of AMPK/ACC pathway activation, leading to the increase in lipid content. These pollutants were also able to increase the transcription of inflammatory cytokines (IL-6 and TNFα). It is worthy to note that the PCB concentrations used are comparable to levels detectable in human adipose tissue. Our data strongly support the hypothesis that NDL-PCBs may interfere with the lipid metabolism contributing to the development of obesity and related diseases.


Subject(s)
Adipocytes/drug effects , Environmental Pollutants/toxicity , Leptin/physiology , Lipid Metabolism/drug effects , Polychlorinated Biphenyls/toxicity , Signal Transduction/drug effects , 3T3-L1 Cells , Animals , Azo Compounds , Blotting, Western , Cell Differentiation , Coloring Agents , Interleukin-6/biosynthesis , Interleukin-6/genetics , Mice , Mitogen-Activated Protein Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/biosynthesis , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Real-Time Polymerase Chain Reaction , Receptors, Leptin/biosynthesis , Receptors, Leptin/drug effects , Receptors, Leptin/genetics , STAT3 Transcription Factor/biosynthesis , STAT3 Transcription Factor/genetics , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/biosynthesis , Suppressor of Cytokine Signaling Proteins/genetics , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/genetics
13.
Cell Mol Neurobiol ; 34(6): 871-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24794794

ABSTRACT

Leptin is a multifunctional hormone produced by the ob gene and is secreted by adipocytes that regulate food intake and energy metabolism. Numerous studies demonstrated that leptin is a novel neuroprotective effector, however, the mechanisms are largely unknown. Herein, we demonstrate the protective activities of leptin after ischemic stroke and provide the first evidence for the involvement of the connexin 43 (Cx43) in leptin-mediated neuroprotection. We found that leptin treatment reduces the infarct volume, improves animal behavioral parameters, and inhibits the elevation of Cx43 expression in vivo. In vitro, leptin reverses ischemia-induced SY5Y and U87 cells Cx43 elevation, secreted glutamate levels in medium and SY5Y cell death, these roles could be abolished by leptin receptor blocker. Additionally, leptin administration upregulated the extracellular signal-regulated kinase1/2 (ERK1/2) phosphorylation. Moreover, ERK1/2 inhibitors pretreatment reversed the effects of leptin on Cx43 expression, glutamate levels and cell apoptosis. In conclusion, the present study demonstrated that leptin can reduce the Cx43 expression and cell death both in vivo and in vitro via ERK1/2 signaling pathway. This result provides a novel regulatory signaling pathway of the neuroprotective effects of leptin and may contribute to ischemic brain injury prevention and therapy.


Subject(s)
Brain Ischemia/drug therapy , Brain/drug effects , Connexin 43/metabolism , Leptin/pharmacology , MAP Kinase Signaling System/drug effects , Neuroprotective Agents/pharmacology , Animals , Brain/metabolism , Brain Ischemia/metabolism , Male , Mice , Neurons/metabolism , Receptors, Leptin/drug effects
14.
Metallomics ; 6(7): 1229-39, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24793162

ABSTRACT

Protein tyrosine phosphatases (PTPs) are key enzymes in cellular regulation. The 107 human PTPs are regulated by redox signalling, phosphorylation, dimerisation, and proteolysis. Recent findings of very strong inhibition of some PTPs by zinc ions at concentrations relevant in a cellular environment suggest yet another mechanism of regulation. One of the most extensively investigated PTPs is PTP1B (PTPN1). It regulates the insulin and leptin signalling pathway and is implicated in cancer and obesity/diabetes. The development of novel assay conditions to investigate zinc inhibition of PTP1B provides estimates of about 5.6 nM affinity for inhibitory zinc(II) ions. Analysis of three PTP1B 3D structures (PDB id: 2CM2, 3I80 and 1A5Y) identified putative zinc binding sites and supports the kinetic studies in suggesting an inhibitory zinc only in the closed and cysteinyl-phosphate intermediate forms of the enzyme. These observations gain significance with regard to recent findings of regulatory roles of zinc ions released from the endoplasmic reticulum.


Subject(s)
Enzyme Inhibitors/pharmacology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Zinc/metabolism , Binding Sites , Edetic Acid/pharmacology , Humans , Molecular Docking Simulation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Receptor, Insulin/drug effects , Receptors, Leptin/drug effects , Zinc/administration & dosage
15.
Curr Pharm Des ; 20(1): 136-45, 2014.
Article in English | MEDLINE | ID: mdl-24180400

ABSTRACT

The OB-receptor or leptin receptor (LR) is crucial for energy homeostasis and regulation of food uptake. Leptin is a 16 kDa hormone that is mainly secreted by fat cells into the bloodstream. Under normal circumstances, circulating leptin levels are proportionate to the fat body mass. Sensing of elevated leptin levels by the hypothalamic neuro-circuitry activates a negative feedback loop resulting in reduced food intake and increased energy expenditure. Decreased leptin concentrations lead to opposite effects. Therefore, rational design of leptin agonists/antagonists could be an appealing challenge in the battle against obesity. The Leptin/LR interactions have been studied in several works by means of different molecular modelling approaches, spreading from homology modelling to manual docking. No small molecules have ever been proposed as agonists of the Ob receptor but researchers' efforts focused only on leptin-related synthetic peptides as receptor antagonists and on peptidomimetics. In this review we try to track a timeline of obtained in silico information to clarify the mechanism of interaction between leptin and its receptor, together to summarize the more recent efforts to propose new drugs usable in anti-obesity therapy. Final considerations could be useful starting points for the rational drug design of new lead compounds.


Subject(s)
Anti-Obesity Agents/pharmacology , Drug Design , Leptin/agonists , Receptors, Leptin/drug effects , Anti-Obesity Agents/chemistry , Body Weight/drug effects , Humans , Infant, Newborn , Leptin/metabolism , Models, Molecular , Obesity/drug therapy , Obesity/metabolism , Protein Binding , Receptors, Leptin/chemistry , Receptors, Leptin/metabolism , Risk Factors , Signal Transduction
16.
Physiol Behav ; 120: 83-92, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23911693

ABSTRACT

Previous studies with chronic decerebrate rats and rats infused with leptin into the 4th ventricle suggest that hindbrain leptin receptors attenuate the catabolic effect of forebrain leptin receptor activation. To test this further, rats were fitted with both 3rd and 4th ventricle cannulae. They were infused for 12 days with different combinations of saline, low dose leptin or leptin receptor antagonist (leptin mutein protein). Infusion of 0.1 µg leptin/day into the 3rd ventricle or 0.6 µg leptin/day into the 4th ventricle had no significant effect on food intake, energy expenditure or body composition. Infusion of 2 µg mutein/day into either ventricle caused a small, but significant weight gain. When mutein was infused into one ventricle and leptin into the other, the rats lost weight irrespective of which combination was applied. Surprisingly, rats that received leptin infusions into both ventricles showed an initial hypophagia, no change in energy expenditure, but a 75% loss of carcass fat after 12 days. These data suggest that neuronal pathways activated by leptin receptors in either the forebrain or hindbrain modulate each other's effects. In normal conditions hindbrain leptin may attenuate the catabolic effect of forebrain leptin, but if activity in one area is blocked with mutein, then the catabolic response to leptin in the other ventricle is exaggerated. When receptors in both areas are activated there is an integration of response to produce negative energy balance. This may ensure that leptin causes a loss of fat only when leptin is elevated in both the CSF and periphery.


Subject(s)
Leptin/pharmacology , Prosencephalon/drug effects , Prosencephalon/metabolism , Receptors, Leptin/drug effects , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Weight Loss/drug effects , Adiposity/drug effects , Animals , Body Composition/drug effects , Body Weight/drug effects , Calorimetry , Eating/drug effects , Fourth Ventricle/metabolism , Injections, Intraventricular , Male , Rats , Rats, Sprague-Dawley , Third Ventricle/metabolism
17.
Ther Deliv ; 4(3): 369-94, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23442082

ABSTRACT

Receptor-based targeting of therapeutics may be a fascinating proposition to improve the therapeutic efficacy of encapsulated drugs. The development of safe and effective nanomedicines is a prerequisite in the current nanotechnological scenario. Currently, the surface engineering of nanocarriers has attracted great attention for targeted therapeutic delivery by selective binding of targeting ligand to the specific receptors present on the surface of cells. In this review, we have discussed the current status of various receptors such as transferrin, lectoferrin, lectin, folate, human EGF receptor, scavenger, nuclear and integrin, which are over-expressed on the surface of cancer cells; along with the relevance of targeted delivery systems such as nanoparticles, polymersomes, dendrimers, liposomes and carbon nanotubes. The review also focuses on the effective utilization of receptor-based targeted delivery systems for the management of cancer in effective ways by minimizing the drug-associated side effects and improving the therapeutic efficacy of developed nano-architectures.


Subject(s)
Drug Delivery Systems , Neoplasms/drug therapy , Receptors, Cell Surface/drug effects , Animals , ErbB Receptors/drug effects , Humans , Hyaluronan Receptors/drug effects , Nanoparticles , Receptors, Leptin/drug effects , Receptors, Transferrin/drug effects , Receptors, Tumor Necrosis Factor/drug effects
20.
J Cell Physiol ; 228(6): 1202-9, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23129404

ABSTRACT

Leptin, a hormone-cytokine produced primarily in the adipose tissue, has pleiotropic effects on many biological systems and in several cell types, including immune cells. Hyperleptinemia is associated with immune dysfunction and carcinogenesis. Natural killer (NK) cells are critical mediators of anti-tumor immunity, and leptin receptor deficiency in mice leads to impaired NK function. It was thus decided to explore the in vitro effects of leptin on human NK cell function. NK-92 cells were cultured during 48 h with different leptin concentrations [absence, 10 (physiological), 100 (obesity), or 200 ng/ml (pharmacology)]. Their metabolic activity was assessed using the resazurin test. NK-92 cell cytotoxicity and intracellular IFN-γ production were analyzed by flow cytometry. NK-92 cell mRNA and protein expression levels of cytotoxic effectors were determined by RT-qPCR and Western blot. In our conditions, leptin exerted a dose-dependent stimulatory effect on NK-92 cell metabolic activity. In addition, high leptin concentrations enhanced NK-92 cell cytotoxicity against K562-EGFP and MDA-MB-231-EGFP target cells and inversely reduced cytotoxicity against the MCF-7-EGFP target. At 100 ng/ml, leptin up-regulated both NK cell granzyme B and TRAIL protein expressions and concomitantly down-regulated perforin expression without affecting Fas-L expression. In response to PMA/ionomycin stimulation, the proportion of IFN-γ expressing NK-92 cells increased with 100 and 200 ng/ml of leptin. In conclusion, leptin concentration, at obesity level, variably increased NK-92 cell metabolic activity and modulated NK cell cytotoxicity according to the target cells. The underlying mechanisms are partly due to an up-regulation of TRAIL and IFN-γ expression and a down-regulation of perforin.


Subject(s)
Cytotoxicity, Immunologic/drug effects , Killer Cells, Natural/drug effects , Leptin/pharmacology , Blotting, Western , Dose-Response Relationship, Drug , Down-Regulation , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Humans , Indicators and Reagents , Interferon-gamma/metabolism , Interferon-gamma Release Tests , Ionomycin/pharmacology , K562 Cells , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , MCF-7 Cells , Oxazines , Perforin/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Leptin/drug effects , Receptors, Leptin/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , TNF-Related Apoptosis-Inducing Ligand/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Time Factors , Transfection , Up-Regulation , Xanthenes
SELECTION OF CITATIONS
SEARCH DETAIL
...