Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Neuropharmacology ; 257: 110058, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38960135

ABSTRACT

Postnatal hippocampal neurogenesis is essential for learning and memory. Hippocampal neural precursor cells (NPCs) can be induced to proliferate and differentiate into either glial cells or dentate granule cells. Notably, hippocampal neurogenesis decreases dramatically with age, partly due to a reduction in the NPC pool and a decrease in their proliferative activity. Alpha-melanocyte-stimulating hormone (α-MSH) improves learning, memory, neuronal survival and plasticity. Here, we used postnatally-isolated hippocampal NPCs from Wistar rat pups (male and female combined) to determine the role of the melanocortin analog [Nle4, D-Phe7]-α-MSH (NDP-MSH) in proliferation and fate acquisition of NPCs. Incubation of growth-factor deprived NPCs with 10 nM NDP-MSH for 6 days increased the proportion of Ki-67- and 5-bromo-2'-deoxyuridine (BrdU)-positive cells, compared to the control group, and these effects were blocked by the MC4R antagonist JKC-363. NDP-MSH also increased the proportion of glial fibrillar acidic protein (GFAP)/Ki-67, GFAP/sex-determining region Y-box2 (SOX2) and neuroepithelial stem cell protein (NESTIN)/Ki-67-double positive cells (type-1 and type-2 precursors). Finally, NDP-MSH induced peroxisome proliferator-activated receptor (PPAR)-γ protein expression, and co-incubation with the PPAR-γ inhibitor GW9662 prevented the effect of NDP-MSH on NPC proliferation and differentiation. Our results indicate that in vitro activation of MC4R in growth-factor-deprived postnatal hippocampal NPCs induces proliferation and promotes the relative expansion of the type-1 and type-2 NPC pool through a PPAR-γ-dependent mechanism. These results shed new light on the mechanisms underlying the beneficial effects of melanocortins in hippocampal plasticity and provide evidence linking the MC4R and PPAR-γ pathways in modulation of hippocampal NPC proliferation and differentiation.

2.
Mol Neurobiol ; 2024 Jan 26.
Article in English | MEDLINE | ID: mdl-38277117

ABSTRACT

Spinal cord injury (SCI) is a destructive neurological trauma that induces permanent sensory and motor impairment as well as a deficit in autonomic physiological function. Melanocortin receptor 4 (MC4R) is a G protein-linked receptor that is extensively expressed in the neural system and contributes to inhibiting inflammation, regulating mitochondrial function, and inducing programmed cell death. However, the effect of MC4R in the modulation of oxidative stress and whether this mechanism is related to the role of absent in melanoma 2 (AIM2) in SCI are not confirmed yet. In the current study, we demonstrated that MC4R is significantly increased in the neurons of spinal cords after trauma and oxidative stimulation of cells. Further, activation of MC4R by RO27-3225 effectively improved functional recovery, inhibited AIM2 activation, maintained mitochondrial homeostasis, repressed oxidative stress, and prevented Drp1 translocation to the mitochondria. Meanwhile, treating Drp1 inhibitors would be beneficial in reducing AIM2 activation, and activating AIM2 could abolish the protective effect of MC4R on neuron homeostasis. In conclusion, we demonstrated that MC4R protects against neural injury through a novel process by inhibiting mitochondrial dysfunction, oxidative stress, as well as AIM2 activation, which may serve as an available candidate for SCI therapy.

3.
J Adv Vet Anim Res ; 10(3): 449-457, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37969798

ABSTRACT

Objective: The objective of this study was to associate the 1016G > A variant of the melanocortin-4 receptor gene with lamb's weight, growth, and meat quality in the Colombian hair sheep breed. Materials and Methods: A total of 168 lambs, weights were measured at birth weight (BW), at weaning adjusted weaning weight (AWW), at 6 months [adjusted weight (AW180)], at slaughter adjusted slaughter weight (ASW), daily weight gain preweaning daily gain (preWDG), and postweaning daily gain (postWDG) weaning, and after slaughter, pH, texture, and cook-loss (CL) in the longissimus thoracis et lumborum (LTL) muscle according to the American Meat Science Association methodology. The 1016G > A genotypes were obtained by sequencing. Genotypic and allele frequencies, heterozygosities, and Hardy-Weinberg equilibrium (HWE) were estimated. Using a generalized linear model, the genotype and the allelic substitution effect were associated with the evaluated traits. Results: The heterozygous genotype (0.48) and G allele (0.61) were the most frequent. Heterozygosities were similar (0.47), indicating HWE. The genotype affected the BW (p < 0.05), with a higher value for the GG genotype (2.69 kg). AWW (12.75 kg), AW180 (19.67 kg), and ASW (31.21 kg) weights and daily weight gain (preWDG = 115.41 gm; postWDG = 96.16 gm) were not associated. Average pH, Warner-Bratzler shear force, and CL were 5.75 units, 49.46 N, and 32.02%, with no genotype effect. The G > A substitution only affected BW at -388 gm (p < 0.05). Conclusion: The 1016G > A variant is polymorphic and affects the BW but not other growth traits or the meat quality of the LTL muscle.

4.
BMC Endocr Disord ; 23(1): 83, 2023 Apr 18.
Article in English | MEDLINE | ID: mdl-37072742

ABSTRACT

BACKGROUND: Previous studies have shown that the minor allele (C allele) for melanocortin 4 receptor (MC4R) rs17782313 may be associated with depressed mood. Moreover, dietary patterns have potentially adverse effects on depression. This study investigates the interactions between the MC4R gene variant (rs17782313) and dietary patterns on depression among Iranian obese and overweight women. METHODS: A total of 289 Iranian overweight and obese women, aged 18-50 years, were enrolled in this cross-sectional study. Biochemical, anthropometric, and body composition indices were assessed in all participants. Moreover, MC4R rs17782313, by the restriction fragment length polymorphism (PCR-RFLP) method, and depression, using the 21-item Depression Anxiety Stress Scales (DASS) questionnaire, were assessed. Food intakes were assessed by completing a 147-item semi-quantitative food frequency questionnaire (FFQ). RESULTS: By the use of factor analysis, 2 major dietary patterns were extracted: healthy dietary pattern (HDP) and unhealthy dietary pattern (UDP). Binary logistic analysis showed that individuals with minor allele risk (CC) with high adherence to the unhealthy pattern increased odds for depression (OR: 8.77, 95%CI: -0.86-18.40, P: 0.07), after controlling for confounders. Also, a logical inverse relationship was observed between CT genotype and HDP on depression in the crude and adjusted models (OR: -0.56, 95% CI: -3.69-2.57, P: 0.72) (OR: -4.17, 95% CI: -9.28-0.94, P: 0.11), although this interaction was not statistically significant. CONCLUSION: According to the above findings, adherence to unhealthy food intake pattern increases odds of depression in MC4R risk allele (C allele) carriers. To confirm these findings, more studies are needed in the form of clinical trials and prospective studies with higher sample sizes.


Subject(s)
Depression , Overweight , Humans , Female , Overweight/epidemiology , Overweight/genetics , Cross-Sectional Studies , Prospective Studies , Iran/epidemiology , Depression/etiology , Depression/genetics , Obesity/genetics , Genotype , Polymorphism, Single Nucleotide , Body Mass Index , Receptor, Melanocortin, Type 4/genetics
5.
Adv Biol (Weinh) ; 7(9): e2300035, 2023 09.
Article in English | MEDLINE | ID: mdl-37043700

ABSTRACT

Obesity has emerged as a critical and urgent health burden during the current global pandemic. Among multiple genetic causes, melanocortin receptor-4 (MC4R), involved in food intake and energy metabolism regulation through various signaling pathways, has been reported to be the lead genetic factor in severe and early onset obesity and hyperphagia disorders. Most previous studies have illustrated the roles of MC4R signaling in energy intake versus expenditure in the central system, while some evidence indicates that MC4R is also expressed in peripheral systems, such as the gut and endocrine organs. However, its physiopathological function remains poorly defined. This review aims to depict the central and peripheral roles of MC4R in energy metabolism and endocrine hormone homeostasis, the diversity of phenotypes, biased downstream signaling caused by distinct MC4R mutations, and current drug development targeting the receptor.


Subject(s)
Hyperphagia , Obesity , Humans , Obesity/genetics , Obesity/metabolism , Hyperphagia/complications , Hyperphagia/metabolism , Receptor, Melanocortin, Type 4/genetics , Receptor, Melanocortin, Type 4/metabolism , Signal Transduction/genetics , Energy Intake
6.
Am J Physiol Cell Physiol ; 324(3): C694-C706, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36717105

ABSTRACT

The inward rectifier potassium channel Kir7.1, encoded by the KCNJ13 gene, is a tetramer composed of two-transmembrane domain-spanning monomers, closer in homology to Kir channels associated with potassium transport such as Kir1.1, 1.2, and 1.3. Compared with other channels, Kir7.1 exhibits small unitary conductance and low dependence on external potassium. Kir7.1 channels also show a phosphatidylinositol 4,5-bisphosphate (PIP2) dependence for opening. Accordingly, retinopathy-associated Kir7.1 mutations mapped at the binding site for PIP2 resulted in channel gating defects leading to channelopathies such as snowflake vitreoretinal degeneration and Leber congenital amaurosis in blind patients. Lately, this channel's role in energy homeostasis was reported due to the direct interaction with the melanocortin type 4 receptor (MC4R) in the hypothalamus. As this channel seems to play a multipronged role in potassium homeostasis and neuronal excitability, we will discuss what is predicted from a structural viewpoint and its possible implications for hunger control.


Subject(s)
Potassium Channels, Inwardly Rectifying , Humans , Mutation , Neurons/metabolism , Potassium/metabolism , Potassium Channels, Inwardly Rectifying/genetics , Potassium Channels, Inwardly Rectifying/metabolism , Protein Domains
7.
World J Clin Cases ; 9(11): 2688-2695, 2021 Apr 16.
Article in English | MEDLINE | ID: mdl-33889637

ABSTRACT

BACKGROUND: Abnormalities in the melanocortin receptor 4 (MC4R) gene often lead to obesity, but are rarely associated with other conditions such as epilepsy and sleep disorder. CASE SUMMARY: Here, we present a case of a male obese child with a heterozygous variant in MC4R (c.494G>A, p.Arg165Gln) inherited from his father, who presented with disordered sleep and abnormal facial movements. Examination through melatonin rhythm testing and electroencephalography led to a diagnosis of sleep disorder and epilepsy, as his melatonin rhythm was markedly distorted and the electroencephalography revealed epileptic discharges. He received treatment with an antiepileptic drug; however, the therapy was ineffective and the sleep disorder appeared to be deteriorating. Subsequently, we initiated adjuvant treatment with melatonin. Upon re-examination, his body mass index had decreased, the sleep disturbance had resolved, and his seizures were well controlled. Electro-encephalography review was normal, and a typical melatonin rhythm was restored. CONCLUSION: We concluded that, in addition to causing obesity, abnormalities in the MC4R gene may contribute to the development of sleep disorders and epilepsy, and that melatonin can be used as an adjuvant therapy to alleviate these symptoms.

8.
Front Immunol ; 10: 2312, 2019.
Article in English | MEDLINE | ID: mdl-31636637

ABSTRACT

To date, available treatment strategies for multiple sclerosis (MS) are ineffective in preventing or reversing progressive neurologic deterioration, creating a high, and unmet medical need. One potential way to fight MS may be by limiting the detrimental effects of reactive astrocytes, a key pathological hallmark for disease progression. One class of compounds that may exert beneficial effects via astrocytes are melanocortin receptor (MCR) agonists. Among the MCR, MC4R is most abundantly expressed in the CNS and several rodent studies have described that MC4R is-besides neurons-expressed by astrocytes. Activation of MC4R in astrocytes has shown to have potent anti-inflammatory as well as neuroprotective effects in vitro, suggesting that this could be a potential target to ameliorate ongoing inflammation, and neurodegeneration in MS. In this study, we set out to investigate human MC4R expression and analyze its downstream effects. We identified MC4R mRNA and protein to be expressed on astrocytes and observed increased astrocytic MC4R expression in active MS lesions. Furthermore, we show that the novel, highly selective MC4R agonist setmelanotide ameliorates the reactive phenotype in astrocytes in vitro and markedly induced interleukin-6 and -11 production, possibly through enhanced cAMP response element-binding protein (CREB) phosphorylation. Notably, stimulation of human macrophages with medium from astrocytes that were exposed to setmelanotide, skewed macrophages toward an anti-inflammatory phenotype. Taken together, these findings suggest that targeting MC4R on astrocytes might be a novel therapeutic strategy to halt inflammation-associated neurodegeneration in MS.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Astrocytes/drug effects , Macrophages/drug effects , Receptor, Melanocortin, Type 4/agonists , alpha-MSH/analogs & derivatives , Adult , Aged , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/metabolism , Female , Humans , Interleukin-11/biosynthesis , Interleukin-6/biosynthesis , Male , Middle Aged , Multiple Sclerosis/drug therapy , Phenotype , Phosphorylation , Receptor, Melanocortin, Type 4/drug effects , Receptor, Melanocortin, Type 4/genetics , alpha-MSH/pharmacology
9.
J Neuroinflammation ; 15(1): 106, 2018 Apr 11.
Article in English | MEDLINE | ID: mdl-29642894

ABSTRACT

BACKGROUND: Neuroinflammation plays an important role in the pathogenesis of intracerebral hemorrhage (ICH)-induced secondary brain injury. Activation of melanocortin receptor 4 (MC4R) has been shown to elicit anti-inflammatory effects in many diseases. The objective of this study was to explore the role of MC4R activation on neuroinflammation in a mouse ICH model and to investigate the contribution of adenosine monophosphate-activated protein kinase (AMPK)/c-Jun N-terminal kinase (JNK)/p38 mitogen-activated protein kinase (p38 MAPK) pathway in MC4R-mediated protection. METHODS: Adult male CD1 mice (n = 189) were subjected to intrastriatal injection of bacterial collagenase or sham surgery. The selective MC4R agonist RO27-3225 was administered by intraperitoneal injection at 1 h after collagenase injection. The specific MC4R antagonist HS024 and selective AMPK inhibitor dorsomorphin were administered prior to RO27-3225 treatment to elucidate potential mechanism. Short- and long-term neurobehavioral assessments, brain water content, immunofluorescence staining, and western blot were performed. RESULTS: The expression of MC4R and p-AMPK increased after ICH with a peak at 24 h. MC4R was expressed by microglia, neurons, and astrocytes. Activation of MC4R with RO27-3225 improved the neurobehavioral functions, decreased brain edema, and suppressed microglia/macrophage activation and neutrophil infiltration after ICH. RO27-3225 administration increased the expression of MC4R and p-AMPK while decreasing p-JNK, p-p38 MAPK, TNF-α, and IL-1ß expression, which was reversed with inhibition of MC4R and AMPK. CONCLUSIONS: Our study demonstrated that activation of MC4R with RO27-3225 attenuated neuroinflammation through AMPK-dependent inhibition of JNK and p38 MAPK signaling pathway, thereby reducing brain edema and improving neurobehavioral functions after experimental ICH in mice. Therefore, the activation of MC4R with RO27-3225 may be a potential therapeutic approach for ICH management.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anti-Inflammatory Agents/therapeutic use , Encephalitis/drug therapy , Peptides/therapeutic use , Receptor, Melanocortin, Type 4/metabolism , Signal Transduction/drug effects , Animals , Brain Edema/drug therapy , Brain Edema/etiology , Calcium-Binding Proteins/metabolism , Cerebral Hemorrhage/complications , Disease Models, Animal , Encephalitis/etiology , Enzyme Inhibitors/therapeutic use , Gene Expression Regulation/drug effects , MAP Kinase Kinase 4/metabolism , Macrophages/drug effects , Male , Mice , Microfilament Proteins/metabolism , Microglia/drug effects , Neutrophil Infiltration/drug effects , Time Factors , p38 Mitogen-Activated Protein Kinases/metabolism
10.
Mol Neurobiol ; 55(6): 4984-4997, 2018 Jun.
Article in English | MEDLINE | ID: mdl-28791594

ABSTRACT

Currently, no description of melanocortin receptor-4 (MC4R) expression or activity is available in human cancer cells, including glioblastoma (GBM). The aim of this study is to evaluate the presence of MC4Rs in GBM cells and the selective inhibition of their activity through the MC4R antagonist ML00253764 alone and in association with temozolomide in vitro and in vivo. MC4R genotyping and gene expression were performed on human GBM cells (U-87 and U-118) with real-time PCR. MC4R western blotting, immunohistochemistry, and immunofluorescence were obtained in both cell lines and in human tissues. Proliferation, cell cycle, and apoptotic assays were performed with ML00253764, whereas the synergism of the simultaneous combination with temozolomide was evaluated by the combination index method. ERK1/2 and Akt phosphorylation were quantified by ELISA. In vivo experiments were performed in U-87 xenografted nude mice. Both GBM cell lines and tumor tissues expressed MC4R receptors. The selective antagonist ML00253764 determined an antiproliferative and proapoptotic activity through the inhibition of the phosphorylation of ERK1/2 and Akt. Moreover, the simultaneous combination of temozolomide and ML00253764 determined a highly synergistic effect on GBM cells. The same combination in vivo showed a strong and significant decrease of GBM tumor volumes if compared to the single drug treatments, with an excellent tolerability profile. In conclusion, MC4R is present in GBM cells and its selective inhibition determined antiproliferative and proapoptotic effects, through the inhibition of ERK1/2 and Akt phosphorylation, and the synergistic enhancement of temozolomide effects in vitro and in vivo.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Imidazoles/pharmacology , Neurons/drug effects , Receptor, Melanocortin, Type 4/metabolism , Temozolomide/pharmacology , Animals , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Humans , Mice , Mice, Nude , Neurons/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism
11.
Mol Neurobiol ; 55(2): 1396-1404, 2018 02.
Article in English | MEDLINE | ID: mdl-28150230

ABSTRACT

Melanocortins are peptides with well-recognized antiinflammatory and neuroprotective activity. No data are currently available on melanocortin receptor-4 (MC4R) gene polymorphisms and tumors, including glioblastomas (GBMs), or their relationship with radiotherapy or chemotherapy. The aim of this study was to evaluate the possible predictive/prognostic role of the MC4R SNPs on GBM patients. Fifty-five patients with a proven diagnosis of GBM, treated with radiotherapy and temozolomide, were consecutively enrolled. MC4R gene SNPs (rs17782313, rs489693, rs8087522, rs17700633) were analyzed by a validated TaqMan® SNP genotyping assays. Univariate and multivariate analyses were performed. A P < 0.0125 (Bonferroni's correction) was considered significant ( Clinicaltrial.gov identifier NCT02458508). The median progression-free survival (PFS) and median overall survival (OS) of these patients were 9.54 (95% CI 5.4-14.3) months and 24.9 (95% CI 17.8-34.6) months, respectively. The MC4R rs489693 AA genotype was significantly associated with a shorter PFS and OS. Indeed, with regard to PFS, patients harboring the rs489693 AA genotype had a median PFS of 2.99 months whereas patients with AC/CC genotypes had a median PFS of 10.82 months (P = 0.009). Interestingly, the rs489693 AA patients also had a lower median OS as compared with the median OS of the AC/CC genotypes (10.75 vs. 29.5 months, respectively, P = 0.0001). This study suggests that the MC4R rs489693 AA genotype is significantly associated with a shorter PFS and OS in patients treated with radiotherapy and temozolomide. These findings represent a relevant effort to identify novel clinical markers for RT-CT therapy in GBM to be validated in future pharmacogenetic clinical trials.


Subject(s)
Brain Neoplasms/genetics , Glioblastoma/genetics , Polymorphism, Single Nucleotide , Receptor, Melanocortin, Type 4/genetics , Adult , Aged , Aged, 80 and over , Brain Neoplasms/mortality , Brain Neoplasms/therapy , Chemoradiotherapy , Female , Glioblastoma/mortality , Glioblastoma/therapy , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , Treatment Outcome , Young Adult
12.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-709991

ABSTRACT

As a pandemic metabolic disease, obesity has become a worldwide problem, which endangering human health. The change in body weight reflects the energy imbalance. And the homeostasis regulation of the central and peripheral melanocortin system may maintain the energy balance and then affect the body weight. Recent studies have found that melanocortin receptor-4 ( MC4R) plays an important role in energy homeostasis and body weight changes, but its specific mechanism needs further elucidation. In this paper, we reviewed the research progress of MC4R in body weight and energy regulation, so as to provide a theoretical basis for further exploring the potential role of MC4R in improving energy metabolism imbalance and obesity.

13.
Biochem Biophys Res Commun ; 469(2): 281-7, 2016 Jan 08.
Article in English | MEDLINE | ID: mdl-26631960

ABSTRACT

Long-term glucocorticoid (GC) usage may cause non-traumatic femoral head osteonecrosis. Dexamethasone (Dex) is shown to exert potent cytotoxic effect to osteoblasts. Here, we investigated the potential activity of α-melanocyte stimulating hormone (α-MSH) against the process. Our data revealed that pretreatment of α-MSH significantly inhibited Dex-induced apoptosis and necrosis in both osteoblastic-like MC3T3-E1 cells and primary murine osteoblasts. Melanocortin receptor 4 (MC4R) acts as the receptor of α-MSH in mediating its actions in osteoblasts. The MC4R antagonist SHU9119, or shRNA-mediated knockdown of MC4R, almost abolished α-MSH-induced activation of downstream signalings (Akt and Erk1/2) and its pro-survival effect in osteoblasts. Further studies showed that α-MSH activated MC4R downstream sphingosine kinase 1 (SphK1) and increased cellular sphingosine-1-phosphate (S1P) content in MC3T3-E1 cells and primary murine osteoblasts, which were blocked by SHU9119 or MC4R shRNAs. SphK1 inhibition by the its inhibitor N,N-dimethylsphingosine (DMS), or SphK1 knockdown by targeted-shRNAs, largely attenuated α-MSH-mediated osteoblast protection against Dex. Together, these results suggest that α-MSH alleviates Dex-induced damages to cultured osteoblasts through activating MC4R-SphK1 signaling.


Subject(s)
Dexamethasone/administration & dosage , Osteoblasts/metabolism , Osteoblasts/pathology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptor, Melanocortin, Type 4/metabolism , alpha-MSH/administration & dosage , Animals , Apoptosis/drug effects , Apoptosis/physiology , Cell Line , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Drug Interactions , Mice , Osteoblasts/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology
14.
Gene ; 563(2): 125-9, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25769385

ABSTRACT

BACKGROUND: Malfunction in the energy homeostasis system is a major cause of developing obesity. Melanocortin-4 receptor (MC4R) plays a crucial role in this system as a key receptor. Although MC4R gene as an obesity candidate gene is associated with higher BMI, only few attempts have been carried out to understand the mechanism underlying body-weight regulation. OBJECTIVE: The aim of this study is to investigate the association between variant rs17782313 near MC4R gene and both dietary energy and macronutrient intakes. METHODS: An Iranian population, 400 adults aged over 22years were selected from the Iranian Multicenter Osteoporosis Study (IMOS). Genotyping for the near MC4R rs17782313 was performed by PCR-RFLP. Weight and height were measured. Dietary intake and physical activity were assessed by using validated questionnaires. Analysis was carried out in two groups with regard to BMI. Multiple linear regression models adjusted for covariates were used to examine the association between rs17782313 and dietary intake. RESULTS: MC4R rs17782313 was associated with high energy intake (P<0.001), and low carbohydrate and protein intakes (P<0.001 and P<0.01 respectively). In addition, the significant association between variant rs17782313 and fat intake disappeared after adjusting for energy. CONCLUSIONS: The rs17782313 variant contributes to the variety of dietary energy and energy-dense macronutrient intakes. Moreover, a novel association was suggested between this polymorphism and dietary fat intake.


Subject(s)
Polymorphism, Genetic/genetics , Receptor, Melanocortin, Type 4/genetics , Adult , Diet , Energy Intake , Female , Genetic Predisposition to Disease , Genotype , Glycoside Hydrolases/metabolism , Humans , Iran , Male , Middle Aged , Obesity/genetics , Obesity/metabolism , Proteins/metabolism , Risk Factors
15.
J Endocrinol ; 220(2): 97-107, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24204009

ABSTRACT

Obesity is a major burden to people and to health care systems around the world. The aim of the study was to characterize the effect of a novel selective α-MSH analog on obesity and insulin sensitivity. The subchronic effects of the selective MC4-R peptide agonist MC4-NN1-0182 were investigated in diet-induced obese (DIO) rats and DIO minipigs by assessing the effects on food intake, energy consumption, and body weight. The acute effect of MC4-NN1-0182 on insulin sensitivity was assessed by a euglycemic-hyperinsulinemic clamp study in normal rats. Three weeks of treatment of DIO rats with MC4-NN1-0182 caused a decrease in food intake and a significant decrease in body weight 7±1%, P<0.05 compared with 3±1% increase with the vehicle control. In DIO minipigs, 8 weeks of treatment with MC4-NN1-0182 resulted in a body weight loss of 13.3±2.5 kg (13±3%), whereas the vehicle control group had gained 3.7±1.4 kg (4±1%). Finally, clamp studies in normal rats showed that acute treatment with MC4-NN1-0182 caused a significant increase in glucose disposal (Rd) compared with vehicle control (Rd, mg/kg per min, 17.0±0.7 vs 13.9±0.6, P<0.01). We demonstrate that treatment of DIO rats or minipigs with a selective MC4-R peptide agonist causes weight loss. Moreover, we have demonstrated weight-independent effects on insulin sensitivity. Our observations identify MC4 agonism as a viable target for the treatment of obesity and insulin resistance.


Subject(s)
Anti-Obesity Agents/pharmacology , Insulin Resistance , Obesity/metabolism , Weight Loss/drug effects , alpha-MSH/analogs & derivatives , alpha-MSH/pharmacology , Animals , Diet , Drug Evaluation, Preclinical , Female , Insulin Resistance/physiology , Male , Obesity/drug therapy , Obesity/etiology , Rats , Rats, Sprague-Dawley , Swine , Swine, Miniature , alpha-MSH/therapeutic use
16.
Gene ; 527(1): 193-200, 2013 Sep 15.
Article in English | MEDLINE | ID: mdl-23774689

ABSTRACT

Agouti-related protein (AgRP) is an important neuropeptide involved in the regulation of feeding in both mammals and fish. In this study, we have cloned the full-length cDNA sequence for AgRP in a cyprinid fish (Schizothorax prenanti). The AgRP gene, encoding 126-amino acids, was strongly expressed in the brain. The AgRP gene was detected in embryos at developmental stages. Further, its mRNA was detectable in unfertilized eggs. An experiment was conducted to determine the expression profile of AgRP during short-term and long-term fasting of the hypothalamus. The expression level of AgRP in unfed fish was significantly increased at 3 and 4h post-fasting than in fed fish but did not affect AgRP mRNA expression after 14 days fasting. Overall, our results suggest that AgRP is a conserved peptide that might be involved in the regulation of short-term feeding and other physiological function in Schizothorax prenanti.


Subject(s)
Agouti-Related Protein/metabolism , Cyprinidae/metabolism , Fish Proteins/metabolism , Hypothalamus/metabolism , Agouti-Related Protein/genetics , Amino Acid Sequence , Animals , Base Sequence , Brain/metabolism , Conserved Sequence , Cyprinidae/genetics , Fasting/metabolism , Female , Fish Proteins/genetics , Gene Expression Regulation, Developmental , Male , Molecular Sequence Data , Organ Specificity , Phylogeny , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Neuroscience ; 247: 43-54, 2013 Sep 05.
Article in English | MEDLINE | ID: mdl-23701881

ABSTRACT

Thyrotropin-releasing hormone (TRH), a neuropeptide contained in neural terminals innervating brainstem vagal motor neurons, enhances vagal outflow to modify multisystemic visceral functions and food intake. Type 2 diabetes (T2D) and obesity are accompanied by impaired vagal functioning. We examined the possibility that impaired brainstem TRH action may contribute to the vagal dysregulation of food intake in Goto-Kakizaki (GK) rats, a T2D model with hyperglycemia and impaired central vagal activation by TRH. Food intake induced by intracisternal injection of TRH analog was reduced significantly by 50% in GK rats, compared to Wistar rats. Similarly, natural food intake in the dark phase or food intake after an overnight fast was reduced by 56-81% in GK rats. Fasting (48h) and refeeding (2h)-associated changes in serum ghrelin, insulin, peptide YY, pancreatic polypeptide and leptin, and the concomitant changes in orexigenic or anorexigenic peptide expression in the brainstem and hypothalamus, all apparent in Wistar rats, were absent or markedly reduced in GK rats, with hormone release stimulated by vagal activation, such as ghrelin and pancreatic polypeptide, decreased substantially. Fasting-induced Fos expression accompanying endogenous brainstem TRH action decreased by 66% and 91%, respectively, in the nucleus tractus solitarius (NTS) and the dorsal motor nucleus of the vagus (DMV) in GK rats, compared to Wistar rats. Refeeding abolished fasting-induced Fos-expression in the NTS, while that in the DMV remained in Wistar but not GK rats. These findings indicate that dysfunctional brainstem TRH-elicited vagal impairment contributes to the disturbed food intake in T2D GK rats, and may provide a pathophysiological mechanism which prevents further weight gain in T2D and obesity.


Subject(s)
Brain Stem/metabolism , Diabetes Mellitus, Type 2/blood , Eating/physiology , Thyrotropin-Releasing Hormone/physiology , Vagus Nerve/metabolism , Animals , Blood Glucose/metabolism , Diabetes Mellitus, Type 2/physiopathology , Fasting/blood , Male , Rats , Rats, Wistar
18.
Mol Metab ; 2(2): 74-85, 2012.
Article in English | MEDLINE | ID: mdl-24199146

ABSTRACT

MicroRNAs (miRNAs) have recently emerged as key regulators of metabolism. However, their potential role in the central regulation of whole-body energy homeostasis is still unknown. In this study we show that the expression of Dicer, an essential endoribonuclease for miRNA maturation, is modulated by nutrient availability and excess in the hypothalamus. Conditional deletion of Dicer in POMC-expressing cells resulted in obesity, characterized by hyperphagia, increased adiposity, hyperleptinemia, defective glucose metabolism and alterations in the pituitary-adrenal axis. The development of the obese phenotype was paralleled by a POMC neuron degenerative process that started around 3 weeks of age. Hypothalamic transcriptomic analysis in presymptomatic POMCDicerKO mice revealed the downregulation of genes implicated in biological pathways associated with classical neurodegenerative disorders, such as MAPK signaling, ubiquitin-proteosome system, autophagy and ribosome biosynthesis. Collectively, our results highlight a key role for miRNAs in POMC neuron survival and the consequent development of neurodegenerative obesity.

SELECTION OF CITATIONS
SEARCH DETAIL
...