Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Cell Rep Med ; 3(11): 100810, 2022 11 15.
Article in English | MEDLINE | ID: mdl-36384093

ABSTRACT

Glucagon analogs show promise as components of next-generation, multi-target, anti-obesity therapeutics. The biology of chronic glucagon treatment, in particular, its ability to induce energy expenditure and weight loss, remains poorly understood. Using a long-acting glucagon analog, G108, we demonstrate that glucagon-mediated body weight loss is intrinsically linked to the hypoaminoacidemia associated with its known amino acid catabolic action. Mechanistic studies reveal an energy-consuming response to low plasma amino acids in G108-treated mice, prevented by dietary amino acid supplementation and mimicked by a rationally designed low amino acid diet. Therefore, low plasma amino acids are a pre-requisite for G108-mediated energy expenditure and weight loss. However, preventing hypoaminoacidemia with additional dietary protein does not affect the ability of G108 to improve glycemia or hepatic steatosis in obese mice. These studies provide a mechanism for glucagon-mediated weight loss and confirm the hepatic glucagon receptor as an attractive molecular target for metabolic disease therapeutics.


Subject(s)
Glucagon , Weight Loss , Mice , Animals , Glucagon/metabolism , Energy Metabolism/physiology , Receptors, Glucagon/metabolism , Mice, Obese , Amino Acids/pharmacology
2.
Ann Hepatol ; 25: 100549, 2021.
Article in English | MEDLINE | ID: mdl-34614431

ABSTRACT

Malnutrition among patients with chronic liver disease (CLD) is a common complication with significant prognostic implications for patients with liver cirrhosis. Micronutrient deficiency has been associated with an increased risk of hepatic decompensation and is an independent risk factor for mortality among cirrhotic patients. Micronutrient deficiencies in patients with CLD include zinc, vitamin A, vitamin D and selenium. This review article aims to evaluate the literature to date on the complications of zinc deficiency in patients with CLD. A management algorithm for zinc replacement has also been proposed.


Subject(s)
Dietary Supplements , Liver Diseases/therapy , Trace Elements/therapeutic use , Zinc/therapeutic use , Chronic Disease , Humans , Liver Diseases/diagnosis , Liver Diseases/etiology
3.
J Pain Res ; 13: 2269-2278, 2020.
Article in English | MEDLINE | ID: mdl-32982390

ABSTRACT

INTRODUCTION: Cannabidiol (CBD) is reported to produce pain relief, but the clinically relevant cellular and molecular mechanisms remain uncertain. The TRPV1 receptor integrates noxious stimuli and plays a key role in pain signaling. Hence, we conducted in vitro studies, to elucidate the efficacy and mechanisms of CBD for inhibiting neuronal hypersensitivity in cultured rat sensory neurons, following activation of TRPV1. METHODS: Adult rat dorsal root ganglion (DRG) neurons were cultured and supplemented with the neurotrophic factors NGF and GDNF, in an established model of neuronal hypersensitivity. Neurons were stimulated with CBD (Adven 150, EMMAC Life Sciences) at 1, 10, 100 nMol/L and 1, 10 and 50 µMol/L, 48 h after plating. In separate experiments, DRG neurons were also stimulated with capsaicin with or without CBD (1 nMol/L to10 µMol/L), in a functional calcium imaging assay. The effects of the adenylyl cyclase activator forskolin and the calcineurin inhibitor cyclosporin were determined. We also measured forskolin-stimulated cAMP levels, without and after treatment with CBD, using a homogenous time-resolved fluorescence (HTRF) assay. The results were analysed using Mann-Whitney test. RESULTS: DRG neurons treated with 10 and 50 µMol/L CBD showed calcium influx, but not at lower doses. Neurons treated with capsaicin demonstrated robust calcium influx, which was dose-dependently reduced in the presence of low dose CBD (IC50 = 100 nMol/L). The inhibition or desensitization by CBD was reversed in the presence of forskolin and cyclosporin. Forskolin-stimulated cAMP levels were significantly reduced in CBD treated neurons. CONCLUSION: CBD at low doses corresponding to plasma concentrations observed physiologically inhibits or desensitizes neuronal TRPV1 signalling by inhibiting the adenylyl cyclase - cAMP pathway, which is essential for maintaining TRPV1 phosphorylation and sensitization. CBD also facilitated calcineurin-mediated TRPV1 inhibition. These mechanisms may underlie nociceptor desensitization and the therapeutic effect of CBD in animal models and patients with acute and chronic pain.

4.
Mol Metab ; 6(1): 48-60, 2017 01.
Article in English | MEDLINE | ID: mdl-28123937

ABSTRACT

OBJECTIVE: Dietary supplementation with fermentable carbohydrate protects against body weight gain. Fermentation by the resident gut microbiota produces short-chain fatty acids, which act at free fatty acid receptor 2 (FFAR2). Our aim was to test the hypothesis that FFAR2 is important in regulating the beneficial effects of fermentable carbohydrate on body weight and to understand the role of gut hormones PYY and GLP-1. METHODS: Wild-type or Ffar2-/- mice were fed an inulin supplemented or control diet. Mice were metabolically characterized and gut hormone concentrations, enteroendocrine cell density measurements were carried out. Intestinal organoids and colonic cultures were utilized to substantiate the in vivo findings. RESULTS: We provide new mechanistic insight into how fermentable carbohydrate regulates metabolism. Using mice that lack FFAR2, we demonstrate that the fermentable carbohydrate inulin acts via this receptor to drive an 87% increase in the density of cells that produce the appetite-suppressing hormone peptide YY (PYY), reduce food intake, and prevent diet-induced obesity. CONCLUSION: Our results demonstrate that FFAR2 is predominantly involved in regulating the effects of fermentable carbohydrate on metabolism and does so, in part, by enhancing PYY cell density and release. This highlights the potential for targeting enteroendocrine cell differentiation to treat obesity.


Subject(s)
Dietary Carbohydrates/metabolism , Peptide YY/metabolism , Receptors, Cell Surface/metabolism , Animals , Body Weight , Colon/cytology , Dietary Supplements , Eating , Fatty Acids, Volatile/metabolism , Fermentation , Fermented Foods , Gastrointestinal Hormones/metabolism , Gastrointestinal Microbiome/physiology , Glucagon-Like Peptide 1/metabolism , Inulin/metabolism , Male , Mice , Mice, Knockout , Obesity/metabolism , Receptors, Cell Surface/physiology , Weight Gain
5.
Brain Struct Funct ; 221(4): 2035-47, 2016 05.
Article in English | MEDLINE | ID: mdl-25758403

ABSTRACT

Kisspeptin (encoded by KISS1) is a crucial activator of reproductive function. The role of kisspeptin has been studied extensively within the hypothalamus but little is known about its significance in other areas of the brain. KISS1 and its cognate receptor are expressed in the amygdala, a key limbic brain structure with inhibitory projections to hypothalamic centers involved in gonadotropin secretion. We therefore hypothesized that kisspeptin has effects on neuronal activation and reproductive pathways beyond the hypothalamus and particularly within the amygdala. To test this, we mapped brain neuronal activity (using manganese-enhanced MRI) associated with peripheral kisspeptin administration in rodents. We also investigated functional relevance by measuring the gonadotropin response to direct intra-medial amygdala (MeA) administration of kisspeptin and kisspeptin antagonist. Peripheral kisspeptin administration resulted in a marked decrease in signal intensity in the amygdala compared to vehicle alone. This was associated with an increase in luteinizing hormone (LH) secretion. In addition, intra-MeA administration of kisspeptin resulted in increased LH secretion, while blocking endogenous kisspeptin signaling within the amygdala by administering intra-MeA kisspeptin antagonist decreased both LH secretion and LH pulse frequency. We provide evidence for the first time that neuronal activity within the amygdala is decreased by peripheral kisspeptin administration and that kisspeptin signaling within the amygdala contributes to the modulation of gonadotropin release and pulsatility. Our data suggest that kisspeptin is a 'master regulator' of reproductive physiology, integrating limbic circuits with the regulation of gonadotropin-releasing hormone neurons and reproductive hormone secretion.


Subject(s)
Amygdala/metabolism , Kisspeptins/physiology , Luteinizing Hormone/metabolism , Reproduction , Amygdala/drug effects , Amygdala/physiology , Animals , Contrast Media , Female , Hypothalamus/drug effects , Hypothalamus/metabolism , Hypothalamus/physiology , Kisspeptins/administration & dosage , Kisspeptins/metabolism , Magnetic Resonance Imaging , Manganese , Mice, Inbred C57BL , Rats , Rats, Sprague-Dawley
6.
Neuroendocrinology ; 100(2-3): 141-52, 2014.
Article in English | MEDLINE | ID: mdl-25247878

ABSTRACT

Neuronal populations that synthesize kisspeptin (KP), neurokinin B (NKB) and substance P (SP) in the hypothalamic infundibular nucleus of humans are partly overlapping. These cells are important upstream regulators of gonadotropin-releasing hormone (GnRH) neurosecretion. Homologous neurons in laboratory animals are thought to modulate episodic GnRH secretion primarily via influencing KP receptors on the hypophysiotropic fiber projections of GnRH neurons. To explore the structural basis of this putative axo-axonal communication in humans, we analyzed the anatomical relationship of KP-immunoreactive (IR), NKB-IR and SP-IR axon plexuses with hypophysiotropic GnRH fiber projections. Immunohistochemical studies were carried out on histological samples from postmenopausal women. The neuropeptide-IR axons innervated densely the portal capillary network in the postinfundibular eminence. Subsets of the fibers formed descending tracts in the infundibular stalk, some reaching the neurohypophysis. KP-IR, NKB-IR and SP-IR plexuses intermingled, and established occasional contacts, with hypophysiotropic GnRH fibers in the postinfundibular eminence and through their lengthy course while descending within the infundibular stalk. Triple-immunofluorescent studies also revealed considerable overlap between the KP, NKB and SP signals in individual fibers, providing evidence that these peptidergic projections arise from neurons of the mediobasal hypothalamus. These neuroanatomical observations indicate that the hypophysiotropic projections of human GnRH neurons in the postinfundibular eminence and the descending GnRH tract coursing through the infundibular stalk to the neurohypophysis are exposed to neurotransmitters/neuropeptides released by dense KP-IR, NKB-IR and SP-IR fiber plexuses. Localization and characterization of axonal neuropeptide receptors will be required to clarify the putative autocrine and paracrine interactions in these anatomical regions.


Subject(s)
Gonadotropin-Releasing Hormone/metabolism , Hypothalamus/metabolism , Kisspeptins/metabolism , Neurokinin B/metabolism , Pituitary Gland/metabolism , Substance P/metabolism , Aged , Aged, 80 and over , Axons/metabolism , Female , Humans , Hypothalamus/cytology , Immunohistochemistry , Middle Aged , Neurons/cytology , Neurons/metabolism , Pituitary Gland/cytology , Postmenopause/metabolism
7.
PLoS One ; 9(5): e97611, 2014.
Article in English | MEDLINE | ID: mdl-24845101

ABSTRACT

Kisspeptin is a hypothalamic peptide hormone that plays a pivotal role in pubertal onset and reproductive function. Previous studies have examined hypothalamic kisspeptin mRNA expression, either through in situ hybridisation or real-time RT-PCR, as a means quantifying kisspeptin gene expression. However, mRNA expression levels are not always reflected in levels of the translated protein. Kisspeptin-immunoreactivity (IR) has been extensively examined using immunohistochemistry, enabling detection and localisation of kisspeptin perikaya in the arcuate nucleus (ARC) and anteroventral periventricular nucleus (AVPV). However, quantification of kisspeptin-IR remains challenging. We developed a specific rodent radioimmunoassay assay (RIA) capable of detecting and quantifying kisspeptin-IR in rodent tissues. The RIA uses kisspeptin-10 as a standard and radioactive tracer, combined with a commercially available antibody raised to the kisspeptin-10 fragment. Adult female wistar rat brain samples were sectioned at 300 µm and the ARC and AVPV punch micro-dissected. Brain punches were homogenised in extraction buffer and assayed with rodent kisspeptin-RIA. In accord with the pattern of kisspeptin mRNA expression, kisspeptin-IR was detected in both the ARC (47.1±6.2 fmol/punch, mean±SEM n = 15) and AVPV (7.6±1.3 fmol/punch, mean±SEM n = 15). Kisspeptin-IR was also detectable in rat placenta (1.26±0.15 fmol/mg). Reverse phase high pressure liquid chromatography analysis showed that hypothalamic kisspeptin-IR had the same elution profile as a synthetic rodent kisspeptin standard. A specific rodent kisspeptin-RIA will allow accurate quantification of kisspeptin peptide levels within specific tissues in rodent experimental models.


Subject(s)
Hypothalamus/metabolism , Kisspeptins/metabolism , Nerve Tissue Proteins/metabolism , Animals , Female , Gene Expression Regulation/physiology , Kisspeptins/immunology , Radioimmunoassay/methods , Rats , Rats, Wistar , Sensitivity and Specificity
8.
Endocrinology ; 153(11): 5428-39, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23011920

ABSTRACT

Peptidergic neurons synthesizing kisspeptin (KP) and neurokinin B (NKB) in the hypothalamic infundibular nucleus have been implicated in negative sex steroid feedback to GnRH neurons. In laboratory rodents, testosterone decreases KP and NKB expression in this region. In the present study, we addressed the hypothesis that the weakening of this inhibitory testosterone feedback in elderly men coincides with enhanced KP and NKB signaling in the infundibular nucleus. This central hypothesis was tested in a series of immunohistochemical studies on hypothalamic sections of male human individuals that were divided into arbitrary "young" (21-49 yr, n = 11) and "aged" (50-67 yr, n = 9) groups. Quantitative immunohistochemical experiments established that the regional densities of NKB-immunoreactive (IR) perikarya and fibers, and the incidence of afferent contacts they formed onto GnRH neurons, exceeded several times those of the KP-IR elements. Robust aging-dependent enhancements were identified in the regional densities of KP-IR perikarya and fibers and the incidence of afferent contacts they established onto GnRH neurons. The abundance of NKB-IR perikarya, fibers, and axonal appositions to GnRH neurons also increased with age, albeit to lower extents. In dual-immunofluorescent studies, the incidence of KP-IR NKB perikarya increased from 36% in young to 68% in aged men. Collectively, these immunohistochemical data suggest an aging-related robust enhancement in central KP signaling and a moderate enhancement in central NKB signaling. These changes are compatible with a reduced testosterone negative feedback to KP and NKB neurons. The heavier KP and NKB inputs to GnRH neurons in aged, compared with young, men may play a role in the enhanced central stimulation of the reproductive axis. It requires clarification to what extent the enhanced KP and NKB signaling upstream from GnRH neurons is an adaptive response to hypogonadism or, alternatively, a consequence of a decline in the androgen sensitivity of KP and NKB neurons.


Subject(s)
Aging/metabolism , Arcuate Nucleus of Hypothalamus/metabolism , Kisspeptins/metabolism , Neurokinin B/metabolism , Neurons/metabolism , Signal Transduction/physiology , Adult , Aged , Axons/metabolism , Gonadotropin-Releasing Hormone/metabolism , Humans , Hypothalamus/metabolism , Male , Middle Aged , Testosterone/metabolism
9.
Exp Diabetes Res ; 2012: 824305, 2012.
Article in English | MEDLINE | ID: mdl-22899902

ABSTRACT

Obesity is one of the major challenges to human health worldwide; however, there are currently no effective pharmacological interventions for obesity. Recent studies have improved our understanding of energy homeostasis by identifying sophisticated neurohumoral networks which convey signals between the brain and gut in order to control food intake. The hypothalamus is a key region which possesses reciprocal connections between the higher cortical centres such as reward-related limbic pathways, and the brainstem. Furthermore, the hypothalamus integrates a number of peripheral signals which modulate food intake and energy expenditure. Gut hormones, such as peptide YY, pancreatic polypeptide, glucagon-like peptide-1, oxyntomodulin, and ghrelin, are modulated by acute food ingestion. In contrast, adiposity signals such as leptin and insulin are implicated in both short- and long-term energy homeostasis. In this paper, we focus on the role of gut hormones and their related neuronal networks (the gut-brain axis) in appetite control, and their potentials as novel therapies for obesity.


Subject(s)
Appetite Regulation/physiology , Appetite , Gastrointestinal Hormones/metabolism , Obesity/complications , Obesity/metabolism , Bariatric Surgery/methods , Hormones/metabolism , Humans , Hypothalamus/pathology , Intestines/microbiology , Models, Biological , Neurons/metabolism , Obesity/therapy , Reward , Risk Factors , Signal Transduction
10.
Neuropharmacology ; 63(1): 18-30, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22369786

ABSTRACT

Neuropeptides released by hypothalamic neurons play a major role in the regulation of feeding, acting both within the hypothalamus, and at other appetite regulating centres throughout the brain. Where classical neurotransmitters signal only within synapses, neuropeptides diffuse over greater distances affecting both nearby and distant neurons expressing the relevant receptors, which are often extrasynaptic. As well as triggering a behavioural output, neuropeptides also act as neuromodulators: altering the response of neurons to both neurotransmitters and circulating signals of nutrient status. The mechanisms of action of hypothalamic neuropeptides with established roles in feeding, including melanin-concentrating hormone (MCH), the orexins, α-melanocyte stimulating hormone (α-MSH), agouti-gene related protein (AgRP), neuropeptide Y, and oxytocin, are reviewed in this article, with emphasis laid on both their effects on appetite regulating centres throughout the brain, and on examining the evidence for their physiological roles. In addition, evidence for the involvement of several putative appetite regulating hypothalamic neuropeptides is assessed including, ghrelin, cocaine and amphetamine-regulated transcript (CART), neuropeptide W and the galanin-like peptides. This article is part of a Special Issue entitled 'Central control of Food Intake'.


Subject(s)
Appetite Regulation/physiology , Hypothalamus/metabolism , Neuropeptides/metabolism , Animals , Eating/drug effects , Eating/physiology , Humans , Hypothalamus/anatomy & histology , Neuropeptides/pharmacology
11.
Neuroimage ; 59(2): 968-78, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-21925279

ABSTRACT

Manganese enhanced MRI (MEMRI) is an imaging paradigm that can be used to assess neuronal activity in vivo. Here we investigate, through the use of MEMRI, the influence of receptor dynamics on neuronal activity in the hypothalamus and hippocampus focusing on the glutamate receptor signalling system. We demonstrate that intraperitoneal (i.p.) administration of monosodium glutamate (MSG) and the ionotropic glutamate receptor (iGluR) agonists NMDA and AMPA resulted in significantly increased signal intensity (SI) in the arcuate nucleus (ARC), the suprachiasmatic nucleus (SCN) and the CA3 region of the hippocampus of mice consistent with increased neuronal activity. Administration of the NMDA receptor antagonist MK-801 resulted in significantly decreased SI in the paraventricular nucleus (PVN) consistent with decreased neuronal activity. Co-administration of MSG and the AMPA receptor antagonist NBQX attenuated the increase in SI observed in the ARC from MSG alone, suggesting MEMRI may be applicable to the study of receptor dynamics in vivo. We also observed that administration of the various iGluR agonists and antagonists modulated SI in the lateral ventricle and that high dose MSG (300 mg) caused a hitherto unseen enhancement in SI in the entire cortical/subarachnoid region. In conclusion, MEMRI reveals changes in neuronal activity in response to iGluR agonists and antagonists in the CNS in vivo as well as revealing multifaceted effects beyond those attributable to neuronal activity alone.


Subject(s)
Excitatory Amino Acid Antagonists/pharmacology , Hippocampus/physiology , Hypothalamus/physiology , Magnetic Resonance Imaging/methods , Manganese , Neurons/physiology , Receptors, Glutamate/metabolism , Action Potentials/drug effects , Action Potentials/physiology , Animals , Brain Mapping/methods , Contrast Media , Hippocampus/drug effects , Hypothalamus/drug effects , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Receptors, Glutamate/drug effects , Reproducibility of Results , Sensitivity and Specificity
12.
Curr Opin Investig Drugs ; 11(10): 1151-7, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20872318

ABSTRACT

Oxyntomodulin, a product of the proglucagon gene, is released from the enteroendocrine L-cells of the gastrointestinal tract after the digestion of food, and acts via glucagon-like peptide 1 receptors in the arcuate nucleus to induce satiety. The administration of oxyntomodulin to animals and humans causes weight loss by reducing food intake in combination with increasing energy expenditure. Thus, the development of potent and long-acting analogs of oxyntomodulin is an exciting new therapeutic avenue for addressing the global obesity epidemic. This review discusses the role of oxyntomodulin in the physiological control of appetite, and presents the currently available evidence suggesting its potential as an obesity treatment.


Subject(s)
Appetite Depressants/pharmacology , Appetite Depressants/therapeutic use , Obesity/drug therapy , Oxyntomodulin/pharmacology , Oxyntomodulin/therapeutic use , Animals , Appetite Regulation/drug effects , Clinical Trials as Topic , Drug Evaluation, Preclinical , Eating/drug effects , Eating/physiology , Energy Metabolism/drug effects , Enteroendocrine Cells/metabolism , Enteroendocrine Cells/physiology , Gastrointestinal Tract/metabolism , Glucagon/metabolism , Glucagon-Like Peptides/pharmacology , Glucagon-Like Peptides/therapeutic use , Humans
13.
Endocr J ; 57(5): 359-72, 2010.
Article in English | MEDLINE | ID: mdl-20424341

ABSTRACT

The World Health Organisation has estimated that by 2015 approximately 2.3 billion adults will be overweight and more than 700 million obese. Obesity is associated with an increased risk of diabetes, cardiovascular events, stroke and cancer. The hypothalamus is a crucial region for integrating signals from central and peripheral pathways and plays a major role in appetite regulation. In addition, there are reciprocal connections with the brainstem and higher cortical centres. In the arcuate nucleus of the hypothalamus, there are two major neuronal populations which stimulate or inhibit food intake and influence energy homeostasis. Within the brainstem, the dorsal vagal complex plays a role in the interpretation and relaying of peripheral signals. Gut hormones act peripherally to modulate digestion and absorption of nutrients. However, they also act as neurotransmitters within the central nervous system to control food intake. Peptide YY, pancreatic polypeptide, glucagon-like peptide-1 and oxyntomodulin suppress appetite, whilst ghrelin increases appetite through afferent vagal fibres to the caudal brainstem or directly to the hypothalamus. A better understanding of the role of these gut hormones may offer the opportunity to develop successful treatments for obesity. Here we review the current understanding of the role of gut hormones and the hypothalamus on food intake and body weight control.


Subject(s)
Appetite Regulation/physiology , Gastrointestinal Hormones/physiology , Hypothalamus/physiology , Adult , Animals , Appetite Regulation/genetics , Brain Stem/metabolism , Brain Stem/physiology , Feeding Behavior/physiology , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/metabolism , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/physiology , Humans , Models, Biological , Obesity/genetics , Obesity/metabolism , Obesity/physiopathology , Reward
14.
Diabetes ; 59(2): 397-406, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19933997

ABSTRACT

OBJECTIVE: Prokineticin 2 (PK2) is a hypothalamic neuropeptide expressed in central nervous system areas known to be involved in food intake. We therefore hypothesized that PK2 plays a role in energy homeostasis. RESEARCH DESIGN AND METHODS: We investigated the effect of nutritional status on hypothalamic PK2 expression and effects of PK2 on the regulation of food intake by intracerebroventricular (ICV) injection of PK2 and anti-PK2 antibody. Subsequently, we investigated the potential mechanism of action by determining sites of neuronal activation after ICV injection of PK2, the hypothalamic site of action of PK2, and interaction between PK2 and other hypothalamic neuropeptides regulating energy homeostasis. To investigate PK2's potential as a therapeutic target, we investigated the effect of chronic administration in lean and obese mice. RESULTS: Hypothalamic PK2 expression was reduced by fasting. ICV administration of PK2 to rats potently inhibited food intake, whereas anti-PK2 antibody increased food intake, suggesting that PK2 is an anorectic neuropeptide. ICV administration of PK2 increased c-fos expression in proopiomelanocortin neurons of the arcuate nucleus (ARC) of the hypothalamus. In keeping with this, PK2 administration into the ARC reduced food intake and PK2 increased the release of alpha-melanocyte-stimulating hormone (alpha-MSH) from ex vivo hypothalamic explants. In addition, ICV coadministration of the alpha-MSH antagonist agouti-related peptide blocked the anorexigenic effects of PK2. Chronic peripheral administration of PK2 reduced food and body weight in lean and obese mice. CONCLUSIONS: This is the first report showing that PK2 has a role in appetite regulation and its anorectic effect is mediated partly via the melanocortin system.


Subject(s)
Energy Intake/drug effects , Gastrointestinal Hormones/pharmacology , Gastrointestinal Hormones/physiology , Neuropeptides/pharmacology , Neuropeptides/physiology , Obesity/physiopathology , Animals , Dose-Response Relationship, Drug , Gastrointestinal Hormones/genetics , Gene Expression Regulation , Hypothalamus/physiology , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Motor Activity/drug effects , Neuropeptides/genetics , RNA, Messenger/genetics , Rats , Rats, Wistar
15.
J Clin Endocrinol Metab ; 94(11): 4315-23, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19820030

ABSTRACT

BACKGROUND: Kisspeptin is a critical regulator of normal reproductive function. A single injection of kisspeptin in healthy human volunteers potently stimulates gonadotropin release. However, the effects of kisspeptin on gonadotropin release in women with hypothalamic amenorrhea (HA) and the effects of repeated administration of kisspeptin to humans are unknown. AIM: The aim of this study was to determine the effects of acute and chronic kisspeptin administration on gonadotropin release in women with HA. METHODS: We performed a prospective, randomized, double-blinded, parallel design study. Women with HA received twice-daily sc injections of kisspeptin (6.4 nmol/kg) or 0.9% saline (n = 5 per group) for 2 wk. Changes in serum gonadotropin and estradiol levels, LH pulsatility, and ultrasound measurements of reproductive activity were assessed. RESULTS: On the first injection day, potent increases in serum LH and FSH were observed after sc kisspeptin injection in women with HA (mean maximal increment from baseline within 4 h after injection: LH, 24.0 +/- 3.5 IU/liter; FSH, 9.1 +/- 2.5 IU/liter). These responses were significantly reduced on the 14th injection day (mean maximal increment from baseline within 4 h postinjection: LH, 2.5 +/- 2.2 IU/liter, P < 0.05; FSH, 0.5 +/- 0.5 IU/liter, P < 0.05). Subjects remained responsive to GnRH after kisspeptin treatment. No significant changes in LH pulsatility or ultrasound measurements of reproductive activity were observed. CONCLUSION: Acute administration of kisspeptin to women with infertility due to HA potently stimulates gonadotropin release, but chronic administration of kisspeptin results in desensitization to its effects on gonadotropin release. These data have important implications for the development of kisspeptin as a novel therapy for reproductive disorders in humans.


Subject(s)
Amenorrhea/drug therapy , Gonadotropins/metabolism , Tachyphylaxis/physiology , Tumor Suppressor Proteins/therapeutic use , Adult , Body Mass Index , Body Weight , Female , Follicle Stimulating Hormone/blood , Gonadotropins/blood , Humans , Hypothalamus/physiopathology , Kisspeptins , Luteinizing Hormone/blood , Spectrometry, Mass, Electrospray Ionization/methods , Tumor Suppressor Proteins/adverse effects , Tumor Suppressor Proteins/chemistry , Weight Gain , Young Adult
16.
Magn Reson Med ; 62(2): 279-83, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19526502

ABSTRACT

Carbon-13 ((13)C) high-resolution magic angle spinning (HR-MAS) spectroscopy was used to investigate the neuroglial coupling mechanisms underlying appetite regulation in the brain of C57BL/6J mice metabolizing [1-(13)C]glucose. Control fed or overnight fasted mice received [1-(13)C]glucose (20 micromol/g intraperitoneally [i.p.]), 15 min prior to brain fixation by focused microwaves. The hypothalamic region was dissected from the rest of the brain and (13)C HR-MAS spectra were obtained from both biopsies. Fasting resulted in a significant increase in hypothalamic [3-(13)C]lactate and [2-(13)C]gamma-aminobutyric acid (GABA) relative to the remaining brain. Administration of the orexigenic peptide ghrelin (0.3 nmol/g i.p.) did not increase hypothalamic [3-(13)C]lactate or [2-(13)C]GABA, suggesting that ghrelin signaling is not sufficient to elicit all the metabolic consequences of hypothalamic activation by fasting. Our results indicate that the hypothalamic regulation of appetite involves, in addition to the well-known neuropeptide signaling, increased neuroglial lactate shuttling and augmented GABA concentrations.


Subject(s)
Fasting/physiology , Hypothalamus/metabolism , Lactic Acid/analysis , Magnetic Resonance Spectroscopy/methods , Animals , Carbon Isotopes/analysis , Male , Mice , Mice, Inbred C57BL
17.
Arq Bras Endocrinol Metabol ; 53(2): 120-8, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19466203

ABSTRACT

Current estimates suggest that over 1 billion people are overweight and over 300 million people are obese. Weight gain is due to an imbalance between energy expenditure and dietary intake. This review discusses the hypothalamic control of appetite and highlights key developments in research that have furthered our understanding of the complex pathways involved. Nuclei within the hypothalamus integrate peripheral signals such as adiposity and caloric intake to regulate important pathways within the central nervous system controlling food intake and energy expenditure. Firmly established pathways involve the orexigenic NPY/AgRP and the anorexigenic POMC/CART neurons in the arcuate nucleus (ARC) of the hypothalamus. These project from the ARC to other important hypothalamic nuclei, including the paraventricular, dorsomedial, ventromedial and lateral hypothalamic nuclei. In addition there are many projections to and from the brainstem, cortical areas and reward pathways, which modulate food intake.


Subject(s)
Appetite Regulation/physiology , Feeding Behavior/physiology , Hypothalamus/physiology , Obesity/physiopathology , Arcuate Nucleus of Hypothalamus/physiology , Gastrointestinal Hormones/physiology , Humans , Obesity/metabolism , Obesity/therapy
18.
Endocrinology ; 150(8): 3513-20, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19359390

ABSTRACT

Ghrelin is a gastric peptide that regulates appetite and GH secretion. Circulating ghrelin levels are elevated by fasting and suppressed postprandially. However, the mechanisms regulating circulating ghrelin levels are unclear. Oxyntomodulin is an anorexic peptide hormone released from L cells in the gut. We investigated the effects of intracerebroventricular (icv) administration of oxyntomodulin on circulating ghrelin levels. The icv administration of 1, 3, or 10 nmol oxyntomodulin reduced circulating acylated and total (acylated and des-acylated) ghrelin 60 min after icv injection. Administration of 1 nmol oxyntomodulin directly into the arcuate nucleus of the hypothalamus significantly reduced total and acylated ghrelin levels, and administration of 3 nmol oxyntomodulin into the lateral ventricle induced c-fos mRNA expression in arcuate nucleus neurons expressing the glucagon-like peptide-1 (GLP-1) receptor. In a final study, the reduction in total ghrelin observed after icv injection of 3 nmol oxyntomodulin was blocked by coadministration of the GLP-1 receptor antagonist exendin (9-39). These studies suggest oxyntomodulin reduces peripheral ghrelin levels via GLP-1 receptor-dependent hypothalamic pathways. Postprandial release of anorexic gut hormones may thus act centrally to contribute to the postprandial reduction in circulating ghrelin.


Subject(s)
Ghrelin/blood , Hypothalamus/drug effects , Hypothalamus/metabolism , Oxyntomodulin/pharmacology , Animals , Blood Glucose/drug effects , Eating/drug effects , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Glucagon-Like Peptide-1 Receptor , Injections , Insulin/blood , Male , Mice , Oxyntomodulin/administration & dosage , Peptide Fragments/pharmacology , Proto-Oncogene Proteins c-fos/genetics , Radioimmunoassay , Rats , Rats, Wistar , Receptors, Glucagon/antagonists & inhibitors , Receptors, Glucagon/metabolism
19.
Arq. bras. endocrinol. metab ; 53(2): 120-128, Mar. 2009. ilus
Article in English | LILACS | ID: lil-513765

ABSTRACT

Current estimates suggest that over 1 billion people are overweight and over 300 million people are obese. Weight gain is due to an imbalance between energy expenditure and dietary intake. This review discusses the hypothalamic control of appetite and highlights key developments in research that have furthered our understanding of the complex pathways involved. Nuclei within the hypothalamus integrate peripheral signals such as adiposity and caloric intake to regulate important pathways within the central nervous system controlling food intake and energy expenditure. Firmly established pathways involve the orexigenic NPY/AgRP and the anorexigenic POMC/CART neurons in the arcuate nucleus (ARC) of the hypothalamus. These project from the ARC to other important hypothalamic nuclei, including the paraventricular, dorsomedial, ventromedial and lateral hypothalamic nuclei. In addition there are many projections to and from the brainstem, cortical areas and reward pathways, which modulate food intake.


As estimativas atuais sugerem que mais de 1 bilhão de pessoas apresentam sobrepeso e 300 milhões são obesas. O ganho de peso representa um desequilíbrio entre o gasto energético e o consumo alimentar. Esta revisão discute o controle hipotalâmico do apetite e destaca os pontos-chave no desenvolvimento de pesquisas para ampliar o nosso entendimento dos complexos mecanismos envolvidos nesta regulação. Núcleos situados no hipotálamo integram uma série de sinais com o sistema nervoso central controlando a ingestão alimentar e o gasto energético. As vias mais estabelecidas envolvem os neurônios orexigênicos NPY/AgRP e os neurônios anorexigênicos POMC/CART no núcleo arqueado (ARC) do hipotálamo. Esses neurônios se projetam do ARC para outros importantes núcleos hipotalâmicos, tais quais: paraventricular, dorsomedial, ventromedial e lateral. Além disso, existem várias projeções que vão e vem do tronco cerebral, das áreas corticais e das vias de retroalimentação que modulam o consumo alimentar.


Subject(s)
Humans , Appetite Regulation/physiology , Feeding Behavior/physiology , Hypothalamus/physiology , Obesity/physiopathology , Arcuate Nucleus of Hypothalamus/physiology , Gastrointestinal Hormones/physiology , Obesity/metabolism , Obesity/therapy
20.
Obesity (Silver Spring) ; 17(6): 1135-43, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19214175

ABSTRACT

Xenin is a 25-amino acid peptide highly homologous to neurotensin. Xenin and neurotensin are reported to have similar biological effects. Both reduce food intake when administered centrally to fasted rats. We aimed to clarify and compare the effects of these peptides on food intake and behavior. We confirm that intracerebroventricular (ICV) administration of xenin or neurotensin reduces food intake in fasted rats, and demonstrate that both reduce food intake in satiated rats during the dark phase. Xenin reduced food intake more potently than neurotensin following ICV administration. ICV injection of either peptide in the dark phase increased resting behavior. Xenin and neurotensin stimulated the release of corticotrophin-releasing hormone (CRH) from ex vivo hypothalamic explants, and administration of alpha-helical CRH attenuated their effects on food intake. Intraperitoneal (IP) administration of xenin or neurotensin acutely reduced food intake in fasted mice and ad libitum fed mice in the dark phase. However, chronic continuous or twice daily peripheral administration of xenin or neurotensin to mice had no significant effect on daily food intake or body weight. These studies confirm that ICV xenin or neurotensin can acutely reduce food intake and demonstrate that peripheral administration of xenin and neurotensin also reduces food intake. This may be partly mediated by changes in hypothalamic CRH release. The lack of chronic effects on body weight observed in our experiments suggests that xenin and neurotensin are unlikely to be useful as obesity therapies.


Subject(s)
Eating , Feeding Behavior , Hypothalamus/metabolism , Neurotensin/metabolism , Peptides/metabolism , Animals , Body Weight , Corticotropin-Releasing Hormone/metabolism , Fasting/metabolism , Infusion Pumps, Implantable , Infusions, Subcutaneous , Injections, Intraperitoneal , Injections, Intraventricular , Male , Mice , Mice, Inbred C57BL , Neurotensin/administration & dosage , Organ Culture Techniques , Peptides/administration & dosage , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL