Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Neurosci Res ; 96(3): 427-435, 2018 03.
Article in English | MEDLINE | ID: mdl-28877366

ABSTRACT

ACTH, a melanocortin peptide used to treat multiple sclerosis (MS) relapses, acts by stimulating adrenal corticosteroid (CS) production via melanocortin receptor 2 (MC2R), but it may also exert a therapeutic effect independent of CS by stimulating other melanocortin receptors (MCR) distributed in many tissues, including the brain. We reported that oligodendroglia (OL) and oligodendroglial precursor cells (OPC) express MC4R, and that ACTH 1-39 protects OL and OPC in vitro from cell death induced by mechanisms likely involved in white matter damage in MS. This study investigates expression of MC1R, MC2R, MC3R and MC5R in OL and MC4R in OPC using immunocytochemistry with MCR subtype specific antibodies. OL express surface MC1R, MC3R and MC5R, in addition to MC4R. To investigate whether these receptors are functional, we asked if signaling through MCR is involved in ACTH protection of cultured rat OL from apoptosis (staurosporine), or cell death induced by excitotoxicity (glutamate), reactive oxygen species (ROS), or an inflammatory mediator (quinolinic acid). Like ACTH 1-39, MCR subtype specific agonists for MC1R, MC3R, MC4R and MC5R all protected OL from these insults. Conversely, antagonists for MC3R and MC4R blocked ACTH protection of OL. We then investigated the role of MC4R, as a prototype MCR, in protection and proliferation of OPC; MC4R agonists protected OPC and increased their proliferation, while antagonists blocked these effects. Our results demonstrate that MCR on OL and OPC are functional and activate signaling pathways that protect against mechanisms involved in OL damage in MS, suggesting potential beneficial effects in neurologic diseases.


Subject(s)
Adrenocorticotropic Hormone/pharmacology , Oligodendroglia/drug effects , Oligodendroglia/metabolism , Receptors, Melanocortin/biosynthesis , Animals , Animals, Newborn , Apoptosis/drug effects , Cell Death/drug effects , Cells, Cultured , Glutamic Acid/toxicity , Hydrogen Peroxide/toxicity , Immunohistochemistry , Primary Cell Culture , Prosencephalon/drug effects , Prosencephalon/metabolism , Quinolinic Acid/toxicity , Rats, Sprague-Dawley , Receptors, Melanocortin/agonists , Receptors, Melanocortin/antagonists & inhibitors , Staurosporine/toxicity
2.
Horm Behav ; 64(1): 95-102, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23707533

ABSTRACT

Neuropeptide Y (NPY) and nuclear factor-kappa B (NF-κB) are involved in regulating anorexia elicited by phenylpropanolamine (PPA), a sympathomimetic drug. This study explored whether NPY Y1 receptor (Y1R) is involved in this process, and a potential role for the proopiomelanocortin system was identified. Rats were given PPA once a day for 4days. Changes in the hypothalamic expression of the NPY, Y1R, NF-κB, and melanocortin receptor 4 (MC4R) levels were assessed and compared. The results indicated that food intake and NPY expression decreased, with the largest reductions observed on Day 2 (approximately 50% and 45%, respectively), whereas NF-κB, MC4R, and Y1R increased, achieving maximums on Day 2 (160%, 200%, and 280%, respectively). To determine the role of Y1R, rats were pretreated with Y1R antisense or a Y1R antagonist via intracerebroventricular injection 1h before the daily PPA dose. Y1R knockdown and inhibition reduced PPA anorexia and partially restored the normal expression of NPY, MC4R, and NF-κB. The data suggest that hypothalamic Y1R participates in the appetite-suppression from PPA by regulating MC4R and NF-κB. The results of this study increase our understanding of the molecular mechanisms in PPA-induced anorexia.


Subject(s)
Appetite Depressants/pharmacology , Feeding Behavior/drug effects , NF-kappa B/drug effects , Phenylpropanolamine/pharmacology , Receptors, Melanocortin/drug effects , Receptors, Neuropeptide Y/antagonists & inhibitors , Animals , Antisense Elements (Genetics) , Blotting, Western , Body Weight/physiology , Catheterization , Cerebral Ventricles/physiology , Hypothalamus/drug effects , Hypothalamus/metabolism , Inferior Colliculi , Injections , Male , NF-kappa B/biosynthesis , Rats , Rats, Wistar , Receptor, Melanocortin, Type 4/biosynthesis , Receptors, Melanocortin/biosynthesis , Receptors, Neuropeptide Y/biosynthesis , Receptors, Neuropeptide Y/genetics
3.
J Endocrinol ; 217(1): R1-11, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23418361

ABSTRACT

The melanocortin receptor (MCR) family consists of five G-protein-coupled receptors (MC1R-MC5R) with diverse physiological roles. MC1R controls pigmentation, MC2R is a critical component of the hypothalamic-pituitary-adrenal axis, MC3R and MC4R have a vital role in energy homeostasis and MC5R is involved in exocrine function. The melanocortin receptor accessory protein (MRAP) and its paralogue MRAP2 are small single-pass transmembrane proteins that have been shown to regulate MCR expression and function. In the adrenal gland, MRAP is an essential accessory factor for the functional expression of the MC2R/ACTH receptor. The importance of MRAP in adrenal gland physiology is demonstrated by the clinical condition familial glucocorticoid deficiency, where inactivating MRAP mutations account for ∼20% of cases. MRAP is highly expressed in both the zona fasciculata and the undifferentiated zone. Expression in the undifferentiated zone suggests that MRAP could also be important in adrenal cell differentiation and/or maintenance. In contrast, the role of adrenal MRAP2, which is highly expressed in the foetal gland, is unclear. The expression of MRAPs outside the adrenal gland is suggestive of a wider physiological purpose, beyond MC2R-mediated adrenal steroidogenesis. In vitro, MRAPs have been shown to reduce surface expression and signalling of all the other MCRs (MC1,3,4,5R). MRAP2 is predominantly expressed in the hypothalamus, a site that also expresses a high level of MC3R and MC4R. This raises the intriguing possibility of a CNS role for the MRAPs.


Subject(s)
Adrenal Glands/metabolism , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Adaptor Proteins, Signal Transducing , Adrenal Glands/embryology , Animals , Carrier Proteins/chemistry , Gene Expression Regulation , Humans , Hypothalamus/metabolism , Membrane Proteins/chemistry , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, Melanocortin/biosynthesis , Receptors, Melanocortin/chemistry , Receptors, Melanocortin/metabolism
4.
Mol Cell Endocrinol ; 371(1-2): 62-70, 2013 May 22.
Article in English | MEDLINE | ID: mdl-23138111

ABSTRACT

Circadian and ultradian variations of basal glucocorticoid secretion and transient elevations during stress are essential for homeostasis. Using intronic qRT-PCR to measure changes in primary transcript (hnRNA) we have shown that secretory events induced by stress or ACTH injection are followed by episodic increases in transcription of rate limiting steroidogenic proteins, such as steroidogenic acute regulatory protein (StAR), cytochrome P450 side chain cleavage and melanocortin receptor associated protein. These transcriptional episodes imply rapid turnover of steroidogenic proteins and the need of de novo synthesis following each secretory event. In addition to episodic ACTH secretion, it is likely that intracellular feedback mechanisms at the adrenal fasciculata level contribute to the generation of episodes of transcription. The time relationship between activation and translocation of the CREB co-activator, transducer of regulated CREB activity (TORC) to the nucleus preceding transcriptional episodes suggest the involvement of TORC in the transcriptional activation of StAR and other steroidogenic proteins.


Subject(s)
Adrenal Glands/metabolism , Glucocorticoids/metabolism , Steroids/biosynthesis , Adrenocorticotropic Hormone/metabolism , Cyclic AMP Response Element-Binding Protein , Gene Expression Regulation , Humans , Phosphoproteins/biosynthesis , Receptors, Melanocortin/biosynthesis , Stress, Physiological , Transcription, Genetic , Transcriptional Activation
5.
Neuroreport ; 23(10): 596-600, 2012 Jul 11.
Article in English | MEDLINE | ID: mdl-22643233

ABSTRACT

The brain melanocortin (MC) system is one of numerous overlapping systems regulating energy balance; it consists of peptides including α-melanocyte-stimulating hormone that act through melanocortin receptors (MCRs). Mutations and polymorphisms in MC3R and MC4R have been identified as one of the most common genetic contributors to obesity in human studies. Brain MC3R and MC4R are known to modulate energy expenditure (EE) and food intake, but much less is known regarding brain MC5R. To test the hypothesis that brain MC modulates physical activity (PA) and EE, we compared brain MCR profiles in rats that consistently show high versus low levels of 'spontaneous' daily PA. Compared with low-activity rats, high-activity rats show enhanced mRNA expression of MCRs in the brain, specifically of MC3R in the paraventricular nucleus (PVN), and MC4R and MC5R in the perifornical lateral hypothalamus. Next, we microinjected the MCR agonist melanotan II into the PVN region and measured PA and EE. Intra-PVN melanotan II induced a dose-dependent increase in PA and this effect was greater in high-activity rats compared with low-activity rats. These results indicate region-specific brain MCR expression in the heightened PA seen in association with high endurance capacity and identify promising targets in the brain MC system that may contribute to interindividual variability in energy balance.


Subject(s)
Brain/metabolism , Phenotype , Receptor, Melanocortin, Type 4/biosynthesis , Receptors, Melanocortin/biosynthesis , Thinness/metabolism , Animals , Eating/physiology , Female , Male , Motor Activity/physiology , Rats , Receptor, Melanocortin, Type 3 , Receptor, Melanocortin, Type 4/genetics , Receptors, Melanocortin/genetics , Thinness/genetics
6.
Eur J Pharmacol ; 680(1-3): 1-7, 2012 Apr 05.
Article in English | MEDLINE | ID: mdl-22314217

ABSTRACT

The melanocortin-5 receptor (MC(5)) of the dogfish Squalus acanthias (SacMC(5) receptor) can be functionally expressed in CHO cells in the absence of the co-expression of an exogenous MRAP cDNA. Both human ACTH(1-24) and dogfish ACTH(1-25) were much better stimulators of the SacMC(5) receptor than any of the mammalian or dogfish MSH ligands that were tested. The order of ligand selectivity for the dogfish melanocortins was ACTH(1-25)>αMSH>γ-MSH=δ-MSH>ß-MSH. Unlike mammalian MC(5) receptors, the functional expression of the SacMC(5) receptor was not negatively impacted when the receptor was co-expressed with a cartilaginous fish (Callorhinchus milii) MRAP2 cDNA. However, co-expression with either mouse mMRAP1 or zebrafish zfMRAP1 increased the sensitivity of SacMC(5) receptor for hACTH(1-24) by at least one order of magnitude. Hence, SacMC(5) receptor has the potential to interact with MRAP1 orthologs and in this regard behaved more like a melanocortin MC(2) receptor ortholog than a melanocortin MC(5) receptor ortholog. These observations are discussed in light of the evolution of the melanocortin receptor gene family in cartilaginous fish, and the physiological implications of these observations are considered.


Subject(s)
Membrane Proteins/genetics , Membrane Proteins/metabolism , Receptors, Melanocortin/biosynthesis , Receptors, Melanocortin/genetics , Squalus acanthias/genetics , Squalus acanthias/metabolism , Amino Acid Sequence , Animals , CHO Cells , Cricetinae , Humans , Ligands , Melanocortins/metabolism , Mice , Molecular Sequence Data , Receptors, Melanocortin/metabolism , Zebrafish
7.
Endocr Pathol ; 19(2): 122-7, 2008.
Article in English | MEDLINE | ID: mdl-18618087

ABSTRACT

Myelolipomas of the adrenal gland are benign, nonfunctioning tumors. Patients with congenital adrenal hyperplasia sometimes develop large and bilateral myelolipomas. Although the precise pathogenesis of myelolipomas remains unclear, prolonged stimulation with high levels of adrenocorticotropic hormone (ACTH) or adrenal androgens are assumed to have a causative role. To clarify the role of ACTH and androgen in the pathogenesis of myelolipoma, we report a case of giant adrenal myelolipoma in a patient with poorly controlled congenital adrenal hyperplasia. A 43-year-old female was diagnosed with congenital adrenal hyperplasia at 6 years of age because of ambiguous genitalia. She had high plasma ACTH and 17-hydroxyprogesterone levels. Abdominal computed tomography showed a huge mass on the left adrenal gland, and an enlarged right adrenal mass. Genetic testing for CYP21A2 was performed and revealed that her genotype was IVS2-13A/C>G/I172N. Adrenalectomy for the left-side tumor was performed. Histological study revealed that the tumor consisted of fat cells and myeloid components, findings compatible with adrenal myelolipoma. Neither ACTH receptors nor androgen receptor was over-expressed in the tumor. Our finding that the tumor did not over-express ACTH or androgen receptor suggests a limited direct role for these hormones in the development of the myelolipoma.


Subject(s)
Adrenal Gland Neoplasms/metabolism , Adrenocorticotropic Hormone/biosynthesis , Myelolipoma/metabolism , Receptors, Androgen/biosynthesis , Steroid 21-Hydroxylase/genetics , Adrenal Gland Neoplasms/enzymology , Adrenal Gland Neoplasms/pathology , Adrenal Hyperplasia, Congenital/complications , Adrenal Hyperplasia, Congenital/pathology , Adrenalectomy , Adrenocorticotropic Hormone/blood , Adult , Female , Genitalia, Female/abnormalities , Genitalia, Female/surgery , Humans , Myelolipoma/enzymology , Myelolipoma/pathology , Receptors, Melanocortin/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Tomography, X-Ray Computed
8.
Biopolymers ; 90(5): 671-82, 2008.
Article in English | MEDLINE | ID: mdl-18655141

ABSTRACT

Backbone cyclization (BC) and N-methylation have been shown to enhance the activity and/or selectivity of biologically active peptides and improve metabolic stability and intestinal permeability. In this study, we describe the synthesis, structure-activity relationship (SAR) and intestinal metabolic stability of a backbone cyclic peptide library, BL3020, based on the linear alpha-Melanocyte stimulating hormone analog Phe-D-Phe-Arg-Trp-Gly. The drug lead, BL3020-1, selected from the BL3020 library (compound 1) has been shown to inhibit weight gain in mice following oral administration. Another member of the BL3020 library, BL3020-17, showed improved biological activity towards the mMC4R, in comparison to BL3020-1, although neither were selective for MC4R or MC5R. N-methylation, which restrains conformational freedom while increasing metabolic stability beyond that which is imparted by BC, was used to find analogs with increased selectivity. N-methylated backbone cyclic libraries were synthesized based on the BL3020 library. SAR studies showed that all the N-methylated backbone cyclic peptides demonstrated reduced biological activity and selectivity for all the analyzed receptors. N-methylation of active backbone cyclic peptides destabilized the active conformation or stabilized an inactive conformation, rendering the peptides biologically inactive. N-methylation of backbone cyclic peptides maintained stability to degradation by intestinal enzymes.


Subject(s)
Melanocortins/chemistry , Melanocortins/metabolism , Peptide Library , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/metabolism , Animals , Cell Line , Cyclization , Humans , Methylation , Mice , Protein Conformation , Receptors, Melanocortin/biosynthesis , Receptors, Melanocortin/genetics , Structure-Activity Relationship
9.
Am J Physiol Endocrinol Metab ; 295(2): E350-5, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18505834

ABSTRACT

Menopause and premature gonadal steroid deficiency are associated with increases in fat mass and body weight. Ovariectomized (OVX) mice also show reduced locomotor activity. Glucose-dependent-insulinotropic-polypeptide (GIP) is known to play an important role both in fat metabolism and locomotor activity. Therefore, we hypothesized that the effects of estrogen on the regulation of body weight, fat mass, and spontaneous physical activity could be mediated in part by GIP signaling. To test this hypothesis, C57BL/6 mice and GIP-receptor knockout mice (Gipr(-/-)) were exposed to OVX or sham operation (n = 10 per group). The effects on body composition, markers of insulin resistance, energy expenditure, locomotor activity, and expression of hypothalamic anorexigenic and orexigenic factors were investigated over 26 wk in all four groups of mice. OVX wild-type mice developed obesity, increased fat mass, and elevated markers of insulin resistance as expected. This was completely prevented in OVX Gipr(-/-) animals, even though their energy expenditure and spontaneous locomotor activity levels did not significantly differ from those of OVX wild-type mice. Cumulative food intake in OVX Gipr(-/-) animals was significantly reduced and associated with significantly lower hypothalamic mRNA expression of the orexigenic neuropeptide Y (NPY) but not of cocaine-amphetamine-related transcript (CART), melanocortin receptors (MCR-3 and MCR-4), or thyrotropin-releasing hormone (TRH). GIP receptors thus interact with estrogens in the hypothalamic regulation of food intake in mice, and their blockade may carry promising potential for the prevention of obesity in gonadal steroid deficiency.


Subject(s)
Energy Metabolism/physiology , Gastric Inhibitory Polypeptide/metabolism , Obesity/metabolism , Receptors, Gastrointestinal Hormone/deficiency , Animals , Body Composition/physiology , Eating/physiology , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/physiology , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Neuropeptide Y/biosynthesis , Neuropeptide Y/genetics , Obesity/etiology , Obesity/genetics , Obesity/prevention & control , Ovariectomy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Receptors, Gastrointestinal Hormone/genetics , Receptors, Gastrointestinal Hormone/metabolism , Receptors, Melanocortin/biosynthesis , Receptors, Melanocortin/genetics , Reverse Transcriptase Polymerase Chain Reaction , Thyrotropin-Releasing Hormone/biosynthesis , Thyrotropin-Releasing Hormone/genetics
10.
J Neurosci ; 24(45): 10176-81, 2004 Nov 10.
Article in English | MEDLINE | ID: mdl-15537888

ABSTRACT

Disruption of melanocortin (MC) signaling, such as by ectopic Agouti overexpression, leads to an obesity syndrome with hyperphagia, obesity, and accelerated body weight gain during high-fat diet. To investigate where in the brain disruption of MC signaling results in obesity, long-term Agouti expression was induced after local injections of recombinant adeno-associated viral particles in selected brain nuclei of adult rats. Agouti expression in the paraventricular nucleus, a hypothalamic region with a high density of MC receptors, induced acute onset hyperphagia and rapid weight gain that persisted for at least 6 weeks. In contrast, obesity and hyperphagia developed with a 3 week delay when Agouti was expressed in the dorsal medial hypothalamus. Agouti expression in the lateral hypothalamus (LH) did not affect food intake and body weight during regular diet, despite the presence of MC receptors in this region. However, during exposure to a high-fat diet, animals with Agouti expression in the LH exhibited a marked increase in body weight. Here we show that the LH is important for the protection against diet-induced obesity by controlling caloric intake during consumption of a high-fat diet. Together, this study provides evidence that different aspects of the Agouti-induced obesity syndrome, such as hyperphagia and diet responsiveness, are mediated by distinct brain regions and opens challenging opportunities for further understanding of pathophysiological processes in the development of the obesity syndrome.


Subject(s)
Hyperphagia/physiopathology , Hypothalamus/physiopathology , Intercellular Signaling Peptides and Proteins/physiology , Obesity/physiopathology , Agouti Signaling Protein , Agouti-Related Protein , Animals , Cell Line , Dietary Fats/toxicity , Energy Intake , Humans , Hyperphagia/genetics , Hypothalamus, Middle/physiopathology , Intercellular Signaling Peptides and Proteins/genetics , Male , Neuropeptide Y/biosynthesis , Neuropeptide Y/genetics , Obesity/genetics , Obesity/prevention & control , Organ Specificity , Paraventricular Hypothalamic Nucleus/physiopathology , Proteins/genetics , Proteins/metabolism , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Rats, Wistar , Receptors, Melanocortin/biosynthesis , Receptors, Melanocortin/physiology , Recombinant Fusion Proteins/physiology , Weight Gain/drug effects , Weight Gain/genetics
11.
Br J Pharmacol ; 143(5): 626-37, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15466451

ABSTRACT

1 Melanocortin (MC) receptors are widely distributed throughout the body of chicken, like in mammals, and participate in a wide range of physiological functions. 2 To clarify the pharmacological impact of ligands acting in the MC system, we expressed the chicken MC1, MC2, MC3, MC4 and MC5 (cMC1-5) receptors in eukaryotic cells and performed comprehensive pharmacological characterization of the potency of endogenous and synthetic melanocortin peptides. 3 Remarkably, the cMC receptors displayed high affinity for ACTH-derived peptides and in general low affinity for alpha-MSH. It is evident that not only the cMC2 receptor but also the other cMC receptors interact with ACTH-derived peptide through an epitope beyond the sequence of alpha-MSH. 4 The synthetic ligand MTII was found to be a potent agonist whereas HS024 was a potent antagonist at the cMC4 receptor, indicating that these ligands are suitable for physiological studies in chicken. 5 We also show the presence of prohormone convertase 1 (PC1) and PC2 genes in chicken, and that these peptides are coexpressed with proopiomelanocortin (POMC) in various tissues.


Subject(s)
Adrenocorticotropic Hormone/metabolism , Chickens/metabolism , Peptides/metabolism , Receptors, Melanocortin/drug effects , Receptors, Melanocortin/metabolism , Adrenocorticotropic Hormone/analogs & derivatives , Adrenocorticotropic Hormone/pharmacology , Animals , Binding, Competitive/drug effects , Cell Line , Cloning, Molecular , Cyclic AMP/metabolism , Humans , Kinetics , Peptides/pharmacology , Phylogeny , Pro-Opiomelanocortin/metabolism , Proprotein Convertase 1/genetics , Proprotein Convertase 1/metabolism , Proprotein Convertase 2/genetics , Proprotein Convertase 2/metabolism , Radioligand Assay , Receptors, Melanocortin/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Tissue Distribution , Transfection , alpha-MSH/pharmacology
12.
Eur J Pharmacol ; 482(1-3): 85-94, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14660008

ABSTRACT

The aim of this study was to characterise the expression of the melanocortin system in the normal and injured rat visual system. Using real-time polymerase chain reaction and immunohistochemistry, we detected melanocortin MC(3), MC(4) and MC(5) receptors and proopiomelanocortin in adult retina and superior colliculus. Melanocortin MC(4) receptor mRNA was the most abundant receptor. Melanocortin MC(3), MC(4) and MC(5) receptors were localised to the ganglion cell and inner nuclear layers and the melanocortin MC(3) and MC(4) receptors were localised to retinal ganglion cells. Transection of the optic nerve leads to ganglion cell death and both melanocortin receptor and proopiomelanocortin expression decreased in superior colliculus after transection whereas the expression was unchanged or even increased in the retina. alpha-Melanocyte-stimulating hormone elicited neurite outgrowth from embryonic retinal explants. Together, these data implicate a role for the melanocortin system in the adult rat retina and that melanocortins can stimulate neurite growth from retinal neurons.


Subject(s)
Optic Nerve Injuries/metabolism , Pro-Opiomelanocortin/biosynthesis , Receptors, Melanocortin/biosynthesis , Retina/metabolism , Superior Colliculi/metabolism , Animals , Chick Embryo , Female , Optic Nerve Injuries/genetics , Pro-Opiomelanocortin/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Rats , Receptor, Melanocortin, Type 3/biosynthesis , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 4/biosynthesis , Receptor, Melanocortin, Type 4/genetics , Receptors, Corticotropin/genetics , Receptors, Corticotropin/metabolism , Receptors, Melanocortin/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...