Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 163
Filter
1.
Vitam Horm ; 111: 147-193, 2019.
Article in English | MEDLINE | ID: mdl-31421699

ABSTRACT

The pentapeptides methionine-enkephalin and leucine-enkephalin belong to the opioid family of peptides, and the non-opiate peptide adrenocorticotropin hormone (ACTH) to the melanocortin peptide family. Enkephalins/ACTH are derived from pro-enkephalin, pro-dynorphin or pro-opiomelanocortin precursors and, via opioid and melanocortin receptors, are responsible for many biological activities. Enkephalins exhibit the highest affinity for the δ receptor, followed by the µ and κ receptors, whereas ACTH binds to the five subtypes of melanocortin receptor, and is the only member of the melanocortin family of peptides that binds to the melanocortin-receptor 2 (ACTH receptor). Enkephalins/ACTH and their receptors exhibit a widespread anatomical distribution. Enkephalins are involved in analgesia, angiogenesis, blood pressure, embryonic development, emotional behavior, feeding, hypoxia, limbic system modulation, neuroprotection, peristalsis, and wound repair; as well as in hepatoprotective, motor, neuroendocrine and respiratory mechanisms. ACTH plays a role in acetylcholine release, aggressive behavior, blood pressure, bone maintenance, hyperalgesia, feeding, fever, grooming, learning, lipolysis, memory, nerve injury repair, neuroprotection, sexual behavior, sleep, social behavior, tissue growth and stimulates the synthesis and secretion of glucocorticoids. Enkephalins/ACTH are also involved in many pathologies. Enkephalins are implicated in alcoholism, cancer, colitis, depression, heart failure, Huntington's disease, influenza A virus infection, ischemia, multiple sclerosis, and stress. ACTH plays a role in Addison's disease, alcoholism, cancer, Cushing's disease, dermatitis, encephalitis, epilepsy, Graves' disease, Guillain-Barré syndrome, multiple sclerosis, podocytopathies, and stress. In this review, we provide an updated description of the enkephalinergic and ACTH systems.


Subject(s)
Adrenocorticotropic Hormone/physiology , Enkephalins/physiology , Nervous System Physiological Phenomena , Adrenocorticotropic Hormone/analysis , Adrenocorticotropic Hormone/genetics , Amino Acid Sequence , Animals , Enkephalins/analysis , Enkephalins/genetics , Humans , Nervous System , Protein Precursors/metabolism , Receptors, Corticotropin/physiology , Receptors, Opioid/physiology , Signal Transduction , Tissue Distribution
2.
Endocr Dev ; 24: 57-66, 2013.
Article in English | MEDLINE | ID: mdl-23392095

ABSTRACT

ACTH resistance is a rare disorder typified by familial glucocorticoid deficiency (FGD), a genetically heterogeneous disease. Previously, genetic defects in FGD have been identified in the ACTH receptor gene (MC2R), its accessory protein (MRAP) and the steroidogenic acute regulatory protein gene (STAR). The defective mechanisms here are failures in ACTH ligand binding and/or receptor trafficking for MC2R and MRAP and, in the case of STAR mutations, inefficient cholesterol transport to allow steroidogenesis to proceed. Novel gene defects in FGD have recently been recognised in mini-chromosome maintenance-deficient 4 homologue (MCM4) and nicotinamide nucleotide transhydrogenase (NNT). MCM4 is one part of a DNA repair complex essential for DNA replication and genome stability, whilst NNT is involved in the glutathione redox system that protects cells against reactive oxygen species. The finding of mutations in these two genes implicates new pathogenetic mechanisms at play in FGD, and implies that the adrenal cortex is exquisitely sensitive to replicative and oxidative stresses.


Subject(s)
Adrenal Insufficiency/genetics , Signal Transduction/genetics , Steroid Metabolism, Inborn Errors/genetics , Adrenal Insufficiency/metabolism , Animals , Humans , Membrane Proteins/genetics , Metabolic Networks and Pathways/genetics , Models, Biological , Mutation/physiology , Receptor, Melanocortin, Type 2/genetics , Receptors, Corticotropin/genetics , Receptors, Corticotropin/metabolism , Receptors, Corticotropin/physiology , Signal Transduction/physiology , Steroid Metabolism, Inborn Errors/metabolism , Steroids/biosynthesis
3.
Minerva Endocrinol ; 37(2): 133-9, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22691887

ABSTRACT

Abnormalities in the cAMP/PKA signaling pathway have been linked to the formation of benign adrenal tumors, as well as a possible predisposition to adrenocortical cancer. Mutations in the G-protein coupled receptor are associated with McCune-Albright syndrome and ACTH-independent macronodular adrenal hyperplasia, while defects in cAMP-dependent protein kinase A can lead to the development of Carney's complex, as well as primary pigmented nodular adrenocortical disease (PPNAD), and micronodular adrenocortical hyperplasia (MAH). Defects in phosphodiesterases, which regulate cAMP levels, have also been demonstrated in PPNAD and MAH. The Wnt signaling pathway, which is involved in oncogenesis in a variety of tumors, has also been implicated in adrenocortical tumorigenesis. MicroRNA profiling has added to our understanding of the signaling pathways involved in tumor formation in the adrenal cortex. Will this all lead to the development of specific targets for pharmacologic therapies? In this article, we review the molecular genetics of adrenocortical tumors and refer to potential targets for pharmacologic therapy.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Carcinoma/genetics , Signal Transduction/physiology , 3',5'-Cyclic-AMP Phosphodiesterases/physiology , Adrenal Cortex Neoplasms/drug therapy , Adrenocorticotropic Hormone/physiology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma/drug therapy , Chromogranins , Cyclic AMP/physiology , Cyclic AMP-Dependent Protein Kinases/physiology , GTP-Binding Protein alpha Subunits, Gs/deficiency , GTP-Binding Protein alpha Subunits, Gs/genetics , GTP-Binding Protein alpha Subunits, Gs/physiology , Humans , MicroRNAs/physiology , Molecular Targeted Therapy , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Receptors, Corticotropin/deficiency , Receptors, Corticotropin/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Wnt Proteins/physiology
4.
Ann N Y Acad Sci ; 1192: 110-6, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20392225

ABSTRACT

Adrenocorticotropin (ACTH) is one of several peptide hormones derived from a larger molecule, proopiomelanocortin (POMC). ACTH is a classic endocrine hormone, processed and secreted from the pituitary to stimulate cortisol production from the fasciculata cells in the adrenal gland. However, ACTH is also produced by other cells, including macrophages, at many sites in the body. ACTH binds to a specific member of the melanocortin receptor family, the MC2R. MC2R is expressed in osteoblastic cells in vivo, as shown by in situ hybridization. MC2R expression is strongest at sites of active bone deposition, and thus ACTH response probably varies with osteoblastic activity or stage of osteoblast differentiation. In vitro ACTH stimulates proliferation of osteoblasts in a dose-dependent manner. ACTH at 10 nM increases collagen I mRNA in the osteoblastic cell line SaOs2, although at lower concentrations ACTH may oppose osteoblast differentiation. ACTH is thus, at high concentrations, anabolic for the osteoblast, and it is highly likely that the hormone has concentration-dependent effects on bone metabolism in vivo.


Subject(s)
Adrenocorticotropic Hormone/physiology , Bone and Bones/anatomy & histology , Adrenocorticotropic Hormone/metabolism , Adrenocorticotropic Hormone/pharmacology , Animals , Bone Density/drug effects , Bone Density/genetics , Bone Density/physiology , Bone and Bones/drug effects , Bone and Bones/metabolism , Humans , Models, Biological , Organ Size/drug effects , Organ Size/genetics , Paracrine Communication/drug effects , Paracrine Communication/genetics , Paracrine Communication/physiology , Pituitary Hormones/pharmacology , Pituitary Hormones/physiology , Receptors, Corticotropin/genetics , Receptors, Corticotropin/metabolism , Receptors, Corticotropin/physiology
5.
J Clin Endocrinol Metab ; 94(3): 750-6, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19066304

ABSTRACT

CONTEXT: Primary adrenal Cushing's syndrome can result from the aberrant adrenal expression of several hormone receptors; this mechanism has not been explored in detail in aldosterone-producing tumors. OBJECTIVE: The objective of the study was to evaluate a 56-yr-old male patient with an aldosteronoma for the regulation of aldosterone secretion by aberrant hormone receptors. RESULTS: Renin-independent stimulation of aldosterone secretion was observed in vivo after a mixed meal, oral glucose, or administration of glucose-dependent insulinotropic peptide (GIP), vasopressin, and tegaserod. The mixed meal-mediated stimulation of aldosterone was not present in five other cases of aldosteronoma. A smaller response of aldosterone after GIP infusion was observed in a normal subject. Aldosterone secretion was stimulated by GIP in primary cultures of this patient's aldosteronoma. Increased expression of GIP receptor was found in this aldosteronoma by real-time RT-PCR and immunohistochemistry. The GIP receptor protein was also found at lower levels in zona glomerulosa cells of the normal adjacent adrenal gland. Increased expression of serotonin 4 and ACTH receptors was also present in this aldosteronoma. CONCLUSIONS: This case report provides new evidence of the implication of aberrant hormone receptors in the regulation of this aldosteronoma and suggests that further detailed studies of the role of aberrant hormone receptors in this frequent pathology should be undertaken.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Adrenocortical Adenoma/metabolism , Aldosterone/metabolism , Receptors, Gastrointestinal Hormone/physiology , Adrenocorticotropic Hormone/pharmacology , Gastric Inhibitory Polypeptide/pharmacology , Humans , Male , Middle Aged , Receptors, Corticotropin/physiology , Receptors, Gastrointestinal Hormone/analysis , Receptors, Serotonin, 5-HT4/physiology , Receptors, Vasopressin/physiology , Reverse Transcriptase Polymerase Chain Reaction
6.
Physiol Genomics ; 35(3): 296-304, 2008 Nov 12.
Article in English | MEDLINE | ID: mdl-18796533

ABSTRACT

The vacuolated lens (vl) mouse mutant arose spontaneously on the C3H/HeSn background and exhibits neural tube defects (NTDs), congenital cataract, and occasionally a white belly spot. We previously reported that 1) the vl phenotypes are due to a mutation in an orphan G protein-coupled receptor (GPCR), Gpr161; 2) the penetrance of the vl NTD and cataract phenotypes are affected by genetic background, allowing three unlinked quantitative trait loci (QTL) to be mapped (modifiers of vacuolated lens, Modvl1-3); and 3) phenotype-based bioinformatics followed by genetic and molecular analysis identified a lens-specific transcription factor that contributes to the cataract-modifying effect of Modvl3. We now extend this analysis in three ways. First, using the Gpr161 mutation to unequivocally identify mutant adults and embryos, we determined that approximately 50% of vl/vl NTD-affected embryos die during development. Second, the MOLF/Ei genetic background suppresses this embryonic lethality but increases the incidence of the adult belly spot phenotype. Additional QTL analysis was performed, and two novel modifiers were mapped [Modvl4, logarithm of odds ratio (LOD) 4.4; Modvl5, LOD 5.0]. Third, phenotype-based bioinformatics identified candidate genes for these modifiers including two GPCRs that cause NTD or skin/pigmentation defects (Modvl4: Frizzled homolog 6; Modvl5: Melanocortin 5 receptor). Because GPCRs form oligomeric complexes, these genes were resequenced and nonsynonymous coding variants were identified. Bioinformatics and protein modeling suggest that these variants may be functional. Our studies further establish vl as a multigenic mouse model for NTDs and identify additional QTL that interact with Gpr161 to regulate neurulation.


Subject(s)
Lens, Crystalline/metabolism , Mutation , Neural Tube Defects/genetics , Quantitative Trait Loci/genetics , Amino Acid Sequence , Animals , Computational Biology , Disease Models, Animal , Female , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Frizzled Receptors/physiology , Genotype , Lens, Crystalline/pathology , Male , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Molecular Sequence Data , Phenotype , Polymorphism, Genetic , Receptors, Corticotropin/genetics , Receptors, Corticotropin/metabolism , Receptors, Corticotropin/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Receptors, G-Protein-Coupled/physiology , Receptors, Melanocortin , Sequence Homology, Amino Acid
7.
Prog Brain Res ; 170: 29-39, 2008.
Article in English | MEDLINE | ID: mdl-18655869

ABSTRACT

Vasopressin (VP) secreted from parvocellular neurons of the hypothalamic paraventricular nucleus (PVN) stimulates pituitary adrenocorticotropic hormone (ACTH) secretion, through interaction with receptors of the V1b subtype (V1bR) in the pituitary corticotroph, mainly by potentiating the stimulatory effects of corticotrophin-releasing hormone (CRH). Chronic stress paradigms associated with corticotroph hyperresponsiveness lead to preferential expression of hypothalamic VP over CRH and upregulation of pituitary V1bR, suggesting that VP has a primary role during adaptation of the hypothalamic pituitary adrenal (HPA) axis to long-term stimulation. However, studies using pharmacological or genetic ablation of V1bR have shown that VP is required for full ACTH responses to some stressors, but not for the sensitization of ACTH responses to a novel stress observed during chronic stress. Studies using minipump infusion of a peptide V1 antagonist in long-term adrenalectomized rats have revealed that VP mediates proliferative responses in the pituitary. Nevertheless, only a minor proportion of cells undergoing mitogenesis co-express markers for differentiated corticotrophs or precursors, suggesting that new corticotrophs are recruited from yet undifferentiated cells. The overall evidence supports a limited role of VP regulating acute ACTH responses to some acute stressors and points to cell proliferation and pituitary remodelling as alternative roles for the marked increases in parvocellular vasopressinergic activity during prolonged activation of the HPA axis.


Subject(s)
Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/physiopathology , Vasopressins/metabolism , Adrenocorticotropic Hormone/metabolism , Anesthetics/therapeutic use , Animals , Chronic Disease , Corticotropin-Releasing Hormone/metabolism , Humans , Paraventricular Hypothalamic Nucleus/physiology , Receptors, Corticotropin/physiology , Receptors, Vasopressin/physiology , Rodentia , Stress, Psychological/physiopathology
8.
Endocr Dev ; 13: 99-116, 2008.
Article in English | MEDLINE | ID: mdl-18493136

ABSTRACT

Familial glucocorticoid deficiency (FGD) and triple A syndrome belong to a rare group of autosomal recessive disorders characterized by adrenocorticotropin (ACTH) insensitivity. Unlike triple A syndrome which presents a range of clinical features, FGD is solely characterized by glucocorticoid deficiency. ACTH regulates steroid biosynthesis in the adrenal cortex by exerting its effects via the ACTH receptor (melanocortin- 2 receptor, MC2R). In FGD, mutations in the MC2R account for only approximately 25% of cases (FGD type 1). The inability to express a functional MC2R in non-adrenal cell lines had implied the presence of an adrenal specific accessory factor(s), essential for MC2R expression. More recently, this factor was identified as melanocortin receptor accessory protein (MRAP). Mutations in MRAP account for 20% of cases (FGD type 2). Like the receptor activity-modifying proteins (RAMPs) and receptor transporter proteins (RTPs), which are well-characterized accessory proteins for G-protein-coupled receptors (GPCRs), MRAP is a small single transmembrane domain protein. MRAP is essential for the functional expression of the MC2R. About 55% of FGD cases have no identifiable gene defect, implying the involvement of additional genes. This chapter briefly describes the clinical and biochemical features of ACTH resistance syndromes. However, we will focus on the recent progress made towards understanding the molecular defect underlying these conditions, in particular the interaction of MC2R and MRAP.


Subject(s)
Adrenal Insufficiency/etiology , Adrenocorticotropic Hormone/physiology , Adrenal Insufficiency/genetics , Adrenocorticotropic Hormone/chemistry , Adrenocorticotropic Hormone/pharmacology , Animals , Glucocorticoids/deficiency , Humans , Membrane Proteins/genetics , Models, Biological , Peptide Fragments/pharmacology , Receptor, Melanocortin, Type 2/genetics , Receptor, Melanocortin, Type 2/metabolism , Receptor, Melanocortin, Type 2/physiology , Receptors, Corticotropin/physiology , Receptors, Melanocortin/physiology , Signal Transduction/genetics , Signal Transduction/physiology , Steroids/biosynthesis , Syndrome
9.
Pediatr Res ; 63(4): 410-4, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18356748

ABSTRACT

The genetic mechanisms underlying the regulation of adrenarche are unknown. The aim of the study was to find out whether ACTH receptor (MC2R) promoter polymorphism associates with premature adrenarche (PA) and its characteristics. DNA samples of 74 prepubertal children with PA and their age- and gender-matched 97 healthy controls were genotyped for the -2 bp T/C diallelic MC2R promoter polymorphism (MC2R -2 T>C) All children were examined clinically, and hormonal measurements after an overnight fast and a low-dose ACTH stimulation test were performed. In controls, the baseline ACTH/cortisol ratio was significantly higher (p = 0.002) in subjects with the polymorphism than in the T/T group indicating decreased ACTH sensitivity. The frequency of the MC2R -2 T>C polymorphism was significantly higher in PA children with premature pubarche than in those with milder signs of PA or in control children (p = 0.04). In children with PA, the polymorphism associated with higher baseline serum dehydroepiandrosterone (p = 0.03), androstenedione (p = 0.02), plasma ACTH (p = 0.03) levels and with lower birth weight (p = 0.02). Our study provides evidence that the MC2R promoter polymorphism modulates the hypothalamo-pituitary-adrenal axis in children and may play a role in altered regulation of adrenarche.


Subject(s)
Adrenarche/genetics , Hypothalamo-Hypophyseal System/physiopathology , Pituitary-Adrenal System/physiopathology , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics , Receptors, Corticotropin/genetics , Receptors, Corticotropin/physiology , Adrenarche/physiology , Adrenocorticotropic Hormone/blood , Case-Control Studies , Child , Dehydroepiandrosterone/blood , Female , Genotype , Humans , Hydrocortisone/blood , Male , Puberty, Precocious/genetics , Puberty, Precocious/physiopathology
10.
Endocr Rev ; 27(7): 736-49, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17077189

ABSTRACT

The melanocortin system refers to a set of hormonal, neuropeptidergic, and paracrine signaling pathways that are defined by components that include the five G protein-coupled melanocortin receptors; peptide agonists derived from the proopiomelanocortin preprohormone precursor; and the endogenous antagonists, agouti and agouti-related protein. This signaling system regulates a remarkably diverse array of physiological functions including pigmentation, adrenocortical steroidogenesis, energy homeostasis, natriuresis, erectile responses, energy homeostasis, and exocrine gland secretion. There are many complex and unique aspects of melanocortin signaling, such as the existence of endogenous antagonists, the agouti proteins, that act at three of the five melanocortin receptors. However, there is an aspect of melanocortin signaling that has facilitated highly reductionist approaches aimed at understanding the physiological functions of each receptor and peptide: in contrast to many peptides, the melanocortin agonists and antagonists are expressed in a limited number of very discrete locations. Similarly, the melanocortin receptors are also expressed in a limited number of discrete locations where they tend to be involved in rather circumscribed physiological functions. This review examines my laboratory's participation in the cloning of the melanocortin receptors and characterization of their physiological roles.


Subject(s)
Melanocortins/physiology , Receptors, Melanocortin/physiology , Signal Transduction/physiology , Agouti Signaling Protein , Agouti-Related Protein , Alleles , Animals , Cachexia/genetics , Cachexia/physiopathology , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/physiology , Melanocortins/genetics , Receptor, Melanocortin, Type 3/genetics , Receptor, Melanocortin, Type 3/physiology , Receptors, Corticotropin/genetics , Receptors, Corticotropin/physiology , Receptors, Melanocortin/genetics , Signal Transduction/genetics
11.
Cell Mol Biol (Noisy-le-grand) ; 52(2): 53-9, 2006 May 30.
Article in English | MEDLINE | ID: mdl-16914087

ABSTRACT

The nervous system contributes to the mechanisms of ocular immune privilege by the constitutive presence of the immunosuppressive neuropeptide alpha-melanocyte stimulating hormone (alpha-MSH) in the eye. Alpha-MSH through the melanocortin 5 receptor (MC5r) mediates induction of CD4+ regulatory T cells that suppress in an antigen specific manner autoimmune disease. We investigated whether there was a role for MC5r expression in ocular immunity and the natural induction of regulatory T cells that emerged following resolution of experimental autoimmune uveoretinitis (EAU). Unlike wild type mice, EAU in MC5r-/- mice caused severe retinal damage, did mice expressed a not induce the emergence of ocular autoantigen regulatory immunity in the spleen, and the MC5r-/- classical memory immune response when reimmunized with ocular autoantigen. There was expression of MC5r in retinal pigment epithelial cells, in the ganglion cell and neural outer plexiform layers of healthy wild type mice retinas. The recovery of the ocular microenvironment from EAU was not dependent on the expression of MC5r, nor was the recovery dependent on the induction of CD4+ regulatory T cells (Treg cells) in the spleen. However, protection of the retina from the inflammatory damage of EAU and the induction of ocular autoantigen-responsive CD4+ Treg cells in the post EAU spleen requires expression of MC5r.


Subject(s)
Autoimmune Diseases/immunology , Autoimmunity/physiology , Receptors, Corticotropin/physiology , Retinitis/immunology , T-Lymphocytes, Regulatory/metabolism , Uveitis/immunology , Animals , Autoantigens/immunology , Autoantigens/metabolism , Autoimmune Diseases/metabolism , Autoimmune Diseases/physiopathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Eye/immunology , Eye/metabolism , Eye/pathology , Female , Immunization/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Corticotropin/genetics , Receptors, Corticotropin/metabolism , Receptors, Melanocortin , Retina/metabolism , Retinitis/metabolism , Retinitis/physiopathology , Spleen/cytology , Spleen/immunology , T-Lymphocytes, Regulatory/immunology , Time Factors , Uveitis/metabolism , Uveitis/physiopathology
12.
Nat Clin Pract Endocrinol Metab ; 2(5): 282-90, 2006 May.
Article in English | MEDLINE | ID: mdl-16932299

ABSTRACT

The action of the peptide hormone adrenocorticotropin (ACTH) to stimulate glucocorticoid production by the adrenal gland is an essential physiologic process, yet is dependent on a single unique genetic component--the ACTH receptor or melanocortin 2 receptor. Genetic defects that cause abnormalities in this receptor or in a protein required for its expression at the cell surface result in a potentially fatal disease (familial glucocorticoid deficiency). Overexpression of this receptor or inability to desensitize it is found in adrenal adenomas or hyperplasia associated with glucocorticoid overproduction (Cushing syndrome). These disorders are uncommon, but there are considerable data to show that the hypothalamo-pituitary-adrenal axis is overactive, or in some circumstances underactive, in more common situations including depressive illness and septic shock. The origin of these latter disturbances is undoubtedly complex and multifactorial, but there is good evidence that a component of this phenomenon is an altered responsiveness of the ACTH receptor to ACTH. Understanding the basis of ACTH responsiveness might, therefore, contribute to the understanding of disorders such as these and perhaps enable the hypothalamo-pituitary-adrenal axis to be manipulated beneficially in these circumstances.


Subject(s)
Endocrine System Diseases/genetics , Receptors, Corticotropin/genetics , Receptors, Corticotropin/physiology , Adrenal Cortex Neoplasms/genetics , Adrenal Glands/metabolism , Adrenocortical Adenoma/genetics , Adrenocorticotropic Hormone/blood , Adrenocorticotropic Hormone/physiology , Animals , Depressive Disorder/blood , Endocrine System Diseases/complications , Glucocorticoids/deficiency , Humans , Models, Biological , Receptor, Melanocortin, Type 2/genetics , Receptor, Melanocortin, Type 2/physiology , Shock, Septic/complications , Steroids/biosynthesis
13.
J Neuroendocrinol ; 18(3): 157-67, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16454799

ABSTRACT

The present series of studies aimed to further our understanding of the role of melanin-concentrating hormone (MCH) neurones in the central regulation of luteinising hormone (LH) release in the female rat. LH release was stimulated when MCH was injected bilaterally into the rostral preoptic area (rPOA) or medial preoptic area (mPOA), but not when injected into the zona incerta (ZI), of oestrogen-primed ovariectomised rats. In rats that were steroid-primed to generate a surge-like release of LH, MCH administration into the ZI blocked this rise in LH release: no such effect occurred when MCH was injected into the rPOA or mPOA. In vitro, MCH stimulated gonadotrophin-releasing hormone (GnRH) release from hypothalamic explants. Double-label immunohistochemistry showed GnRH-immunoreactive neurones in the vicinity of and intermingled with immunoreactive MCH processes. MCH is the endogenous ligand of the MCH type 1 receptor (MCH1-R). Previously, we have shown a role for melanocortin-5 receptors (MC5-R) in the stimulatory action of MCH, so we next investigated the involvement of both MCH1-R and/or MC5-R in mediating the actions of MCH on GnRH and hence LH release. The stimulatory action of MCH in the rPOA was inhibited by administration of antagonists for either MCH1-R or MC5-R. However, in the mPOA, the action of MCH was blocked only by the MC5-R antagonist. LH release was stimulated by an agonist for MC5-R injected into the rPOA or mPOA; this was blocked by the MC5-R antagonist but not the MCH1-R antagonist. These results indicate that both MCH1-R and MC5-R are involved in the central control of LH release by MCH.


Subject(s)
Hypothalamic Hormones/pharmacology , Luteinizing Hormone/metabolism , Melanins/pharmacology , Pituitary Hormones/pharmacology , Receptors, Corticotropin/physiology , Receptors, Somatostatin/physiology , Animals , Female , Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Immunohistochemistry , Ovariectomy , Rats , Rats, Wistar , Receptors, Melanocortin
14.
Peptides ; 26(10): 1929-35, 2005 Oct.
Article in English | MEDLINE | ID: mdl-15979763

ABSTRACT

Expression of melanocortin-4 receptor (MC4R) mRNA in developing rat limb buds, teeth, and skull bone first indicated a possible role for MC4R in bone metabolism. We therefore investigated whether MC4R mRNA was expressed in the rat osteosarcoma UMR106.06 cell line and in primary rat osteoblast cells. Reverse transcriptase-polymerase chain reaction (RT-PCR), Northern blot analysis, and ribonuclease protection assay (RPA) were used to demonstrate MC4R mRNA expression in UMR106.06 and primary osteoblast cells. MC4R mRNA was found to be localized to the periosteum of mouse bone using in situ hybridization. We also used RT-PCR and rat specific MC2R and MC5R oligonucleotides to amplify the correct size DNA fragments for these melanocortin receptors from rat primary osteoblasts. In conclusion, melanocortin receptor expression in mouse periosteum and rat osteoblasts suggests a direct role for POMC derived peptides in bone development and bone metabolism.


Subject(s)
Bone and Bones/metabolism , Peptides/physiology , Receptor, Melanocortin, Type 4/physiology , Animals , Blotting, Northern , Cell Line, Tumor , Cells, Cultured , Mice , Osteoblasts/metabolism , Peptides/genetics , Periosteum/metabolism , Pro-Opiomelanocortin/metabolism , Pro-Opiomelanocortin/physiology , RNA, Messenger/biosynthesis , Rats , Receptor, Melanocortin, Type 2/biosynthesis , Receptor, Melanocortin, Type 2/genetics , Receptor, Melanocortin, Type 2/physiology , Receptor, Melanocortin, Type 4/genetics , Receptors, Corticotropin/biosynthesis , Receptors, Corticotropin/genetics , Receptors, Corticotropin/physiology , Receptors, Melanocortin
15.
J Clin Endocrinol Metab ; 90(5): 3009-16, 2005 May.
Article in English | MEDLINE | ID: mdl-15705925

ABSTRACT

CONTEXT: Cortisol secretion is usually under the control of ACTH. However, cortisol secretion occurs in response to gastric inhibitory polypeptide (GIP) in rare cases of food-dependent Cushing's syndrome (CS). OBJECTIVE: We have investigated whether chronic ACTH stimulation or activation of the ACTH signaling pathway might be associated with GIP receptor (GIPR) expression. DESIGN: RT-PCR analysis and primary culture of hyperplastic adrenals. PATIENTS: All patients presented with CS: 20 unilateral adrenal adenomas, five Cushing's disease, one food-dependent CS. RESULTS: RT-PCR revealed GIPR expression in all hyperplastic adrenals studied. No RT-PCR product could be detected in two normal adrenals or 20 hyperfunctioning adrenal adenomas. Primary culture revealed a significant cAMP response to ACTH in all adrenals available for study (EC50, 8.1 x 10(-10) M in normals, 4.7 x 10(-10) M in Cushing's disease, and 4.4 x 10(-10) M in food-dependent disease). However, cultures taken from all four ACTH-dependent and the one food-dependent hyperplastic adrenals studied were also responsive to GIP (EC50 for cAMP, 1.3 x 10(-9) M in Cushing's disease and 4.1 x 10(-10) M in food-dependent disease). Fasting cortisol levels were low in the case of food-dependant Cushing's, rising postprandially as predicted. However, there was no trend toward low fasting or high postprandial cortisol in the other cases, suggesting that the presence of detectable GIPR alone, albeit with definite function in vitro, is not sufficient to cause clinically food-dependent CS. CONCLUSIONS: These data are consistent with the hypothesis that chronic ACTH stimulation or constitutive activation of the ACTH signaling pathway may be associated with aberrant GIPR expression, and suggest one mechanism for the pathogenesis of this phenomenon.


Subject(s)
Adrenal Cortex/pathology , Adrenocorticotropic Hormone/pharmacology , Gene Expression Regulation , Pituitary ACTH Hypersecretion/metabolism , Receptors, Gastrointestinal Hormone/genetics , Adult , Aged , Cells, Cultured , Child , Female , Gastric Inhibitory Polypeptide/pharmacology , Humans , Hydrocortisone/blood , Hyperplasia , Middle Aged , Pituitary ACTH Hypersecretion/pathology , Receptors, Corticotropin/physiology , Signal Transduction , Up-Regulation
16.
Behav Brain Res ; 152(2): 177-85, 2004 Jul 09.
Article in English | MEDLINE | ID: mdl-15196785

ABSTRACT

Siberian hamsters accumulate fat reserves in long photoperiods, but show a long-term decrease in food intake and body weight when exposed to a short winter photoperiod. The aim of this study was to determine the role of central melanocortin 3/4 receptors (MC3/4-R) in generating this chronic catabolic state by investigating the effects of SHU9119, a MC3/4-R antagonist, on food intake and associated behaviours. In adult male hamsters, intra-cerebroventricular infusions of SHU9119 significantly increased food intake in a dose-dependent manner. The time course of action was slow, food intake being increased between 4 and 24 h after intra-cerebroventricular administration. A similar degree of increase in food intake occurred in fat hamsters in long days and in lean hamsters chronically exposed to short days. Intra-cerebroventricular treatment with MTII (a MC3/4-R agonist) significantly decreased food intake for up to 24 h after treatment, and SHU9119 reversed these suppressive effects between 4 and 24 h after treatment, a similar time course to that observed when SHU9119 was administered alone. We conclude that endogenous melanocortin peptides acting via MC3/4-R are involved in the regulation of food intake in hamsters in both anabolic and catabolic states, but these acute studies do not provide evidence that increased activity of this hypothalamic system underlies the seasonal decrease in food intake that contributes to the long-term catabolic state in short days.


Subject(s)
Eating/drug effects , Feeding Behavior/drug effects , Melanocyte-Stimulating Hormones/pharmacology , Obesity/physiopathology , Receptors, Corticotropin/antagonists & inhibitors , alpha-MSH/analogs & derivatives , Analysis of Variance , Animals , Body Weight/drug effects , Body Weight/physiology , Cricetinae , Dose-Response Relationship, Drug , Drug Interactions , Grooming/drug effects , Injections, Intraventricular/methods , Male , Motor Activity/drug effects , Peptides, Cyclic/pharmacology , Photoperiod , Receptors, Corticotropin/physiology , Time Factors , alpha-MSH/pharmacology
17.
Horm Behav ; 45(1): 58-63, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14733892

ABSTRACT

The endogenous melanocortin, alpha-melanocyte-stimulating hormone (alpha-MSH), is a neurohormone secreted by the neurointermediate lobe of the pituitary. Alpha-MSH promotes intermale aggression in mice by influencing pheromone secretion, but the role of specific melanocortin receptors has not been determined. We assessed mice made deficient in the gene for the melanocortin-5 receptor (MC5R) to determine its role in pheromone-regulated behavior. In heterotypic pairs assessed in the social interaction test (SIT), MC5R-deficient mice exhibited less aggressive behavior and more defensive behavior than their wild-type opponents. By contrast, when assessed in homotypic pairs and against stimulus animals in the SIT, MC5R-deficient and wild-type mice behaved similarly. Moreover, urine from MC5R deficient mice stimulated more aggression than did urine from wild-type mice. The results suggest that MC5R deficiency disinhibits an aggression-suppressing pheromonal signal.


Subject(s)
Aggression/physiology , Behavior, Animal/physiology , Pheromones/physiology , Receptors, Corticotropin/deficiency , Receptors, Corticotropin/physiology , Social Behavior , Animals , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Odorants , Receptors, Melanocortin
19.
Curr Drug Targets ; 4(7): 586-97, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14535656

ABSTRACT

The melanocortins are a group of small protein hormones derived by post-translational cleavage of the proopiomelanocortin (POMC) gene product. The known melanocortin hormones include alpha-melanocyte stimulating hormone (MSH), beta-MSH, gamma-MSH and adrenocorticotropic hormone (ACTH). Five melanocortin receptors (MCIR through to MC5R) have been identified and most of these show tissue-specific expression patterns, as well as different binding affinities for each of the melanocortin hormones. The central melanocortin system consists of alpha-MSH, agouti-related protein (AGRP), MC3R and MC4R. AGRP and alpha-MSH are believed to be the natural antagonist and agonist respectively of MC3R and MC4R. This central melanocortin system is thought to play a fundamental role in the control of feeding and body weight. Knock-out mice models and genetic studies have pointed to the importance of the melanocortins in complex human pathways such as pigmentation, lipolysis, food intake, thermogenesis, sexual behaviour, memory and inflammatory response. Recently the melanocortins and their receptors have been the target for drug-based treatment of human physiological processes. MC3R and MC4R are likely targets for controlling body weight; MCIR may be used in the treatment of inflammation and MC2R for the treatment of glucocortical deficiency. A role for MCSR still remains unclear, but the evidence suggests an exocrine gland function.


Subject(s)
Melanocyte-Stimulating Hormones , Receptors, Corticotropin , Agouti Signaling Protein , Agouti-Related Protein , Animals , Humans , Intercellular Signaling Peptides and Proteins/physiology , Melanocyte-Stimulating Hormones/antagonists & inhibitors , Melanocyte-Stimulating Hormones/physiology , Proteins/physiology , Receptor, Melanocortin, Type 2 , Receptor, Melanocortin, Type 3 , Receptor, Melanocortin, Type 4 , Receptors, Corticotropin/antagonists & inhibitors , Receptors, Corticotropin/physiology , Receptors, Melanocortin
20.
ANZ J Surg ; 73(9): 727-38, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12956790

ABSTRACT

Adrenal masses are a common problem affecting 3-7% of the population. The majority turn out to be benign adrenocortical adenomas, which may be functional or non-functional. Much more rarely, these masses represent a primary adrenal carcinoma. It is becoming increasingly recognized that of the benign functioning adenomas or hyperplasias, the majority will hypersecrete aldosterone and this will be more frequently detected when hypertensive populations are screened for this disease. In contrast, the incidence of primary adrenocortical carcinoma has remained steady and for this disease, surgery represents the mainstay of treatment. The advent of laparoscopic adrenal surgery has lowered the threshold size for recommending surgery for asymptomatic adrenal masses and as such, an increased proportion of adrenocortical cancers are being resected and detected at an earlier stage. Recent progress has been made in our understanding of the key genetic changes which underpin the biology of this disease. Progression from adrenal adenoma to carcinoma involves a monoclonal proliferation of cells which, among other defects, have undergone chromosomal duplication at the 11p15.5 locus leading to overexpression of the IGF2 gene and abrogation of expression of the CDKN1C and H19 genes. TP53 is involved in progression to carcinoma in a subset of patients and the frequency of ACTH receptor deletion needs to be more fully explored. Other key oncogenes and tumour suppressor genes remain to be identified although the chromosomal loci in which they lie can be identified at 17p, 1p, 2p16 and 11q13 for tumour suppressor genes and chromosomes 4, 5 and 12 for oncogenes.


Subject(s)
Adrenal Cortex Neoplasms , Adrenal Cortex Neoplasms/genetics , Adrenal Cortex Neoplasms/physiopathology , Adrenal Cortex Neoplasms/surgery , Chromosomes, Human, Pair 11/genetics , Genes, Tumor Suppressor/physiology , Humans , Hydrocortisone/blood , Hyperaldosteronism/etiology , Hyperaldosteronism/physiopathology , Prognosis , Receptors, Corticotropin/physiology
SELECTION OF CITATIONS
SEARCH DETAIL