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










Publication year range
1.
Endocrinology ; 165(7)2024 May 27.
Article in English | MEDLINE | ID: mdl-38815068

ABSTRACT

The growth hormone secretagogue receptor (GHSR), primarily known as the receptor for the hunger hormone ghrelin, potently controls food intake, yet the specific Ghsr-expressing cells mediating the orexigenic effects of this receptor remain incompletely characterized. Since Ghsr is expressed in gamma-aminobutyric acid (GABA)-producing neurons, we sought to investigate whether the selective expression of Ghsr in a subset of GABA neurons is sufficient to mediate GHSR's effects on feeding. First, we crossed mice that express a tamoxifen-dependent Cre recombinase in the subset of GABA neurons that express glutamic acid decarboxylase 2 (Gad2) enzyme (Gad2-CreER mice) with reporter mice, and found that ghrelin mainly targets a subset of Gad2-expressing neurons located in the hypothalamic arcuate nucleus (ARH) and that is predominantly segregated from Agouti-related protein (AgRP)-expressing neurons. Analysis of various single-cell RNA-sequencing datasets further corroborated that the primary subset of cells coexpressing Gad2 and Ghsr in the mouse brain are non-AgRP ARH neurons. Next, we crossed Gad2-CreER mice with reactivable GHSR-deficient mice to generate mice expressing Ghsr only in Gad2-expressing neurons (Gad2-GHSR mice). We found that ghrelin treatment induced the expression of the marker of transcriptional activation c-Fos in the ARH of Gad2-GHSR mice, yet failed to induce food intake. In contrast, food deprivation-induced refeeding was higher in Gad2-GHSR mice than in GHSR-deficient mice and similar to wild-type mice, suggesting that ghrelin-independent roles of GHSR in a subset of GABA neurons is sufficient for eliciting full compensatory hyperphagia in mice.


Subject(s)
Arcuate Nucleus of Hypothalamus , Food Deprivation , GABAergic Neurons , Ghrelin , Glutamate Decarboxylase , Hyperphagia , Receptors, Ghrelin , Animals , Male , Mice , GABAergic Neurons/metabolism , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Hyperphagia/metabolism , Ghrelin/metabolism , Ghrelin/pharmacology , Arcuate Nucleus of Hypothalamus/metabolism , Food Deprivation/physiology , Glutamate Decarboxylase/metabolism , Glutamate Decarboxylase/genetics , Mice, Transgenic , Agouti-Related Protein/metabolism , Agouti-Related Protein/genetics , Mice, Inbred C57BL
2.
Endocrinology ; 164(11)2023 09 23.
Article in English | MEDLINE | ID: mdl-37823477

ABSTRACT

The hormone ghrelin displays several well-characterized functions, including some with pharmaceutical interest. The receptor for ghrelin, the growth hormone secretagogue receptor (GHSR), is expressed in the hypothalamic paraventricular nucleus (PVH), a critical hub for the integration of metabolic, neuroendocrine, autonomic, and behavioral functions. Here, we performed a neuroanatomical and functional characterization of the neuronal types mediating ghrelin actions in the PVH of male mice. We found that fluorescent ghrelin mainly labels PVH neurons immunoreactive for nitric oxide synthase 1 (NOS1), which catalyze the production of nitric oxide [NO]). Centrally injected ghrelin increases c-Fos in NOS1 PVH neurons and NOS1 phosphorylation in the PVH. We also found that a high dose of systemically injected ghrelin increases the ghrelin level in the cerebrospinal fluid and in the periventricular PVH, and induces c-Fos in NOS1 PVH neurons. Such a high dose of systemically injected ghrelin activates a subset of NOS1 PVH neurons, which do not express oxytocin, via an arcuate nucleus-independent mechanism. Finally, we found that pharmacological inhibition of NO production fully abrogates ghrelin-induced increase of calcium concentration in corticotropin-releasing hormone neurons of the PVH whereas it partially impairs ghrelin-induced increase of plasma glucocorticoid levels. Thus, plasma ghrelin can directly target a subset of NO-producing neurons of the PVH that is involved in ghrelin-induced activation of the hypothalamic-pituitary-adrenal neuroendocrine axis.


Subject(s)
Corticotropin-Releasing Hormone , Ghrelin , Mice , Male , Animals , Corticotropin-Releasing Hormone/metabolism , Ghrelin/pharmacology , Ghrelin/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Hypothalamo-Hypophyseal System/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Neurons/metabolism
3.
Cell Mol Life Sci ; 79(5): 277, 2022 May 04.
Article in English | MEDLINE | ID: mdl-35504998

ABSTRACT

OBJECTIVE: Prolonged fasting is a major challenge for living organisms. An appropriate metabolic response to food deprivation requires the activation of the corticotropin-releasing factor-producing neurons of the hypothalamic paraventricular nucleus (PVHCRF neurons), which are a part of the hypothalamic-pituitary-adrenal axis (HPA), as well as the growth hormone secretagogue receptor (GHSR) signaling, whose activity is up- or down-regulated, respectively, by the hormones ghrelin and the liver-expressed antimicrobial peptide 2 (LEAP2). Since ghrelin treatment potently up-regulates the HPA axis, we studied the role of GHSR in mediating food deprivation-induced activation of the PVHCRF neurons in mice. METHODS: We estimated the activation of the PVHCRF neurons, using immuno-staining against CRF and the marker of neuronal activation c-Fos in brain sections, and assessed plasma levels of corticosterone and glucose in different pharmacologically or genetically manipulated mouse models exposed, or not, to a 2-day food deprivation protocol. In particular, we investigated ad libitum fed or food-deprived male mice that: (1) lacked GHSR gene expression, (2) had genetic deletion of the ghrelin gene, (3) displayed neurotoxic ablation of the hypothalamic arcuate nucleus, (4) were centrally treated with an anti-ghrelin antibody to block central ghrelin action, (5) were centrally treated with a GHSR ligand that blocks ghrelin-evoked and constitutive GHSR activities, or (6) received a continuous systemic infusion of LEAP2(1-12). RESULTS: We found that food deprivation results in the activation of the PVHCRF neurons and in a rise of the ghrelin/LEAP2 molar ratio. Food deprivation-induced activation of PVHCRF neurons required the presence and the signaling of GHSR at hypothalamic level, but not of ghrelin. Finally, we found that preventing the food deprivation-induced fall of LEAP2 reverses the activation of the PVHCRF neurons in food-deprived mice, although it has no effect on body weight or blood glucose. CONCLUSION: Food deprivation-induced activation of the PVHCRF neurons involves ghrelin-independent actions of GHSR at hypothalamic level and requires a decrease of plasma LEAP2 levels. We propose that the up-regulation of the actions of GHSR associated to the fall of plasma LEAP2 level are physiologically relevant neuroendocrine signals during a prolonged fasting.


Subject(s)
Antimicrobial Cationic Peptides/metabolism , Food Deprivation , Paraventricular Hypothalamic Nucleus , Receptors, Ghrelin/metabolism , Animals , Corticotropin-Releasing Hormone/metabolism , Corticotropin-Releasing Hormone/pharmacology , Eating , Ghrelin/metabolism , Ghrelin/pharmacology , Hypothalamo-Hypophyseal System/metabolism , Male , Mice , Neurons/metabolism , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/metabolism , Pituitary-Adrenal System/metabolism , Receptors, Ghrelin/genetics
4.
Psychoneuroendocrinology ; 139: 105716, 2022 05.
Article in English | MEDLINE | ID: mdl-35290931

ABSTRACT

Ghrelin is a stomach-derived hormone that acts via the growth hormone secretagogue receptor (GHSR). Recent evidence suggests that some of ghrelin's actions may be mediated via the supramammillary nucleus (SuM). Not only does ghrelin bind to cells within the mouse SuM, but ghrelin also activates SuM cells and intra-SuM ghrelin administration induces feeding in rats. In the current study, we aimed to further characterize ghrelin action in the SuM. We first investigated a mouse model expressing enhanced green fluorescent protein (eGFP) under the promoter of GHSR (GHSR-eGFP mice). We found that the SuM of GHSR-eGFP mice contains a significant amount of eGFP cells, some of which express neuronal nitric oxide synthase. Centrally-, but not systemically-, injected ghrelin reached the SuM, where it induced c-Fos expression. Furthermore, a 5-day 40% calorie restriction protocol, but not a 2-day fast, increased c-Fos expression in non-eGFP+ cells of the SuM of GHSR-eGFP mice, whereas c-Fos induction by calorie restriction was not observed in GHSR-deficient mice. Exposure of satiated mice to a binge-like eating protocol also increased c-Fos expression in non-eGFP+ cells of the SuM of GHSR-eGFP mice in a GHSR-dependent manner. Finally, intra-SuM-injected ghrelin did not acutely affect food intake, locomotor activity, behavioral arousal or spatial memory but increased recognition memory. Thus, we provide a compelling neuroanatomical characterization of GHSR SuM neurons and its behavioral implications in mice.


Subject(s)
Neurons , Nitric Oxide , Receptors, Ghrelin , Animals , Ghrelin/metabolism , Hypothalamus, Posterior , Mice , Neurons/metabolism , Nitric Oxide/metabolism , Rats , Receptors, Ghrelin/metabolism , Signal Transduction
5.
Mol Metab ; 32: 69-84, 2020 02.
Article in English | MEDLINE | ID: mdl-32029231

ABSTRACT

OBJECTIVE: Arcuate nucleus (ARC) neurons producing Agouti-related peptide (AgRP) and neuropeptide Y (NPY; ARCAgRP/NPY neurons) are activated under energy-deficit states. ARCAgRP/NPY neurons innervate the hypothalamic paraventricular nucleus (PVH), and ARC→PVH projections are recognized as key regulators of food intake. Plasma ghrelin levels increase under energy-deficit states and activate ARCAgRP/NPY neurons by acting on the growth hormone secretagogue receptor (GHSR). Here, we hypothesized that activation of ARCAgRP/NPY neurons in fasted mice would promote morphological remodeling of the ARCAgRP/NPY→PVH projections in a GHSR-dependent manner. METHODS: We performed 1) fluorescent immunohistochemistry, 2) imaging of green fluorescent protein (GFP) signal in NPY-GFP mice, and 3) DiI axonal labeling in brains of ad libitum fed or fasted mice with pharmacological or genetic blockage of the GHSR signaling and then estimated the density and strength of ARCAgRP/NPY→PVH fibers by assessing the mean fluorescence intensity, the absolute area with fluorescent signal, and the intensity of the fluorescent signal in the fluorescent area of the PVH. RESULTS: We found that 1) the density and strength of ARCAgRP/NPY fibers increase in the PVH of fasted mice, 2) the morphological remodeling of the ARCAgRP/NPY→PVH projections correlates with the activation of PVH neurons, and 3) PVH neurons are not activated in ARC-ablated mice. We also found that fasting-induced remodeling of ARCAgRP/NPY→PVH fibers and PVH activation are impaired in mice with pharmacological or genetic blockage of GHSR signaling. CONCLUSION: This evidence shows that the connectivity between hypothalamic circuits controlling food intake can be remodeled in the adult brain, depending on the energy balance conditions, and that GHSR activity is a key regulator of this phenomenon.


Subject(s)
Arcuate Nucleus of Hypothalamus/metabolism , Fasting/metabolism , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, Ghrelin/metabolism , Animals , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Ghrelin/deficiency
6.
Mol Cell Endocrinol ; 498: 110573, 2019 12 01.
Article in English | MEDLINE | ID: mdl-31499133

ABSTRACT

Liver-expressed antimicrobial peptide 2 (LEAP2) was recently recognized as an endogenous ligand for the growth hormone secretagogue receptor (GHSR), which also is a receptor for the hormone ghrelin. LEAP2 blocks ghrelin-induced activation of GHSR and inhibits GHSR constitutive activity. Since fluorescence-based imaging and pharmacological analyses to investigate the biology of GHSR require reliable probes, we developed a novel fluorescent GHSR ligand based on the N-terminal LEAP2 sequence, hereafter named F-LEAP2. In vitro, F-LEAP2 displayed binding affinity and inverse agonism to GHSR similar to LEAP2. In a heterologous expression system, F-LEAP2 labeling was specifically observed in the surface of GHSR-expressing cells, in contrast to fluorescent ghrelin labeling that was mainly observed inside the GHSR-expressing cells. In mice, centrally-injected F-LEAP2 reduced ghrelin-induced food intake, in a similar fashion to LEAP2, and specifically labeled cells in GHSR-expressing brain areas. Thus, F-LEAP2 represents a valuable tool to study the biology of GHSR in vitro and in vivo.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Antimicrobial Cationic Peptides/metabolism , Brain/metabolism , Fluorescent Dyes/chemistry , Ghrelin/metabolism , Kidney/metabolism , Animals , Cells, Cultured , Eating , Humans , Ligands , Mice , Mice, Inbred C57BL , Protein Domains , Signal Transduction
7.
Mol Neurobiol ; 56(6): 4120-4134, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30276663

ABSTRACT

The stomach-derived hormone ghrelin mainly acts in the brain. Studies in mice have shown that the accessibility of ghrelin into the brain is limited and that it mainly takes place in some circumventricular organs, such as the median eminence. Notably, some known brain targets of ghrelin are distantly located from the circumventricular organs. Thus, we hypothesized that ghrelin could also access the brain via the blood-cerebrospinal fluid (CSF) barrier, which consists of the choroid plexus and the hypothalamic tanycytes. Using systemic injection of ghrelin or fluorescent-ghrelin in mice, we found that cells of the blood-CSF barrier internalize these molecules. In time-response studies, we found that peripherally injected fluorescent-ghrelin quickly reaches hypothalamic regions located in apposition to the median eminence and more slowly reaches the periventricular hypothalamic parenchyma, adjacent to the dorsal part of the third ventricle. Additionally, we found that CSF ghrelin levels increase after the systemic administration of ghrelin, and that central infusions of either an anti-ghrelin antibody, which immuno-neutralizes CSF ghrelin, or a scrambled version of ghrelin, which is also internalized by cells of the blood-CSF barrier, partially impair the orexigenic effect of peripherally injected ghrelin. Thus, current evidence suggests that the blood-CSF barrier can transport circulating ghrelin into the brain, and that the access of ghrelin into the CSF is required for its full orexigenic effect.


Subject(s)
Blood-Brain Barrier/metabolism , Cerebrospinal Fluid/metabolism , Ghrelin/blood , Animals , Antibodies/metabolism , Cerebral Ventricles/metabolism , Ghrelin/administration & dosage , Ghrelin/cerebrospinal fluid , Male , Mice, Inbred C57BL , Orexins/metabolism
8.
J Med Chem ; 62(2): 965-973, 2019 01 24.
Article in English | MEDLINE | ID: mdl-30543423

ABSTRACT

The ghrelin receptor or growth hormone secretagogue receptor (GHSR) is a G-protein-coupled receptor that controls growth hormone and insulin secretion, food intake, and reward-seeking behaviors. Liver-expressed antimicrobial peptide 2 (LEAP2) was recently described as an endogenous antagonist of GHSR. Here, we present a study aimed at delineating the structural determinants required for LEAP2 activity toward GHSR. We demonstrate that the entire sequence of LEAP2 is not necessary for its actions. Indeed, the N-terminal part alone confers receptor binding and activity to LEAP2. We found that both LEAP2 and its N-terminal part behave as inverse agonists of GHSR and as competitive antagonists of ghrelin-induced inositol phosphate production and calcium mobilization. Accordingly, the N-terminal region of LEAP2 is able to inhibit ghrelin-induced food intake in mice. These data demonstrate an unexpected pharmacological activity for LEAP2 that is likely to have an important role in the control of ghrelin response under normal and pathological conditions.


Subject(s)
Antimicrobial Cationic Peptides/chemistry , Receptors, Ghrelin/agonists , Amino Acid Sequence , Animals , Antimicrobial Cationic Peptides/metabolism , Antimicrobial Cationic Peptides/pharmacology , Binding, Competitive , Drug Inverse Agonism , HEK293 Cells , Humans , Inositol Phosphates/metabolism , Islets of Langerhans/cytology , Islets of Langerhans/drug effects , Islets of Langerhans/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Binding , Rats , Receptors, Ghrelin/antagonists & inhibitors , Receptors, Ghrelin/metabolism
9.
J Neuroendocrinol ; 31(7): e12677, 2019 07.
Article in English | MEDLINE | ID: mdl-30582239

ABSTRACT

Ghrelin is a hormone produced in the gastrointestinal tract that acts via the growth hormone secretagogue receptor. In the central nervous system, ghrelin signalling is able to recruit different neuronal targets that regulate the behavioural, neuroendocrine, metabolic and autonomic effects of the hormone. Notably, several studies using radioactive or fluorescent variants of ghrelin have found that the accessibility of circulating ghrelin into the mouse brain is both strikingly low and restricted to some specific brain areas. A variety of studies addressing central effects of systemically injected ghrelin in mice have also provided indirect evidence that the accessibility of plasma ghrelin into the brain is limited. Here, we review these previous observations and discuss the putative pathways that would allow plasma ghrelin to gain access into the brain together with their physiological implications. Additionally, we discuss some potential features regarding the accessibility of plasma ghrelin into the human brain based on the observations reported by studies that investigate the consequences of ghrelin administration to humans.


Subject(s)
Brain/physiology , Ghrelin/physiology , Neurons/physiology , Animals , Blood-Brain Barrier/metabolism , Brain/metabolism , Ghrelin/blood , Humans , Neurons/metabolism
10.
Endocrinology ; 159(2): 1021-1034, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29300858

ABSTRACT

Ghrelin is a potent orexigenic peptide hormone that acts through the growth hormone secretagogue receptor (GHSR), a G protein-coupled receptor highly expressed in the hypothalamus. In vitro studies have shown that GHSR displays a high constitutive activity, whose physiological relevance is uncertain. As GHSR gene expression in the hypothalamus is known to increase in fasting conditions, we tested the hypothesis that constitutive GHSR activity at the hypothalamic level drives the fasting-induced hyperphagia. We found that refed wild-type (WT) mice displayed a robust hyperphagia that continued for 5 days after refeeding and changed their food intake daily pattern. Fasted WT mice showed an increase in plasma ghrelin levels, as well as in GHSR expression levels and ghrelin binding sites in the hypothalamic arcuate nucleus. When fasting-refeeding responses were evaluated in ghrelin- or GHSR-deficient mice, only the latter displayed an ∼15% smaller hyperphagia, compared with WT mice. Finally, fasting-induced hyperphagia of WT mice was significantly smaller in mice centrally treated with the GHSR inverse agonist K-(D-1-Nal)-FwLL-NH2, compared with mice treated with vehicle, whereas it was unaffected in mice centrally treated with the GHSR antagonists D-Lys3-growth hormone-releasing peptide 6 or JMV2959. Taken together, genetic models and pharmacological results support the notion that constitutive GHSR activity modulates the magnitude of the compensatory hyperphagia triggered by fasting. Thus, the hypothalamic GHSR signaling system could affect the set point of daily food intake, independently of plasma ghrelin levels, in situations of negative energy balance.


Subject(s)
Fasting/physiology , Ghrelin/physiology , Hyperphagia/etiology , Receptors, Ghrelin/physiology , Animals , Eating/physiology , Ghrelin/metabolism , Hyperphagia/genetics , Hyperphagia/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , Signal Transduction/genetics
11.
Int J Mol Sci ; 18(3)2017 Mar 15.
Article in English | MEDLINE | ID: mdl-28294994

ABSTRACT

Ghrelin is an octanoylated peptide that acts via its specific receptor, the growth hormone secretagogue receptor type 1a (GHSR-1a), and regulates a vast variety of physiological functions. It is well established that ghrelin is predominantly synthesized by a distinct population of endocrine cells located within the gastric oxyntic mucosa. In addition, some studies have reported that ghrelin could also be synthesized in some brain regions, such as the hypothalamus. However, evidences of neuronal production of ghrelin have been inconsistent and, as a consequence, it is still as a matter of debate if ghrelin can be centrally produced. Here, we provide a comprehensive review and discussion of the data supporting, or not, the notion that the mammalian central nervous system can synthetize ghrelin. We conclude that no irrefutable and reproducible evidence exists supporting the notion that ghrelin is synthetized, at physiologically relevant levels, in the central nervous system of adult mammals.


Subject(s)
Central Nervous System/metabolism , Ghrelin/biosynthesis , Animals , Gene Expression , Gene Knockout Techniques , Ghrelin/genetics , Humans , Neurons/metabolism , Protein Precursors/biosynthesis , Protein Precursors/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Endocrinology ; 158(5): 1436-1449, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28204197

ABSTRACT

Ghrelin is known to act on the area postrema (AP), a sensory circumventricular organ located in the medulla oblongata that regulates a variety of important physiological functions. However, the neuronal targets of ghrelin in the AP and their potential role are currently unknown. In this study, we used wild-type and genetically modified mice to gain insights into the neurons of the AP expressing the ghrelin receptor [growth hormone secretagogue receptor (GHSR)] and their role. We show that circulating ghrelin mainly accesses the AP but not to the adjacent nucleus of the solitary tract. Also, we show that both peripheral administration of ghrelin and fasting induce an increase of c-Fos, a marker of neuronal activation, in GHSR-expressing neurons of the AP, and that GHSR expression is necessary for the fasting-induced activation of AP neurons. Additionally, we show that ghrelin-sensitive neurons of the AP are mainly γ-aminobutyric acid (GABA)ergic, and that an intact AP is required for ghrelin-induced gastric emptying. Overall, we show that the capacity of circulating ghrelin to acutely induce gastric emptying in mice requires the integrity of the AP, which contains a population of GABA neurons that are a target of plasma ghrelin.


Subject(s)
Area Postrema/physiology , GABAergic Neurons/physiology , Ghrelin/blood , Animals , Area Postrema/drug effects , Fasting , GABAergic Neurons/drug effects , Gastric Emptying/drug effects , Ghrelin/administration & dosage , Ghrelin/metabolism , Male , Mice , Proto-Oncogene Proteins c-fos/genetics , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , gamma-Aminobutyric Acid/metabolism
13.
Endocrinology ; 157(10): 3946-3957, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27490185

ABSTRACT

Ghrelin exists in two forms in circulation, acyl ghrelin and des-acyl ghrelin, both of which have distinct and fundamental roles in a variety of physiological functions. Despite this fact, a large proportion of papers simply measure and refer to plasma ghrelin without specifying the acylation status. It is therefore critical to assess and state the acylation status of plasma ghrelin in all studies. In this study we tested the effect of des-acyl ghrelin administration on the hypothalamic-pituitary-adrenal axis and on anxiety-like behavior of mice lacking endogenous ghrelin and in ghrelin-O-acyltransferase (GOAT) knockout (KO) mice that have no endogenous acyl ghrelin and high endogenous des-acyl ghrelin. Our results show des-acyl ghrelin produces an anxiogenic effect under nonstressed conditions, but this switches to an anxiolytic effect under stress. Des-acyl ghrelin influences plasma corticosterone under both nonstressed and stressed conditions, although c-fos activation in the paraventricular nucleus of the hypothalamus is not different. By contrast, GOAT KO are anxious under both nonstressed and stressed conditions, although this is not due to corticosterone release from the adrenals but rather from impaired feedback actions in the paraventricular nucleus of the hypothalamus, as assessed by c-fos activation. These results reveal des-acyl ghrelin treatment and GOAT deletion have differential effects on the hypothalamic-pituitary-adrenal axis and anxiety-like behavior, suggesting that anxiety-like behavior in GOAT KO mice is not due to high plasma des-acyl ghrelin.


Subject(s)
Acyltransferases/metabolism , Anxiety/physiopathology , Ghrelin/physiology , Hypothalamo-Hypophyseal System/physiology , Pituitary-Adrenal System/physiology , Acylation , Acyltransferases/genetics , Animals , Anxiety/psychology , Female , Male , Membrane Proteins , Mice, Inbred C57BL , Mice, Knockout
14.
Psychoneuroendocrinology ; 67: 27-39, 2016 May.
Article in English | MEDLINE | ID: mdl-26874559

ABSTRACT

Previous work has established that the hormone ghrelin engages the hypothalamic-pituitary-adrenal neuroendocrine axis via activation of corticotropin-releasing factor (CRF) neurons of the hypothalamic paraventricular nucleus (PVN). The neuronal circuitry that mediates this effect of ghrelin is currently unknown. Here, we show that ghrelin-induced activation of PVN CRF neurons involved inhibition of γ-aminobutyric acid (GABA) inputs, likely via ghrelin binding sites that were localized at GABAergic terminals within the PVN. While ghrelin activated PVN CRF neurons in the presence of neuropeptide Y (NPY) receptor antagonists or in arcuate nucleus (ARC)-ablated mice, it failed to do it so in mice with ghrelin receptor expression limited to ARC agouti gene related protein (AgRP)/NPY neurons. These data support the notion that ghrelin activates PVN CRF neurons via inhibition of local GABAergic tone, in an ARC-independent manner. Furthermore, these data suggest that the neuronal circuits mediating ghrelin's orexigenic action vs. its role as a stress signal are anatomically dissociated.


Subject(s)
Arcuate Nucleus of Hypothalamus/drug effects , Corticotropin-Releasing Hormone/metabolism , Ghrelin/pharmacology , Neurons/drug effects , Neurons/metabolism , Animals , Arginine/analogs & derivatives , Arginine/pharmacology , Corticosterone/blood , GABA Antagonists , Gene Knockdown Techniques , Ghrelin/administration & dosage , Infusions, Intraventricular , Male , Mice , Muscimol/pharmacology , Neuropeptide Y/antagonists & inhibitors , Paraventricular Hypothalamic Nucleus/metabolism , Receptors, Ghrelin/drug effects , Receptors, Ghrelin/genetics , Receptors, Ghrelin/metabolism , gamma-Aminobutyric Acid/metabolism
15.
J Gen Physiol ; 146(3): 205-19, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26283199

ABSTRACT

The growth hormone secretagogue receptor type 1a (GHSR1a) has the highest known constitutive activity of any G protein-coupled receptor (GPCR). GHSR1a mediates the action of the hormone ghrelin, and its activation increases transcriptional and electrical activity in hypothalamic neurons. Although GHSR1a is present at GABAergic presynaptic terminals, its effect on neurotransmitter release remains unclear. The activities of the voltage-gated calcium channels, CaV2.1 and CaV2.2, which mediate neurotransmitter release at presynaptic terminals, are modulated by many GPCRs. Here, we show that both constitutive and agonist-dependent GHSR1a activity elicit a strong impairment of CaV2.1 and CaV2.2 currents in rat and mouse hypothalamic neurons and in a heterologous expression system. Constitutive GHSR1a activity reduces CaV2 currents by a Gi/o-dependent mechanism that involves persistent reduction in channel density at the plasma membrane, whereas ghrelin-dependent GHSR1a inhibition is reversible and involves altered CaV2 gating via a Gq-dependent pathway. Thus, GHSR1a differentially inhibits CaV2 channels by Gi/o or Gq protein pathways depending on its mode of activation. Moreover, we present evidence suggesting that GHSR1a-mediated inhibition of CaV2 attenuates GABA release in hypothalamic neurons, a mechanism that could contribute to neuronal activation through the disinhibition of postsynaptic neurons.


Subject(s)
Calcium Channels, N-Type/metabolism , Ghrelin/metabolism , Hypothalamus/physiology , Neurons/physiology , Receptors, Ghrelin/metabolism , Animals , Base Sequence , Calcium/metabolism , Calcium Signaling/physiology , Cells, Cultured , HEK293 Cells , Humans , Ion Channel Gating/physiology , Mice , Molecular Sequence Data , Rats , Rats, Sprague-Dawley , Receptors, Ghrelin/genetics
16.
Endocrinology ; 156(7): 2713-23, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25942072

ABSTRACT

Fasting down-regulates the hypothalamus-pituitary-thyroid (HPT) axis activity through a reduction of TRH synthesis in neurons of the parvocellular paraventricular nucleus of the hypothalamus (PVN). These TRH neurons project to the median eminence (ME), where TRH terminals are close to the cytoplasmic extensions of ß2 tanycytes. Tanycytes express pyroglutamyl peptidase II (PPII), the TRH-degrading ectoenzyme that controls the amount of TRH that reaches the anterior pituitary. We tested the hypothesis that regulation of ME PPII activity is another mechanism by which fasting affects the activity of the HPT axis. Semiquantitative in situ hybridization histochemistry data indicated that PPII and deiodinase 2 mRNA levels increased in tanycytes after 48 hours of fasting. This increase was transitory, followed by an increase of PPII activity in the ME, and a partial reversion of the reduction in PVN pro-TRH mRNA levels and the number of TRH neurons detected by immunohistochemistry. In fed animals, adrenalectomy and corticosterone treatment did not change ME PPII activity 72 hours later. Methimazole-induced hypothyroidism produced a profound drop in tanycytes PPII mRNA levels, which was reverted by 3 days of treatment with T4. The activity of thyroliberinase, the serum isoform of PPII, was increased at most fasting time points studied. We conclude that delayed increases in both the ME PPII as well as the thyroliberinase activities in fasted male rats may facilitate the maintenance of the deep down-regulation of the HPT axis function, despite a partial reactivation of TRH expression in the PVN.


Subject(s)
Aminopeptidases/genetics , Ependymoglial Cells/enzymology , Fasting/metabolism , Hypothalamo-Hypophyseal System/metabolism , Hypothalamus/metabolism , Median Eminence/metabolism , Pyrrolidonecarboxylic Acid/analogs & derivatives , RNA, Messenger/metabolism , Thyrotropin-Releasing Hormone/metabolism , Adrenalectomy , Aminopeptidases/drug effects , Aminopeptidases/metabolism , Animals , Antithyroid Agents/pharmacology , Corticosterone/pharmacology , Ependymoglial Cells/drug effects , Ependymoglial Cells/metabolism , Hypothalamus/cytology , Hypothalamus/drug effects , Hypothyroidism , Iodide Peroxidase/genetics , Male , Methimazole/pharmacology , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Protein Precursors/genetics , Pyrrolidonecarboxylic Acid/metabolism , RNA, Messenger/drug effects , Rats , Thyrotropin-Releasing Hormone/drug effects , Thyrotropin-Releasing Hormone/genetics , Thyroxine/pharmacology , Iodothyronine Deiodinase Type II
18.
Eur J Neurosci ; 40(5): 2755-65, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24943127

ABSTRACT

The melanocortin 4 receptor (MC4R) is a G protein-coupled receptor involved in food intake and energy expenditure regulation. MC4R activation modifies neuronal activity but the molecular mechanisms by which this regulation occurs remain unclear. Here, we tested the hypothesis that MC4R activation regulates the activity of voltage-gated calcium channels and, as a consequence, synaptic activity. We also tested whether the proposed effect occurs in the amygdala, a brain area known to mediate the anorexigenic actions of MC4R signaling. Using the patch-clamp technique, we found that the activation of MC4R with its agonist melanotan II specifically inhibited 34.5 ± 1.5% of N-type calcium currents in transiently transfected HEK293 cells. This inhibition was concentration-dependent, voltage-independent and occluded by the Gαs pathway inhibitor cholera toxin. Moreover, we found that melanotan II specifically inhibited 25.9 ± 2.0% of native N-type calcium currents and 55.4 ± 14.4% of evoked inhibitory postsynaptic currents in mouse cultured amygdala neurons. In vivo, we found that the MC4R agonist RO27-3225 increased the marker of cellular activity c-Fos in several components of the amygdala, whereas the N-type channel blocker ω conotoxin GVIA increased c-Fos expression exclusively in the central subdivision of the amygdala. Thus, MC4R specifically inhibited the presynaptic N-type channel subtype, and this inhibition may be important for the effects of melanocortin in the central subdivision of the amygdala.


Subject(s)
Amygdala/physiology , Calcium Channels, N-Type/metabolism , Presynaptic Terminals/physiology , Receptor, Melanocortin, Type 4/metabolism , Amygdala/drug effects , Animals , Calcium Channel Blockers/pharmacology , Cells, Cultured , Central Nervous System Agents/pharmacology , Cholera Toxin/pharmacology , HEK293 Cells , Humans , Inhibitory Postsynaptic Potentials/drug effects , Inhibitory Postsynaptic Potentials/physiology , Male , Mice , Peptides/pharmacology , Peptides, Cyclic/metabolism , Presynaptic Terminals/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Receptor, Melanocortin, Type 4/agonists , alpha-MSH/analogs & derivatives , alpha-MSH/metabolism , omega-Conotoxin GVIA/pharmacology
19.
Neurosci Lett ; 518(2): 86-91, 2012 Jun 19.
Article in English | MEDLINE | ID: mdl-22580206

ABSTRACT

The neuropeptide thyrotropin releasing hormone (TRH) is necessary for adequate cold-induced thermogenesis. TRH increases body temperature via both neuroendocrine and autonomic mechanisms. TRH neurons of the hypothalamic paraventricular nucleus (PVN) regulate thermogenesis through the activation of the hypothalamic-pituitary-thyroid axis during cold exposure. However, little is known about the role that TRH neurons play in mediating the sympathetic response to cold exposure. Here, we examined the response of TRH neurons of rats to cold exposure in hypothalamic regions including the PVN, the dorsomedial nucleus and the lateral hypothalamus along with areas of the ventral medulla including raphe obscurus, raphe pallidus (RPa) and parapyramidal regions. Our results using a double immunohistochemistry protocol to identify TRH and c-Fos (as a marker of cellular activity) followed by analysis of preproTRH gene expression demonstrate that only TRH neurons located in the PVN and the RPa are activated in animals exposed to short-term cold conditions.


Subject(s)
Cold Temperature , Hypothalamus/physiology , Neurons/metabolism , Thermogenesis/physiology , Thyrotropin-Releasing Hormone/metabolism , Animals , Immunohistochemistry , Male , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction
20.
PLoS One ; 7(2): e31462, 2012.
Article in English | MEDLINE | ID: mdl-22363652

ABSTRACT

Ghrelin is a stomach-derived hormone that regulates food intake and neuroendocrine function by acting on its receptor, GHSR (Growth Hormone Secretagogue Receptor). Recent evidence indicates that a key function of ghrelin is to signal stress to the brain. It has been suggested that one of the potential stress-related ghrelin targets is the CRF (Corticotropin-Releasing Factor)-producing neurons of the hypothalamic paraventricular nucleus, which secrete the CRF neuropeptide into the median eminence and activate the hypothalamic-pituitary-adrenal axis. However, the neural circuits that mediate the ghrelin-induced activation of this neuroendocrine axis are mostly uncharacterized. In the current study, we characterized in vivo the mechanism by which ghrelin activates the hypophysiotropic CRF neurons in mice. We found that peripheral or intra-cerebro-ventricular administration of ghrelin strongly activates c-fos--a marker of cellular activation--in CRF-producing neurons. Also, ghrelin activates CRF gene expression in the paraventricular nucleus of the hypothalamus and the hypothalamic-pituitary-adrenal axis at peripheral level. Ghrelin administration directly into the paraventricular nucleus of the hypothalamus also induces c-fos within the CRF-producing neurons and the hypothalamic-pituitary-adrenal axis, without any significant effect on the food intake. Interestingly, dual-label immunohistochemical analysis and ghrelin binding studies failed to show GHSR expression in CRF neurons. Thus, we conclude that ghrelin activates hypophysiotropic CRF neurons, albeit indirectly.


Subject(s)
Corticotropin-Releasing Hormone/metabolism , Ghrelin/pharmacology , Hypothalamo-Hypophyseal System/cytology , Hypothalamo-Hypophyseal System/drug effects , Neurons/cytology , Neurons/drug effects , Animals , Ghrelin/administration & dosage , Male , Mice , Mice, Inbred C57BL , Microinjections , Paraventricular Hypothalamic Nucleus/cytology , Paraventricular Hypothalamic Nucleus/drug effects , Pituitary-Adrenal System/cytology , Pituitary-Adrenal System/drug effects , Protein Binding/drug effects , Receptors, Ghrelin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...