51
|
Kawamura K, Mori W, Fujinaga M, Yamasaki T, Zhang Y, Wakizaka H, Hatori A, Xie L, Kumata K, Ohkubo T, Kurihara Y, Ogawa M, Nengaki N, Zhang MR. Radiosynthesis and in vivo evaluation of 11C-labeled BMS-193885 and its desmethyl analog as PET tracers for neuropeptide Y1 receptors. EJNMMI Radiopharm Chem 2019; 4:4. [PMID: 31659508 PMCID: PMC6379498 DOI: 10.1186/s41181-019-0056-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 02/04/2019] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Neuropeptide Y (NPY) has been implicated in a wide variety of physiological processes, including feeding, learning, memory, emotion, cardiovascular homeostasis, hormone secretion, and circadian rhythms. NPY Yl receptor (NPY Y1-R) is the most widely studied NPY receptor, and is involved in many of these processes. BMS-193885 (1) was previously developed as a potent and selective NPY Y1-R antagonist, which has good systemic bioavailability and brain penetration. To evaluate the characteristics of 1 in vivo, we developed 11C-labeled BMS-193885 ([11C]1) and its desmethyl analog ([11C]2) for potential use as two new positron emission tomography (PET) tracers. RESULTS [11C]1 was synthesized from [11C]methyl iodide using 2. [11C]2 was synthesized from [11C]phosgene using its aniline and amine derivatives. The mean ± S.D. decay-corrected radiochemical yields of [11C]1 and [11C]2 from 11CO2 at the end of radionuclide production were 23 ± 3.2% (n = 6) and 24 ± 1.5% (n = 4), respectively. In biodistribution on mice, radioactivity levels for both tracers were relatively high in the kidney, small intestine, and liver at 60 min post-injection. The radioactivity levels in the kidney, lung, and spleen of mice at 30 min post-injection with [11C]1 were significantly reduced by pretreatment with 1 (10 mg/kg), and levels of [11C]1 in the brain of mice were significantly increased by pretreatment with the P-glycoprotein and breast cancer resistance protein inhibitor elacridar (10 mg/kg). In metabolite analysis using mouse plasma, [11C]1 and [11C]2 were rapidly metabolized within 30 min post-injection, and [11C]1 was mainly metabolized into unlabeled 2 and radiolabeled components. CONCLUSION [11C]1 and [11C]2 were successfully synthesized with sufficient amount of radioactivity and high quality for use in vivo. Our study of [11C]1 and its desmethyl analog [11C]2 was useful in that it helped to elucidate the in vivo characteristics of 1.
Collapse
Affiliation(s)
- Kazunori Kawamura
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.
| | - Wakana Mori
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Masayuki Fujinaga
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Tomoteru Yamasaki
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Yiding Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Hidekatsu Wakizaka
- Department of Medical Physics, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, Chiba, 263-8555, Japan
| | - Akiko Hatori
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Lin Xie
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Katsushi Kumata
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| | - Takayuki Ohkubo
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.,SHI Accelerator Service Ltd, Tokyo, 141-0032, Japan
| | - Yusuke Kurihara
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.,SHI Accelerator Service Ltd, Tokyo, 141-0032, Japan
| | - Masanao Ogawa
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.,SHI Accelerator Service Ltd, Tokyo, 141-0032, Japan
| | - Nobuki Nengaki
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan.,SHI Accelerator Service Ltd, Tokyo, 141-0032, Japan
| | - Ming-Rong Zhang
- Department of Radiopharmaceuticals Development, National Institute of Radiological Sciences, National Institutes for Quantum and Radiological Science and Technology, 4-9-1 Anagawa, Inage-ku, Chiba, 263-8555, Japan
| |
Collapse
|
52
|
Huston NJ, Brenner LA, Taylor ZC, Ritter RC. NPY2 receptor activation in the dorsal vagal complex increases food intake and attenuates CCK-induced satiation in male rats. Am J Physiol Regul Integr Comp Physiol 2019; 316:R406-R416. [PMID: 30726118 DOI: 10.1152/ajpregu.00011.2019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Neuropeptide Y (NPY), peptide YY (PYY), and their cognate receptors (YR) are expressed by subpopulations of central and peripheral nervous system neurons. Intracerebroventricular injections of NPY or PYY increase food intake, and intrahypothalamic NPY1 or NPY5 receptor agonist injections also increase food intake. In contrast, injection of PYY in the periphery reduces food intake, apparently by activating peripheral Y2R. The dorsal vagal complex (DVC) of the hindbrain is the site where vagal afferents relay gut satiation signals to the brain. While contributions of the DVC are increasingly investigated, a role for DVC YR in control of food intake has not been examined systematically. We used in situ hybridization to confirm expression of Y1R and Y2R, but not Y5R, in the DVC and vagal afferent neurons. We found that nanoinjections of a Y2R agonist, PYY-(3-36), into the DVC significantly increased food intake over a 4-h period in satiated male rats. PYY-(3-36)-evoked food intake was prevented by injection of a selective Y2R antagonist. Injection of a Y1R/Y5R-preferring agonist into the DVC failed to increase food intake at doses reported to increase food intake following hypothalamic injection. Finally, injection of PYY-(3-36) into the DVC prevented reduction of 30-min food intake following intraperitoneal injection of cholecystokinin (CCK). Our results indicate that activation of DVC Y2R, unlike hypothalamic or peripheral Y2R, increases food intake. Furthermore, in the context of available electrophysiological observations, our results are consistent with the hypothesis that DVC Y2R control food intake by dampening vagally mediated satiation signals in the DVC.
Collapse
Affiliation(s)
- Nathaneal J Huston
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| | - Lynne A Brenner
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| | - Zachary C Taylor
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| | - Robert C Ritter
- Department of Integrative Physiology and Neuroscience, Washington State University , Pullman, Washington
| |
Collapse
|
53
|
Kjaergaard M, Salinas CBG, Rehfeld JF, Secher A, Raun K, Wulff BS. PYY(3-36) and exendin-4 reduce food intake and activate neuronal circuits in a synergistic manner in mice. Neuropeptides 2019; 73:89-95. [PMID: 30471778 DOI: 10.1016/j.npep.2018.11.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/17/2018] [Accepted: 11/18/2018] [Indexed: 12/11/2022]
Abstract
Peptide YY(3-36) ((PYY(3-36)) and glucagon like peptide 1 (GLP-1) in combination reduce food intake and body weight in an additive or synergistic manner in animal models and in humans. Nevertheless, the mechanisms behind are not completely understood. The present study aims to investigate the effect of combining PYY(3-36) and the GLP-1 receptor agonist exendin-4 (Ex4) by examining acute food intake and global neuronal activation as measured by c-fos in C57BL/6 J mice. An additive reduction in food intake was found 1.5 h after s.c dosing with the combination of PYY(3-36) (200 μg/kg) and Ex4 (2.5 μg/kg). This was associated with a synergistic enhancement of c-fos reactivity in central amygdalar nucleus (CeA), rostral part of the mediobasal arcuate nucleus (ARH), supratrigeminal nucleus (SUT), lateral parabrachial nucleus (PB), area postrema (AP) and nucleus tractus solitarius (NTS) compared to vehicle, PYY(3-36) and Ex4 individually dosed mice. The regions activated by Ex4 individually and PYY(3-36) and Ex4 in combination resembled each other, but the combination group had a significantly stronger c-fos response. Twenty-five brain areas were activated by PYY(3-36) and Ex4 in combination compared to vehicle versus nine brain areas by Ex4 individually. No significant increase in c-fos reactivity was found by PYY(3-36) compared to vehicle dosed mice. The neuronal activation of ARH and the AP/NTS to PB to CeA pathway is important for appetite regulation while SUT has not previously been reported in the regulation of energy balance. As PYY(3-36) and Ex4 act on different neurons leading to recruitment of different signalling pathways within and to the brain, an interaction of these pathways may contribute to their additive/synergistic action. Thus, PYY(3-36) boosts the effect of Ex4 possibly by inducing less inhibition of neuronal activity leading to an enhanced neuronal activity induced by Ex4.
Collapse
Affiliation(s)
- Marina Kjaergaard
- Histology and Imaging, Novo Nordisk A/S, 2760 Måløv, Denmark.; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark..
| | | | - Jens F Rehfeld
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, 2200 Copenhagen N, Denmark
| | - Anna Secher
- Histology and Imaging, Novo Nordisk A/S, 2760 Måløv, Denmark
| | - Kirsten Raun
- Obesity Research, Novo Nordisk A/S, 2760 Måløv, Denmark
| | | |
Collapse
|
54
|
Huang S, Li Z, Liu Y, Gao D, Zhang X, Hao J, Yang F. Neural regulation of bone remodeling: Identifying novel neural molecules and pathways between brain and bone. J Cell Physiol 2018; 234:5466-5477. [PMID: 29377116 DOI: 10.1002/jcp.26502] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 01/20/2018] [Indexed: 02/05/2023]
Affiliation(s)
- Shishu Huang
- Department of Orthopaedic Surgery West China Hospital, Sichuan University Chengdu China
| | - Zhenxia Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Department of Orthodontics West China Hospital of Stomatology, Sichuan University Chengdu China
| | - Yunhui Liu
- The Brain Cognition & Brain Disease Institute, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen China
| | - Dashuang Gao
- The Brain Cognition & Brain Disease Institute, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen China
| | - Xinzhou Zhang
- Department of Nephrology Shenzhen People's Hospital, Second Clinical Medical College, Jinan University Shenzhen China
| | - Jin Hao
- Program in Biological Sciences in Dental Medicine, Harvard School of Dental Medicine Boston Massachusetts
| | - Fan Yang
- The Brain Cognition & Brain Disease Institute, Shenzhen Institutes of Advanced Technology Chinese Academy of Sciences Shenzhen China
| |
Collapse
|
55
|
Endocannabinoid and nitric oxide systems of the hypothalamic paraventricular nucleus mediate effects of NPY on energy expenditure. Mol Metab 2018; 18:120-133. [PMID: 30274714 PMCID: PMC6308028 DOI: 10.1016/j.molmet.2018.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/14/2018] [Accepted: 08/22/2018] [Indexed: 12/27/2022] Open
Abstract
Objective Neuropeptide Y (NPY) is one of the most potent orexigenic peptides. The hypothalamic paraventricular nucleus (PVN) is a major locus where NPY exerts its effects on energy homeostasis. We investigated how NPY exerts its effect within the PVN. Methods Patch clamp electrophysiology and Ca2+ imaging were used to understand the involvement of Ca2+ signaling and retrograde transmitter systems in the mediation of NPY induced effects in the PVN. Immuno-electron microscopy were performed to elucidate the subcellular localization of the elements of nitric oxide (NO) system in the parvocellular PVN. In vivo metabolic profiling was performed to understand the role of the endocannabinoid and NO systems of the PVN in the mediation of NPY induced changes of energy homeostasis. Results We demonstrated that NPY inhibits synaptic inputs of parvocellular neurons in the PVN by activating endocannabinoid and NO retrograde transmitter systems via mobilization of Ca2+ from the endoplasmic reticulum, suggesting that NPY gates the synaptic inputs of parvocellular neurons in the PVN to prevent the influence of non-feeding-related inputs. While intraPVN administered NPY regulates food intake and locomotor activity via NO signaling, the endocannabinoid system of the PVN selectively mediates NPY-induced decrease in energy expenditure. Conclusion Thus, within the PVN, NPY stimulates the release of endocannabinoids and NO via Ca2+-influx from the endoplasmic reticulum. Both transmitter systems appear to have unique roles in the mediation of the NPY-induced regulation of energy homeostasis, suggesting that NPY regulates food intake, energy expenditure, and locomotor activity through different neuronal networks of this nucleus. NPY increases the intracellular Ca2+ level of PVN neurons by mobilizing the Ca2+ from ER. NPY inhibits the input of these neurons by endocannabinoids and NO. IntraPVN administered NPY regulates food intake and locomotor activity via NO signaling. IntraPVN administered NPY regulates energy expenditure via the endocannabinoid system. NPY regulates the energy expenditure and food intake via different neuronal networks of the PVN.
Collapse
|
56
|
Neurobiological characteristics underlying metabolic differences between males and females. Prog Neurobiol 2018; 176:18-32. [PMID: 30194984 DOI: 10.1016/j.pneurobio.2018.09.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 08/22/2018] [Accepted: 09/01/2018] [Indexed: 12/24/2022]
Abstract
The hypothalamus is the main integrating center for metabolic control. Our understanding of how hypothalamic circuits function to control appetite and energy expenditure has increased dramatically in recent years, due to the rapid rise in the incidence of obesity and the search for effective treatments. Increasing evidence indicates that these treatments will most likely differ between males and females. Indeed, sex differences in metabolism have been demonstrated at various levels, including in two of the most studied neuronal populations involved in metabolic control: the anorexigenic proopiomelanocortin neurons and the orexigenic neuropeptide Y/Agouti-related protein neurons. Here we review what is known to date regarding the sex differences in these two neuronal populations, as well as other neuronal populations involved in metabolic control and glial cells.
Collapse
|
57
|
Liu X, Liu H, Xiong Y, Yang L, Wang C, Zhang R, Zhu X. Postmenopausal osteoporosis is associated with the regulation of SP, CGRP, VIP, and NPY. Biomed Pharmacother 2018; 104:742-750. [PMID: 29807224 DOI: 10.1016/j.biopha.2018.04.044] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 12/13/2022] Open
Abstract
Estrogen deficiency is the main factor underlying postmenopausal osteoporosis. A large number of neuropeptides, which regulate skeletal metabolism, potentially represent a regulatory pathway for the pathogenesis of osteoporosis. The aim of this study was to explore factors involved in the regulation of bone-related neuropeptides and their association with estrogen deficiency and bone metabolism. Thirty adult female Sprague-Dawley (SD) rats were randomly divided into a control group with sham surgery (n = 15) and an ovariectomy group with bilateral oophorectomy (n = 15). After 16 weeks, serum estrogen was reduced,CTX-1 was increased and P1NP was not significantly affected in the ovariectomy group and a model of osteoporosis was established. We then investigate the gene expression and protein levels of a range of neuropeptides and their receptors, including substance P (SP) and tachykinin receptor 1 (TACR1), calcitonin gene-related peptide (CGRP) and calcitonin receptor-like (CALCRL), vasoactive intestinal polypeptide (VIP) and receptor 1 and 2 (VPAC1, 2), neuropeptide Y (NPY) and receptor Y1 and Y2, in the brain and femora. Ovariectomy reduced TACR1, CGRP, CALCRL, NPY, NPY Y2 in the brain, but increased TACR1 and decreased SP, CALCRL, VIP, VPAC2 in the bone. Collectively, our data revealed that the pathogenesis of postmenopausal osteoporosis is associated with the regulation of SP, CGRP, VIP, and NPY. These novel results are of significant importance in the development of neuropeptides as therapeutic targets.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Jinan University College of Traditional Chinese Medicine, Guangzhou 510632, PR China
| | - Hengrui Liu
- Jinan University College of Pharmacy, Guangzhou 510632, PR China
| | - Yingquan Xiong
- Jinan University College of Pharmacy, Guangzhou 510632, PR China
| | - Li Yang
- Jinan University College of Pharmacy, Guangzhou 510632, PR China
| | - Chaopeng Wang
- Jinan University College of Pharmacy, Guangzhou 510632, PR China
| | - Ronghua Zhang
- Jinan University College of Pharmacy, Guangzhou 510632, PR China.
| | - Xiaofeng Zhu
- First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510632, PR China.
| |
Collapse
|
58
|
Bliss ES, Whiteside E. The Gut-Brain Axis, the Human Gut Microbiota and Their Integration in the Development of Obesity. Front Physiol 2018; 9:900. [PMID: 30050464 PMCID: PMC6052131 DOI: 10.3389/fphys.2018.00900] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Accepted: 06/21/2018] [Indexed: 12/17/2022] Open
Abstract
Obesity is a global epidemic, placing socioeconomic strain on public healthcare systems, especially within the so-called Western countries, such as Australia, United States, United Kingdom, and Canada. Obesity results from an imbalance between energy intake and energy expenditure, where energy intake exceeds expenditure. Current non-invasive treatments lack efficacy in combating obesity, suggesting that obesity is a multi-faceted and more complex disease than previously thought. This has led to an increase in research exploring energy homeostasis and the discovery of a complex bidirectional communication axis referred to as the gut-brain axis. The gut-brain axis is comprised of various neurohumoral components that allow the gut and brain to communicate with each other. Communication occurs within the axis via local, paracrine and/or endocrine mechanisms involving a variety of gut-derived peptides produced from enteroendocrine cells (EECs), including glucagon-like peptide 1 (GLP1), cholecystokinin (CCK), peptide YY3-36 (PYY), pancreatic polypeptide (PP), and oxyntomodulin. Neural networks, such as the enteric nervous system (ENS) and vagus nerve also convey information within the gut-brain axis. Emerging evidence suggests the human gut microbiota, a complex ecosystem residing in the gastrointestinal tract (GIT), may influence weight-gain through several inter-dependent pathways including energy harvesting, short-chain fatty-acids (SCFA) signalling, behaviour modifications, controlling satiety and modulating inflammatory responses within the host. Hence, the gut-brain axis, the microbiota and the link between these elements and the role each plays in either promoting or regulating energy and thereby contributing to obesity will be explored in this review.
Collapse
Affiliation(s)
- Edward S. Bliss
- School of Health and Wellbeing, University of Southern Queensland, Toowoomba, QLD, Australia
| | | |
Collapse
|
59
|
Domingues MF, de Assis DR, Piovesan AR, Belo CAD, da Costa JC. Peptide YY (3-36) modulates intracellular calcium through activation of the phosphatidylinositol pathway in hippocampal neurons. Neuropeptides 2018; 67:1-8. [PMID: 29157865 DOI: 10.1016/j.npep.2017.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Revised: 11/03/2017] [Accepted: 11/05/2017] [Indexed: 10/18/2022]
Abstract
Peptide YY (PYY) belongs to the neuropeptide Y (NPY) family, which also includes the pancreatic polypeptide (PP) and NPY. PYY is secreted by the intestinal L cells, being present in the blood stream in two active forms capable of crossing the blood brain barrier, PYY (1-36) and its cleavage product, PYY (3-36). PYY is a selective agonist for the Y2 receptor (Y2R) and these receptors are abundant in the hippocampus. Here we investigated the mechanisms by which PYY (3-36) regulates intracellular Ca2+ concentrations ([Ca2+]i) in hippocampal neurons by employing a calcium imaging technique in hippocampal cultures. Alterations in [Ca2+]i were detected by changes in the Fluo-4 AM reagent emission. PYY (3-36) significantly increased [Ca2+] from the concentration of 10-11M as compared to the controls (infusion of HEPES-buffered solution (HBS) solution alone). The PYY (3-36)-increase in [Ca2+]i remained unchanged even in Ca2+-free extracellular solutions. Sarcoplasmic/endoplasmic reticulum Ca2+-ATPase pump (SERCA pump) inhibition partially prevent the PYY (3-36)-increase of [Ca2+]i and inositol 1,4,5-triphosphate receptor (IP3R) inhibition also decreased the PYY (3-36)-increase of [Ca2+]i. Taken together, our data strongly suggest that PYY (3-36) mobilizes calcium from the neuronal endoplasmic reticulum (ER) stores towards the cytoplasm. Next, we showed that PYY (3-36) inhibited high K+-induced increases of [Ca2+]i, suggesting that PYY (3-36) could also act by activating G-protein coupled inwardly rectifying potassium K+ channels. Finally, the co-infusion of the Y2 receptor (Y2R) antagonist BIIE0246 with PYY (3-36) abolished the [Ca2+]i increase induced by the peptide, suggesting that PYY (3-36)-induced [Ca2+]i increase in hippocampal neurons occurs via Y2Rs.
Collapse
Affiliation(s)
- Michelle Flores Domingues
- Graduate Program in Cellular and Molecular Biology - Center for Biotechnology, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, Brazil; Brain Institute (BraIns), Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, Brazil
| | - Dênis Reis de Assis
- Brain Institute (BraIns), Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, Brazil
| | - Angela Regina Piovesan
- Graduate Program in Cellular and Molecular Biology - Center for Biotechnology, Universidade Federal do Rio Grande do Sul, UFRGS, Porto Alegre, Brazil; Brain Institute (BraIns), Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, Brazil
| | - Cháriston André Dal Belo
- Brain Institute (BraIns), Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, Brazil; Laboratory of Neurobiology and Toxinology, LANETOX, Universidade Federal do Pampa, UNIPAMPA, São Gabriel, Brazil; Graduate Program in Biological Sciences: Biochemical Toxicology, PPGBTox, Universidade Federal de Santa Maria, UFSM, Santa Maria, Brazil.
| | - Jaderson Costa da Costa
- Brain Institute (BraIns), Pontifícia Universidade Católica do Rio Grande do Sul, PUCRS, Porto Alegre, Brazil
| |
Collapse
|
60
|
Manfredi-Lozano M, Roa J, Tena-Sempere M. Connecting metabolism and gonadal function: Novel central neuropeptide pathways involved in the metabolic control of puberty and fertility. Front Neuroendocrinol 2018; 48:37-49. [PMID: 28754629 DOI: 10.1016/j.yfrne.2017.07.008] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 07/20/2017] [Accepted: 07/23/2017] [Indexed: 02/08/2023]
Abstract
Albeit essential for perpetuation of species, reproduction is an energy-demanding function that can be adjusted to body metabolic status. Reproductive maturation and function can be suppressed in conditions of energy deficit, but can be altered also in situations of persistent energy excess, e.g., morbid obesity. This metabolic-reproductive integration, of considerable pathophysiological relevance to explain different forms of perturbed puberty and sub/infertility, is implemented by the concerted action of numerous central and peripheral regulators, which impinge at different levels of the hypothalamic-pituitary-gonadal (HPG) axis, permitting a tight fit between nutritional/energy status and gonadal function. We summarize here the major physiological mechanisms whereby nutritional and metabolic cues modulate the maturation and function of the HPG axis. We will focus on recent progress on the major central neuropeptide pathways, including kisspeptins, neurokinin B and the products of POMC and NPY neurons, which convey metabolic information to GnRH neurons, as major hierarchical hub of our reproductive brain.
Collapse
Affiliation(s)
- M Manfredi-Lozano
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain; Laboratory of Development and Plasticity of the Neuroendocrine Brain, Jean-Pierre Aubert Research Centre, INSERM, U1172, Lille, France
| | - J Roa
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain.
| | - M Tena-Sempere
- Instituto Maimónides de Investigación Biomédica de Cordoba (IMIBIC), Spain; Department of Cell Biology, Physiology and Immunology, University of Cordoba, Spain; Hospital Universitario Reina Sofia, 14004 Cordoba, Spain; CIBER Fisiopatología de la Obesidad y Nutrición, Instituto de Salud Carlos III, 14004 Cordoba, Spain; FiDiPro Program, Department of Physiology, University of Turku, Kiinamyllynkatu 10, FIN-20520 Turku, Finland.
| |
Collapse
|
61
|
Reiner DJ, Mietlicki-Baase EG, Olivos DR, McGrath LE, Zimmer DJ, Koch-Laskowski K, Krawczyk J, Turner CA, Noble EE, Hahn JD, Schmidt HD, Kanoski SE, Hayes MR. Amylin Acts in the Lateral Dorsal Tegmental Nucleus to Regulate Energy Balance Through Gamma-Aminobutyric Acid Signaling. Biol Psychiatry 2017; 82:828-838. [PMID: 28237459 PMCID: PMC5503810 DOI: 10.1016/j.biopsych.2016.12.028] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 12/13/2016] [Accepted: 12/28/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND The pancreatic- and brain-derived hormone amylin promotes negative energy balance and is receiving increasing attention as a promising obesity therapeutic. However, the neurobiological substrates mediating amylin's effects are not fully characterized. We postulated that amylin acts in the lateral dorsal tegmental nucleus (LDTg), an understudied neural processing hub for reward and homeostatic feeding signals. METHODS We used immunohistochemical and quantitative polymerase chain reaction analyses to examine expression of the amylin receptor complex in rat LDTg tissue. Behavioral experiments were performed to examine the mechanisms underlying the hypophagic effects of amylin receptor activation in the LDTg. RESULTS Immunohistochemical and quantitative polymerase chain reaction analyses show expression of the amylin receptor complex in the LDTg. Activation of LDTg amylin receptors by the agonist salmon calcitonin dose-dependently reduces body weight, food intake, and motivated feeding behaviors. Acute pharmacological studies and longer-term adeno-associated viral knockdown experiments indicate that LDTg amylin receptor signaling is physiologically and potentially preclinically relevant for energy balance control. Finally, immunohistochemical data indicate that LDTg amylin receptors are expressed on gamma-aminobutyric acidergic neurons, and behavioral results suggest that local gamma-aminobutyric acid receptor signaling mediates the hypophagia after LDTg amylin receptor activation. CONCLUSIONS These findings identify the LDTg as a novel nucleus with therapeutic potential in mediating amylin's effects on energy balance through gamma-aminobutyric acid receptor signaling.
Collapse
Affiliation(s)
- David J Reiner
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Elizabeth G Mietlicki-Baase
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Diana R Olivos
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Lauren E McGrath
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Derek J Zimmer
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Kieran Koch-Laskowski
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Joanna Krawczyk
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Christopher A Turner
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania; Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Emily E Noble
- Department of Biological Sciences, Human and Evolutionary Biology Section, Los Angeles, California
| | - Joel D Hahn
- Neurobiology Section, University of Southern California, Los Angeles, California
| | - Heath D Schmidt
- Department of Biobehavioral Health Sciences, School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Scott E Kanoski
- Department of Biological Sciences, Human and Evolutionary Biology Section, Los Angeles, California
| | - Matthew R Hayes
- Translational Neuroscience Program, Department of Psychiatry, Perelman School of Medicine, Philadelphia, Pennsylvania.
| |
Collapse
|
62
|
Abstract
Although the brain is well established as a master regulator of homeostasis in peripheral tissues, central regulation of bone mass represents a novel and rapidly expanding field of study. This review examines the current understanding of central regulation of the skeleton, exploring several of the key pathways connecting brain to bone and their implications both in mice and the clinical setting. Our understanding of central bone regulation has largely progressed through examination of skeletal responses downstream of nutrient regulatory pathways in the hypothalamus. Mutations and modulation of these pathways, in cases such as leptin deficiency, induce marked bone phenotypes, which have provided vital insights into central bone regulation. These studies have identified several central neuropeptide pathways that stimulate well-defined changes in bone cell activity in response to changes in energy homeostasis. In addition, this work has highlighted the endocrine nature of the skeleton, revealing a complex cross talk that directly regulates other organ systems. Our laboratory has studied bone-active neuropeptide pathways and defined osteoblast-based actions that recapitulate central pathways linking bone, fat, and glucose homeostasis. Studies of neural control of bone have produced paradigm-shifting changes in our understanding of the skeleton and its relationship with the wider array of organ systems.
Collapse
Affiliation(s)
- Alexander Corr
- 1 The Division of Bone Biology, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,2 Faculty of Science, University of Bath, Bath, United Kingdom
| | - James Smith
- 1 The Division of Bone Biology, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,2 Faculty of Science, University of Bath, Bath, United Kingdom
| | - Paul Baldock
- 1 The Division of Bone Biology, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.,3 Faculty of Medicine, St Vincent's Clinical School, University of New South Wales, Sydney, New South Wales, Australia.,4 School of Medicine Sydney, University of Notre Dame Australia, Sydney, New South Wales, Australia
| |
Collapse
|
63
|
Thomas MA, Xue B. Mechanisms for AgRP neuron-mediated regulation of appetitive behaviors in rodents. Physiol Behav 2017; 190:34-42. [PMID: 29031550 DOI: 10.1016/j.physbeh.2017.10.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2017] [Revised: 08/29/2017] [Accepted: 10/06/2017] [Indexed: 12/19/2022]
Abstract
The obesity epidemic is a major health and economic burden facing both developed and developing countries worldwide. Interrogation of the central and peripheral mechanisms regulating ingestive behaviors have primarily focused on food intake, and in the process uncovered a detailed neuroanatomical framework controlling this behavior. However, these studies have largely ignored the behaviors that bring animals, including humans, in contact with food. It is therefore useful to dichotomize ingestive behaviors as appetitive (motivation to find and store food) and consummatory (consumption of food once found), and utilize an animal model that naturally displays these behaviors. Recent advances in genetics have facilitated the identification of several neuronal populations critical for regulating ingestive behaviors in mice, and novel functions of these neurons and neuropeptides in regulating appetitive behaviors in Siberian hamsters, a natural model of food foraging and food hoarding, have been identified. To this end, hypothalamic agouti-related protein/neuropeptide Y expressing neurons (AgRP neurons) have emerged as a critical regulator of ingestive behaviors. Recent studies by Dr. Timothy Bartness and others have identified several discrete mechanisms through which peripheral endocrine signals regulate AgRP neurons to control food foraging, food hoarding, and food intake. We review here recent advances in our understanding of the neuroendocrine control of ingestive behaviors in Siberian hamsters and other laboratory rodents, and identify novel mechanisms through which AgRP neurons mediate appetitive and consummatory behaviors.
Collapse
Affiliation(s)
- M Alex Thomas
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA
| | - Bingzhong Xue
- Department of Biology, Georgia State University, Atlanta, GA 30302, USA; Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302, USA; Neuroscience Institute, Georgia State University, Atlanta, GA 30302, USA.
| |
Collapse
|
64
|
Guarino D, Nannipieri M, Iervasi G, Taddei S, Bruno RM. The Role of the Autonomic Nervous System in the Pathophysiology of Obesity. Front Physiol 2017; 8:665. [PMID: 28966594 PMCID: PMC5606212 DOI: 10.3389/fphys.2017.00665] [Citation(s) in RCA: 161] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/22/2017] [Indexed: 12/18/2022] Open
Abstract
Obesity is reaching epidemic proportions globally and represents a major cause of comorbidities, mostly related to cardiovascular disease. The autonomic nervous system (ANS) dysfunction has a two-way relationship with obesity. Indeed, alterations of the ANS might be involved in the pathogenesis of obesity, acting on different pathways. On the other hand, the excess weight induces ANS dysfunction, which may be involved in the haemodynamic and metabolic alterations that increase the cardiovascular risk of obese individuals, i.e., hypertension, insulin resistance and dyslipidemia. This article will review current evidence about the role of the ANS in short-term and long-term regulation of energy homeostasis. Furthermore, an increased sympathetic activity has been demonstrated in obese patients, particularly in the muscle vasculature and in the kidneys, possibily contributing to increased cardiovascular risk. Selective leptin resistance, obstructive sleep apnea syndrome, hyperinsulinemia and low ghrelin levels are possible mechanisms underlying sympathetic activation in obesity. Weight loss is able to reverse metabolic and autonomic alterations associated with obesity. Given the crucial role of autonomic dysfunction in the pathophysiology of obesity and its cardiovascular complications, vagal nerve modulation and sympathetic inhibition may serve as therapeutic targets in this condition.
Collapse
Affiliation(s)
- Daniela Guarino
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy.,Institute of Clinical Physiology of CNRPisa, Italy.,Scuola Superiore Sant'AnnaPisa, Italy
| | - Monica Nannipieri
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | | | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Rosa Maria Bruno
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| |
Collapse
|
65
|
Robinson SL, Thiele TE. The Role of Neuropeptide Y (NPY) in Alcohol and Drug Abuse Disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2017; 136:177-197. [PMID: 29056151 DOI: 10.1016/bs.irn.2017.06.005] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neuropeptide Y (NPY) is a neuromodulator that is widely expressed throughout the central nervous system (CNS) and which is cosecreted with classic neurotransmitters including GABA and glutamate. There is a long history of research implicating a role for NPY in modulating neurobiological responses to alcohol (ethanol) as well as other drugs of abuse. Both ethanol exposure and withdrawal from chronic ethanol have been shown to produce changes in NPY and NPY receptor protein levels and mRNA expression in the CNS. Importantly, manipulations of NPY Y1 and Y2 receptor signaling have been shown to alter ethanol consumption and self-administration in a brain region-specific manner, with Y1 receptor activation and Y2 receptor blockade in regions of the extended amygdala promoting robust reductions of ethanol intake. Similar observations have been made in studies examining neurobiological responses to nicotine, psychostimulants, and opioids. When taken together with observations of potential genetic linkage between the NPY system and the human alcohol abuse disorders, NPY represents a promising target for treating problematic alcohol and drug use, and in protecting individuals from relapse during abstinence.
Collapse
Affiliation(s)
- Stacey L Robinson
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Todd E Thiele
- University of North Carolina at Chapel Hill, Chapel Hill, NC, United States.
| |
Collapse
|
66
|
Kornhuber J, Zoicas I. Neuropeptide Y prolongs non-social memory and differentially affects acquisition, consolidation, and retrieval of non-social and social memory in male mice. Sci Rep 2017; 7:6821. [PMID: 28754895 PMCID: PMC5533709 DOI: 10.1038/s41598-017-07273-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 06/23/2017] [Indexed: 01/08/2023] Open
Abstract
Neuropeptide Y (NPY) and its receptors (especially Y1, Y2, and Y5) are highly expressed in brain regions involved in learning and memory processes. Accordingly, NPY was shown to modulate cognitive functions in rodents. Here, we investigated possible memory-enhancing effects of NPY and determined the role of the NPY system in the acquisition, consolidation, and retrieval of non-social and social memory in mice, using the object and social discrimination tests, respectively. Intracerebroventricular (icv) infusion of NPY (1 nmol/2 µl) prolonged retention of non-social (object) memory, but not of social memory. This effect was blocked by the Y1 receptor antagonist BIBO3304 trifluoroacetate (2 nmol/2 µl), but not by the Y2 receptor antagonist BIIE0246 (2 nmol/2 µl). While icv infusion of NPY did not affect the acquisition, consolidation, and retrieval of non-social and social memory, icv infusion of BIBO3304 trifluoroacetate and BIIE0246 blocked the consolidation of non-social memory and the retrieval of both non-social and social memory. This study suggests that NPY has memory-enhancing effects in a non-social context by specifically acting on Y1 receptors. It further suggests that the central NPY system exerts differential effects on the sequential phases of non-social and social memory.
Collapse
Affiliation(s)
- Johannes Kornhuber
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, 91054, Erlangen, Germany
| | - Iulia Zoicas
- Department of Psychiatry and Psychotherapy, University Hospital, Friedrich-Alexander-University Erlangen-Nuremberg, 91054, Erlangen, Germany.
| |
Collapse
|
67
|
Abstract
The rising incidence of metabolic diseases worldwide has prompted renewed interest in the study of intermediary metabolism and cellular bioenergetics. The application of modern biochemical methods for quantitating fuel substrate metabolism with advanced mouse genetic approaches has greatly increased understanding of the mechanisms that integrate energy metabolism in the whole organism. Examination of the intermediary metabolism of skeletal cells has been sparked by a series of unanticipated observations in genetically modified mice that suggest the existence of novel endocrine pathways through which bone cells communicate their energy status to other centers of metabolic control. The recognition of this expanded role of the skeleton has in turn led to new lines of inquiry directed at defining the fuel requirements and bioenergetic properties of bone cells. This article provides a comprehensive review of historical and contemporary studies on the metabolic properties of bone cells and the mechanisms that control energy substrate utilization and bioenergetics. Special attention is devoted to identifying gaps in our current understanding of this new area of skeletal biology that will require additional research to better define the physiological significance of skeletal cell bioenergetics in human health and disease.
Collapse
Affiliation(s)
- Ryan C Riddle
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, Maryland; and The Baltimore Veterans Administration Medical Center, Baltimore, Maryland
| | - Thomas L Clemens
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, Maryland; and The Baltimore Veterans Administration Medical Center, Baltimore, Maryland
| |
Collapse
|
68
|
Goadsby PJ, Holland PR, Martins-Oliveira M, Hoffmann J, Schankin C, Akerman S. Pathophysiology of Migraine: A Disorder of Sensory Processing. Physiol Rev 2017; 97:553-622. [PMID: 28179394 PMCID: PMC5539409 DOI: 10.1152/physrev.00034.2015] [Citation(s) in RCA: 1146] [Impact Index Per Article: 143.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Plaguing humans for more than two millennia, manifest on every continent studied, and with more than one billion patients having an attack in any year, migraine stands as the sixth most common cause of disability on the planet. The pathophysiology of migraine has emerged from a historical consideration of the "humors" through mid-20th century distraction of the now defunct Vascular Theory to a clear place as a neurological disorder. It could be said there are three questions: why, how, and when? Why: migraine is largely accepted to be an inherited tendency for the brain to lose control of its inputs. How: the now classical trigeminal durovascular afferent pathway has been explored in laboratory and clinic; interrogated with immunohistochemistry to functional brain imaging to offer a roadmap of the attack. When: migraine attacks emerge due to a disorder of brain sensory processing that itself likely cycles, influenced by genetics and the environment. In the first, premonitory, phase that precedes headache, brain stem and diencephalic systems modulating afferent signals, light-photophobia or sound-phonophobia, begin to dysfunction and eventually to evolve to the pain phase and with time the resolution or postdromal phase. Understanding the biology of migraine through careful bench-based research has led to major classes of therapeutics being identified: triptans, serotonin 5-HT1B/1D receptor agonists; gepants, calcitonin gene-related peptide (CGRP) receptor antagonists; ditans, 5-HT1F receptor agonists, CGRP mechanisms monoclonal antibodies; and glurants, mGlu5 modulators; with the promise of more to come. Investment in understanding migraine has been very successful and leaves us at a new dawn, able to transform its impact on a global scale, as well as understand fundamental aspects of human biology.
Collapse
Affiliation(s)
- Peter J Goadsby
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| | - Philip R Holland
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| | - Margarida Martins-Oliveira
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| | - Jan Hoffmann
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| | - Christoph Schankin
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| | - Simon Akerman
- Basic and Clinical Neurosciences, Institute of Psychiatry, Psychology and Neuroscience, King's College, London, United Kingdom; Department of Neurology, University of California, San Francisco, San Francisco, California; Department of Neurology, University of Hamburg-Eppendorf, Hamburg, Germany; and Department of Neurology, University Hospital Bern-Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|
69
|
Abstract
In vivo electrophysiology in migraine animal model shows that neuropeptide Y dose dependently inhibits dural-evoked trigeminal activity, through NPY Y1 receptor activation. Implications for pain and premonitory symptomatology. Migraine is a painful neurologic disorder with premonitory symptomatology that can include disturbed appetite. Migraine pathophysiology involves abnormal activation of trigeminocervical complex (TCC) neurons. Neuropeptide Y (NPY) is synthesized in the brain and is involved in pain modulation. NPY receptors are present in trigeminal ganglia and trigeminal nucleus caudalis suggesting a role in migraine pathophysiology. The present study aimed to determine the effect of systemic administration of NPY on TCC neuronal activity in response to dural nociceptive trigeminovascular activation. We performed in vivo electrophysiology in anesthetized rats, administered NPY (10, 30, and 100 µg·kg−1), and investigated the receptors involved by studying NPY Y1 (30 µg·kg−1), Y2 (30 µg·kg−1), and Y5 receptor agonists (100·µg·kg−1), and NPY Y1 receptor antagonist (30 µg·kg−1). NPY (30 and 100 µg·kg−1) significantly reduced TCC neuronal firing in response to dural-evoked trigeminovascular activation, but only NPY (30 µg·kg−1) significantly reduced spontaneous trigeminal firing. NPY Y1 receptor agonist also significantly reduced dural-evoked and spontaneous TCC neuronal firing. NPY (10 µg·kg−1), NPY Y2, and Y5 receptor agonists, and the NPY Y1 receptor antagonist had no significant effects on nociceptive dural-evoked neuronal firing in the TCC or spontaneous trigeminal firing. This study demonstrates that NPY dose dependently inhibits dural-evoked trigeminal activity, through NPY Y1 receptor activation, indicating antinociceptive actions of NPY in a migraine animal model. Based on the role of NPY in appetite regulation, it is possible that disruption of the NPY system might explain changes of appetite in migraineurs.
Collapse
|
70
|
Aoki K, Kondo M, Okuda M, Saneyasu T, Honda K, Kamisoyama H. Identification, expression analysis, and functional characterization of peptide YY in chickens (Gallus gallus domesticus). Gen Comp Endocrinol 2017; 242:11-17. [PMID: 27118705 DOI: 10.1016/j.ygcen.2016.04.021] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 04/05/2016] [Accepted: 04/22/2016] [Indexed: 01/04/2023]
Abstract
Peptide YY (PYY) functions as a postprandial satiety signal in mammals. However, the genomic information and physiological roles of chicken PYY have not yet been clarified, although PYY peptide was isolated from chicken intestines in 1992. In this study, we identified a full-length complementary DNA (cDNA) sequence encoding the chicken PYY precursor. The deduced amino acid sequence of chicken PYY was completely consistent with the previously identified peptide sequence. PYY mRNA was abundantly expressed in the small intestine compared with the large intestine. PYY mRNA levels in the jejunum were significantly higher during ad libitum feeding compared with fasting, suggesting that intestinal PYY expression is altered in response to nutritional status in chicks. Intravenous administration of PYY significantly suppressed food intake in chicks. Furthermore, neuropeptide Y receptor Y2, a possible target of PYY, was expressed in various brain regions including the appetite-regulating centers in chicks. This is the first evidence that the intestinal hormone PYY may function as an anorexigenic hormone in chicks.
Collapse
Affiliation(s)
- Koji Aoki
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Makoto Kondo
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Mika Okuda
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Takaoki Saneyasu
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| | - Kazuhisa Honda
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan.
| | - Hiroshi Kamisoyama
- Department of Bioresource Science, Graduate School of Agricultural Science, Kobe University, Kobe 657-8501, Japan
| |
Collapse
|
71
|
Murase SI, Shiiya T, Higuchi H. Neuropeptide Y Y 5 receptor localization in mouse central nervous system. Brain Res 2017; 1655:216-232. [PMID: 27984021 DOI: 10.1016/j.brainres.2016.10.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2016] [Revised: 10/12/2016] [Accepted: 10/27/2016] [Indexed: 10/20/2022]
Abstract
Neuropeptide Y (NPY) and its receptors affect blood pressure, feeding behavior, and neurogenesis. In this study, the distribution of neurons expressing NPY Y5 receptor (Y5) was examined in adult mouse central nervous system by immunohistochemistry. Y5 protein localization was investigated using polyclonal anti-Y5 antibody, which was successfully preabsorbed with Y5 knockout brain tissues. The preabsorbed anti-Y5 antibody did not react with Y5 knockout brain tissues, thus meeting the "hard specificity criterion," which is the absence of staining in tissues genetically deficient for the antigen (Pradidarcheep et al., 2008). Y5-positive neurons were found in most brain areas. Most Y5 immunoreactivities were observed as dot-like structures adjacent to the plasma membrane, as expected for a cell membrane receptor. In situ hybridization showed that the Y5 mRNA expression was correlated with the Y5 protein level in each case and that it was probably controlled by the transcriptional regulation of the Y5 gene. In the nuclei where Y5 was expressed, Y5 immunoreactivities were found mainly in the somatic and dendritic areas. The distribution patterns of the Y5-positive cells that were broader than previously expected suggest important biological activities of the Y5 in many brain areas.
Collapse
Affiliation(s)
- Shin-Ichi Murase
- Division of Pharmacology, Niigata University, Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| | - Tomohiro Shiiya
- Division of Pharmacology, Niigata University, Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Hiroshi Higuchi
- Division of Pharmacology, Niigata University, Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan.
| |
Collapse
|
72
|
Gao S, Zhang J, He C, Meng F, Bu G, Zhu G, Li J, Wang Y. Molecular characterization of neuropeptide Y (NPY) receptors (Y1, Y4 and Y6) and investigation of the tissue expression of their ligands (NPY, PYY and PP) in chickens. Gen Comp Endocrinol 2017; 240:46-60. [PMID: 27641685 DOI: 10.1016/j.ygcen.2016.09.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Revised: 08/05/2016] [Accepted: 09/14/2016] [Indexed: 01/06/2023]
Abstract
Neuropeptide Y (NPY) receptors and its ligands, NPY, peptide YY (PYY) and pancreatic polypeptide (PP), are suggested to regulate many physiological processes including food intake in birds. However, our knowledge regarding this avian NPY system remains rather limited. Here, we examined the tissue expression of NPY, PYY and PP and the gene structure, expression and signaling of three NPY receptors (cY1, cY4 and cY6) in chickens. The results showed that 1) NPY is widely expressed in chicken tissues with abundance noted in the hypothalamus via quantitative real-time PCR, whereas PYY is highly expressed in the pancreas, gastrointestinal tract and various brain regions, and PP is expressed almost exclusively in the pancreas; 2) cY1, cY4 and cY6 contain novel non-coding exon(s) at their 5'-UTR; 3) The wide tissue distribution of cY1 and cY4 and cY6 were detected in chickens by quantitative real-time PCR and their expression is controlled by the promoter near exon 1, which displays strong promoter activity in DF-1 cells as demonstrated by Dual-luciferase reporter assay; 4) Monitored by luciferase reporter assays, activation of cY1 and cY4 expressed in HEK293 cells by chicken NPY1-36, PYY1-37, and PP1-36 treatment inhibits cAMP/PKA and activates MAPK/ERK signaling pathways, while cY6-expressing cells show little response to peptide treatment, indicating that cY1 and cY4, and not cY6, can transmit signals in vitro. Taken together, our study offers novel information about the expression and functionality of cY1, cY4, cY6 and their ligands in birds, and helps to decipher their conserved roles in vertebrates.
Collapse
Affiliation(s)
- Shunyu Gao
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, PR China; College of Chemistry and Life Sciences, Chuxiong Normal University, Chuxiong 675000, PR China
| | - Jiannan Zhang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, PR China
| | - Chen He
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, PR China
| | - Fengyan Meng
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, PR China
| | - Guixian Bu
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, PR China
| | - Guoqiang Zhu
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, PR China
| | - Juan Li
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, PR China
| | - Yajun Wang
- Key Laboratory of Bio-resources and Eco-environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu 610064, PR China.
| |
Collapse
|
73
|
Gu XC, Zhang XB, Hu B, Zi Y, Li M. Neuropeptide Y accelerates post-fracture bone healing by promoting osteogenesis of mesenchymal stem cells. Neuropeptides 2016; 60:61-66. [PMID: 27720230 DOI: 10.1016/j.npep.2016.09.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2016] [Revised: 09/09/2016] [Accepted: 09/11/2016] [Indexed: 01/27/2023]
Abstract
Fracture repair is a complex yet well orchestrated regenerative process involving numerous signaling and cell types including osteoblasts. Here we showed that NPY, a neurotransmitter with regulatory functions in bone homeostasis, may contribute to the post-fracture bone healing in patients with traumatic brain injury-fracture combined injuries. Our results suggested NPY levels were increased in patients with the combined injuries, accomplished by arising of bone healing markers, such as ALP, OC, PICP and ICTP, than in those with simple fractures, and NPY have direct actions on MSCs to promote their osteogenic differentiation. Our results provided clinical evidences for NPY participating in the bone healing process in a nonhypothalamic manner, most probably by directly promoting osteogenesis of mesenchymal stem cells.
Collapse
Affiliation(s)
- Xiao-Chuan Gu
- Department of Orthopedics, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Xiao-Bin Zhang
- Department of Emergency Medicine, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China
| | - Bing Hu
- Department of Medical Oncology, Shanghai Minhang District Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Ying Zi
- Department of Emergency, 463rd Hospital of PLA, Shenyang 110042, China.
| | - Ming Li
- Department of Orthopedics, Changhai Hospital, Second Military Medical University, Shanghai, China.
| |
Collapse
|
74
|
Burkert K, Zellmann T, Meier R, Kaiser A, Stichel J, Meiler J, Mittapalli GK, Roberts E, Beck-Sickinger AG. A Deep Hydrophobic Binding Cavity is the Main Interaction for Different Y 2 R Antagonists. ChemMedChem 2016; 12:75-85. [PMID: 27874262 DOI: 10.1002/cmdc.201600433] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Revised: 10/28/2016] [Indexed: 12/29/2022]
Abstract
The neuropeptide Y2 receptor (Y2 R) is involved in various pathophysiological processes such as epilepsy, mood disorders, angiogenesis, and tumor growth. Therefore, the Y2 R is an interesting target for drug development. A detailed understanding of the binding pocket could facilitate the development of highly selective antagonists to study the role of Y2 R in vitro and in vivo. In this study, several residues crucial to the interaction of BIIE0246 and SF-11 derivatives with Y2 R were investigated by signal transduction assays. Using the experimental results as constraints, the antagonists were docked into a comparative structural model of the Y2 R. Despite differences in size and structure, all three antagonists display a similar binding site, including a deep hydrophobic cavity formed by transmembrane helices (TM) 4, 5, and 6, as well as a hydrophobic patch at the top of TM2 and 7. Additionally, we suggest that the antagonists block Q3.32 , a position that has been shown to be crucial for binding of the amidated C terminus of NPY and thus for receptor activation.
Collapse
Affiliation(s)
- Kerstin Burkert
- Faculty of Biosciences, Pharmacy and Psychology, Leipzig University, Institute of Biochemistry, Brüderstr. 34, 04103, Leipzig, Germany
| | - Tristan Zellmann
- Faculty of Biosciences, Pharmacy and Psychology, Leipzig University, Institute of Biochemistry, Brüderstr. 34, 04103, Leipzig, Germany
| | - René Meier
- Faculty of Biosciences, Pharmacy and Psychology, Leipzig University, Institute of Biochemistry, Brüderstr. 34, 04103, Leipzig, Germany
| | - Anette Kaiser
- Faculty of Biosciences, Pharmacy and Psychology, Leipzig University, Institute of Biochemistry, Brüderstr. 34, 04103, Leipzig, Germany
| | - Jan Stichel
- Faculty of Biosciences, Pharmacy and Psychology, Leipzig University, Institute of Biochemistry, Brüderstr. 34, 04103, Leipzig, Germany
| | - Jens Meiler
- Center for Structural Biology, Vanderbilt University, 465 21st Avenue South, Nashville, TN, 37203, USA
| | - Gopi K Mittapalli
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Edward Roberts
- Department of Chemistry, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA, 92037, USA
| | - Annette G Beck-Sickinger
- Faculty of Biosciences, Pharmacy and Psychology, Leipzig University, Institute of Biochemistry, Brüderstr. 34, 04103, Leipzig, Germany
| |
Collapse
|
75
|
Muroi Y, Ishii T. A novel neuropeptide Y neuronal pathway linking energy state and reproductive behavior. Neuropeptides 2016; 59:1-8. [PMID: 27659234 DOI: 10.1016/j.npep.2016.09.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 09/08/2016] [Accepted: 09/08/2016] [Indexed: 01/12/2023]
Abstract
Animals consume energy for reproduction, as well as survival. Excess or insufficient energy investment into reproduction, respectively, threatens the survival of parents or leads to the failure of reproduction. Management of energy consumption in reproduction is important, not only for the success of the process, but also for the survival of the parents. Reproductive behaviors, such as mating and parental behavior, are indispensable for achieving each event of reproduction including gametogamy, parturition, and lactation. Therefore, reproductive behavior is one of the important factors in managing energy consumption for reproduction. Orexigenic and anorexigenic molecules in the hypothalamus have been implicated in the regulation of reproductive functions. An orexigenic neuropeptide, neuropeptide Y (NPY), has been also implicated in the regulation of both reproduction and energy state of animals. In this review, we will first summarize the neuronal mechanism for regulating reproductive functions by orexigenic and anorexigenic molecules in the hypothalamus. Second, we will focus on the NPY neuronal pathways regulating reproductive behavior in the intra- and extra-hypothalamic brain areas. We will highlight the NPY neuronal pathway from the arcuate nucleus to the dorsal raphe nucleus as a novel extra-hypothalamic pathway for energy state-dependent regulation of reproductive behavior. Finally, we will propose a biological significance of the extra-hypothalamic NPY neuronal pathway, which plays an important role in the associative control of feeding and reproductive behaviors.
Collapse
Affiliation(s)
- Yoshikage Muroi
- Department of Basic Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan.
| | - Toshiaki Ishii
- Department of Basic Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| |
Collapse
|
76
|
Shipp SL, Cline MA, Gilbert ER. Recent advances in the understanding of how neuropeptide Y and α-melanocyte stimulating hormone function in adipose physiology. Adipocyte 2016; 5:333-350. [PMID: 27994947 DOI: 10.1080/21623945.2016.1208867] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 06/28/2016] [Accepted: 06/28/2016] [Indexed: 12/20/2022] Open
Abstract
Communication between the brain and the adipose tissue has been the focus of many studies in recent years, with the "brain-fat axis" identified as a system that orchestrates the assimilation and usage of energy to maintain body mass and adequate fat stores. It is now well-known that appetite-regulating peptides that were studied as neurotransmitters in the central nervous system can act both on the hypothalamus to regulate feeding behavior and also on the adipose tissue to modulate the storage of energy. Energy balance is thus partly controlled by factors that can alter both energy intake and storage/expenditure. Two such factors involved in these processes are neuropeptide Y (NPY) and α-melanocyte stimulating hormone (α-MSH). NPY, an orexigenic factor, is associated with promoting adipogenesis in both mammals and chickens, while α-MSH, an anorexigenic factor, stimulates lipolysis in rodents. There is also evidence of interaction between the 2 peptides. This review aims to summarize recent advances in the study of NPY and α-MSH regarding their role in adipose tissue physiology, with an emphasis on the cellular and molecular mechanisms. A greater understanding of the brain-fat axis and regulation of adiposity by bioactive peptides may provide insights on strategies to prevent or treat obesity and also enhance nutrient utilization efficiency in agriculturally-important species.
Collapse
|
77
|
Domin H, Święch D, Piergies N, Pięta E, Kim Y, Proniewicz E. Characterization of the surface geometry of acetyl-[Leu 28,31 ]-NPY(24-36), a selective Y 2 receptor agonist, onto the Ag and Au surfaces. VIBRATIONAL SPECTROSCOPY 2016; 85:1-6. [DOI: 10.1016/j.vibspec.2016.03.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
|
78
|
Borrow AP, Stranahan AM, Suchecki D, Yunes R. Neuroendocrine Regulation of Anxiety: Beyond the Hypothalamic-Pituitary-Adrenal Axis. J Neuroendocrinol 2016; 28. [PMID: 27318180 DOI: 10.1111/jne.12403] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 05/20/2016] [Accepted: 06/17/2016] [Indexed: 02/06/2023]
Abstract
The central nervous system regulates and responds to endocrine signals, and this reciprocal relationship determines emotional processing and behavioural anxiety. Although the hypothalamic-pituitary-adrenal (HPA) axis remains the best-characterised system for this relationship, other steroid and peptide hormones are increasingly recognised for their effects on anxiety-like behaviour and reward. The present review examines recent developments related to the role of a number of different hormones in anxiety, including pregnane neurosteroids, gut peptides, neuropeptides and hormonal signals derived from fatty acids. Findings from both basic and clinical studies suggest that these alternative systems may complement or occlude stress-induced changes in anxiety and anxiety-like behaviour. By broadening the scope of mechanisms for depression and anxiety, it may be possible to develop novel strategies to attenuate stress-related psychiatric conditions. The targets for these potential therapies, as discussed in this review, encompass multiple circuits and systems, including those outside of the HPA axis.
Collapse
Affiliation(s)
- A P Borrow
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - A M Stranahan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Georgia Regents University, Augusta, GA, USA
| | - D Suchecki
- Department of Psychobiology, Universidade Federal de São Paulo, São Paulo, Brazil
| | - R Yunes
- Instituto de Investigaciones Biomédicas, Facultad de Ciencias de la Salud, Universidad de Mendoza, Mendoza, Argentina
- Área de Farmacología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| |
Collapse
|
79
|
Cabral A, Portiansky E, Sánchez-Jaramillo E, Zigman JM, Perello M. Ghrelin activates hypophysiotropic corticotropin-releasing factor neurons independently of the arcuate nucleus. Psychoneuroendocrinology 2016; 67:27-39. [PMID: 26874559 PMCID: PMC4808343 DOI: 10.1016/j.psyneuen.2016.01.027] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 01/28/2016] [Accepted: 01/29/2016] [Indexed: 10/22/2022]
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.
Collapse
Affiliation(s)
| | | | | | | | - Mario Perello
- Laboratory of Neurophysiology, Multidisciplinary Institute of Cell Biology [IMBICE-Argentine Research Council (CONICET) and Scientific Research Commission, Province of Buenos Aires (CIC-PBA)], La Plata, Buenos Aires, Argentina.
| |
Collapse
|
80
|
Aerts E, Beckers S, Zegers D, Van Hoorenbeeck K, Massa G, Verrijken A, Verhulst SL, Van Gaal LF, Van Hul W. CNV analysis and mutation screening indicate an important role for the NPY4R gene in human obesity. Obesity (Silver Spring) 2016; 24:970-6. [PMID: 26921218 DOI: 10.1002/oby.21435] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/26/2015] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Genome-wide copy number variation (CNV) analyses have associated the 10q11.22 CNV with obesity. As the NPY4R gene is the most interesting candidate gene in this region, it was hypothesized that both genetic and structural variation in NPY4R might be implicated in the pathogenesis of obesity. METHODS In the first part of this study, 326 children and adolescents with obesity and 298 healthy lean individuals were screened for CNV in the NPY4R-containing chr.10q11.22 region. In the second part of this study, a mutation screen for variants in the NPY4R coding region was performed in 356 children and adolescents with obesity and 337 healthy lean adults. RESULTS Our CNV analysis demonstrated a significantly higher frequency of NPY4R containing 10q11.22 CNV loss in the patient population (P = 0.0003), while CNV gain in this region was more prevalent in the control population (P = 0.031). Mutation analysis resulted in the identification of 15 rare non-synonymous heterozygous variants. For two variants that could only be identified in the patient population, receptor dysfunction and thus a pathogenic effect were demonstrated. CONCLUSIONS In conclusion, these data support an essential role for genetic and structural variation within the NPY4R gene in the pathogenesis of obesity.
Collapse
Affiliation(s)
- Evi Aerts
- Centre of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Sigri Beckers
- Centre of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Doreen Zegers
- Centre of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | | | - Guy Massa
- Department of Pediatrics, Jessa Hospital, Hasselt, Belgium
| | - An Verrijken
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Antwerp, Belgium
| | - Stijn L Verhulst
- Department of Pediatrics, Antwerp University Hospital, Antwerp, Belgium
| | - Luc F Van Gaal
- Department of Endocrinology, Diabetology and Metabolism, Antwerp University Hospital, Antwerp, Belgium
| | - Wim Van Hul
- Centre of Medical Genetics, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
81
|
Horsnell H, Baldock PA. Osteoblastic Actions of the Neuropeptide Y System to Regulate Bone and Energy Homeostasis. Curr Osteoporos Rep 2016; 14:26-31. [PMID: 26872458 DOI: 10.1007/s11914-016-0300-9] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Neural pathways are now a well-appreciated factor in the regulatory milieu controlling the maintenance of bone mass. A number of neural pathways from the brain to bone have been identified. These pathways often involve elements of the energy homeostatic apparatus, indicating links between the regulation of bone metabolism and energy balance. Neuropeptide Y is one such factor that co-regulates these two processes. Initial studies outlined the skeletal actions of NPY from within the brain and the interactions with energy homeostatic processes. However, in recent years, an appreciation for the actions of NPY within bone cells has expanded. Cells of the osteoblastic lineage express both NPY ligand and a cognate receptor NPY, Y1R. Murine studies have demonstrated that both ligand and receptor actively control bone mass and osteoblast activity and interact with mechanical signals to integrate with the local loading environment. Local NPY signalling regulates osteoprogenitor production and differentiation, to cover the entire osteoblastic lineage. In addition, several recent studies have demonstrated extra-skeletal actions of osteoblastic NPY signalling, to regulate energy expenditure and with it adiposity, and in a separate study, to control release of a factor-controlling beta cell mass and insulin production/release and with it glucose tolerance. Thus, osteoblastic neuropeptide production and signalling illustrates the rapidly widening sphere of influence of skeletal tissue, and suggests a far more complex and interconnected physiology then is currently appreciated.
Collapse
Affiliation(s)
- Harry Horsnell
- Osteoporosis and Bone Biology Division, Garvan Institute of Medical Research, St Vincent's Hospital, 390 Victoria St, Darlinghurst, Sydney, NSW, 2010, Australia
- Department of Biology and Biochemistry, Bath University, Claverton Down Rd, Bath, North East Somerset, BA2 7AY, UK
| | - Paul A Baldock
- Osteoporosis and Bone Biology Division, Garvan Institute of Medical Research, St Vincent's Hospital, 390 Victoria St, Darlinghurst, Sydney, NSW, 2010, Australia.
- School of Medicine, The University of Notre Dame Australia, 160 Oxford St, Darlinghurst, Sydney, NSW, 2010, Australia.
- Faculty of Medicine, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
82
|
Kaplowitz ET, Savenkova M, Karatsoreos IN, Romeo RD. Somatic and Neuroendocrine Changes in Response to Chronic Corticosterone Exposure During Adolescence in Male and Female Rats. J Neuroendocrinol 2016; 28:12336. [PMID: 26568535 DOI: 10.1111/jne.12336] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 10/08/2015] [Accepted: 11/06/2015] [Indexed: 01/18/2023]
Abstract
Prolonged stress and repeated activation of the hypothalamic-pituitary-adrenal axis can result in many sex-dependent behavioural and metabolic changes in rats, including alterations in feeding behaviour and reduced body weight. In adults, these effects of stress can be mimicked by corticosterone, a major output of the hypothalamic-pituitary-adrenal axis, and recapitulate the stress-induced sex difference, such that corticosterone-treated males show greater weight loss than females. Similar to adults, chronic stress during adolescence leads to reduced weight gain, particularly in males. However, it is currently unknown whether corticosterone mediates this somatic change and whether additional measures of neuroendocrine function are affected by chronic corticosterone exposure during adolescence in a sex-dependent manner. Therefore, we examined the effects of non-invasively administered corticosterone (150 or 300 μg/ml) in the drinking water of male and female rats throughout adolescent development (30-58 days of age). We found that adolescent animals exposed to chronic corticosterone gain significantly less weight than controls, which may be partly mediated by the effects of corticosterone on food consumption, fluid intake and gonadal hormone function. Our data further show that, despite similar circulating corticosterone levels, males demonstrate a greater sensitivity to these changes than females. We also found that Npy1 and Npy5 receptor mRNA expression, genes implicated in appetite regulation, was significantly reduced in the ventral medial hypothalamus of corticosterone-treated males and females compared to controls. Finally, parameters of gonadal function, such as plasma sex steroid concentrations and weight of reproductive tissues, were reduced by adolescent corticosterone treatment, although only in males. The data obtained in the present study indicate that chronic corticosterone exposure throughout adolescent development results in significant and sex-dependent somatic and neuroendocrine changes, and the results also provide an experimental framework for further investigating the impact of corticosterone on metabolic and neuroendocrine function during adolescence.
Collapse
Affiliation(s)
- E T Kaplowitz
- Department of Psychology and Neuroscience and Behavior Program, Barnard College of Columbia University, New York, NY, USA
| | - M Savenkova
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - I N Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, Pullman, WA, USA
| | - R D Romeo
- Department of Psychology and Neuroscience and Behavior Program, Barnard College of Columbia University, New York, NY, USA
| |
Collapse
|
83
|
Abstract
Stress is defined as an adverse condition that disturbs the homeostasis of the body and activates adaptation responses. Among the many pathways and mediators involved, neuropeptide Y (NPY) stands out due to its unique stress-relieving, anxiolytic and neuroprotective properties. Stress exposure alters the biosynthesis of NPY in distinct brain regions, the magnitude and direction of this effect varying with the duration and type of stress. NPY is expressed in particular neurons of the brainstem, hypothalamus and limbic system, which explains why NPY has an impact on stress-related changes in emotional-affective behaviour and feeding as well as on stress coping. The biological actions of NPY in mammals are mediated by the Y1, Y2, Y4 and Y5 receptors, Y1 receptor stimulation being anxiolytic whereas Y2 receptor activation is anxiogenic. Emerging evidence attributes NPY a role in stress resilience, the ability to cope with stress. Thus there is a negative correlation between stress-induced behavioural disruption and cerebral NPY expression in animal models of post-traumatic stress disorder. Exogenous NPY prevents the negative consequences of stress, and polymorphisms of the NPY gene are predictive of impaired stress processing and increased risk of neuropsychiatric diseases. Stress is also a factor contributing to, and resulting from, neurodegenerative diseases such as Alzheimer's, Parkinson's and Huntington's disease, in which NPY appears to play an important neuroprotective role. This review summarizes the evidence for an implication of NPY in stress-related and neurodegenerative pathologies and addresses the cerebral NPY system as a therapeutic target.
Collapse
Affiliation(s)
- Florian Reichmann
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, A-8010 Graz, Austria.
| | - Peter Holzer
- Research Unit of Translational Neurogastroenterology, Institute of Experimental and Clinical Pharmacology, Medical University of Graz, Universitätsplatz 4, A-8010 Graz, Austria
| |
Collapse
|
84
|
Khandekar N, Berning BA, Sainsbury A, Lin S. The role of pancreatic polypeptide in the regulation of energy homeostasis. Mol Cell Endocrinol 2015; 418 Pt 1:33-41. [PMID: 26123585 DOI: 10.1016/j.mce.2015.06.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 03/16/2015] [Accepted: 06/03/2015] [Indexed: 12/13/2022]
Abstract
Imbalances in normal regulation of food intake can cause obesity and related disorders. Inadequate therapies for such disorders necessitate better understanding of mechanisms that regulate energy homeostasis. Pancreatic polypeptide (PP), a robust anorexigenic hormone, effectively modulates food intake and energy homeostasis, thus potentially aiding anti-obesity therapeutics. Intra-gastric and intra-intestinal infusion of nutrients stimulate PP secretion from the gastrointestinal tract, leading to vagal stimulation that mediates complex actions via the neuropeptide Y4 receptor in arcuate nucleus of the hypothalamus, subsequently activating key hypothalamic nuclei and dorsal vagal complex of the brainstem to influence energy homeostasis and body composition. Novel studies indicate affinity of PP for the relatively underexplored neuropeptide y6 receptor, mediating actions via the suprachiasmatic nucleus and pathways involving vasoactive intestinal polypeptide and insulin like growth factor 1. This review highlights detailed mechanisms by which PP mediates its actions on energy balance through various areas in the brain.
Collapse
Affiliation(s)
- Neeta Khandekar
- Neurological Diseases Division, Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | - Britt A Berning
- Neurological Diseases Division, Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia
| | - Amanda Sainsbury
- The Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, Sydney Medical School, The University of Sydney, NSW 2006, Australia
| | - Shu Lin
- Neurological Diseases Division, Research Program, Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, Sydney, NSW 2010, Australia; School of Medical Sciences, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
85
|
Xu B, Lagman D, Sundström G, Larhammar D. Neuropeptide Y family receptors Y1 and Y2 from sea lamprey, Petromyzon marinus. Gen Comp Endocrinol 2015; 222:106-15. [PMID: 26255155 DOI: 10.1016/j.ygcen.2015.08.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Revised: 07/31/2015] [Accepted: 08/05/2015] [Indexed: 12/15/2022]
Abstract
The vertebrate gene family for neuropeptide Y (NPY) receptors expanded by duplication of the chromosome carrying the ancestral Y1-Y2-Y5 gene triplet. After loss of some duplicates, the ancestral jawed vertebrate had seven receptor subtypes forming the Y1 (including Y1, Y4, Y6, Y8), Y2 (including Y2, Y7) and Y5 (only Y5) subfamilies. Lampreys are considered to have experienced the same chromosome duplications as gnathostomes and should also be expected to have multiple receptor genes. However, previously only a Y4-like and a Y5 receptor have been cloned and characterized. Here we report the cloning and characterization of two additional receptors from the sea lamprey Petromyzon marinus. Sequence phylogeny alone could not with certainty assign their identity, but based on synteny comparisons of P. marinus and the Arctic lamprey, Lethenteron camtschaticum, with jawed vertebrates, the two receptors most likely are Y1 and Y2. Both receptors were expressed in human HEK293 cells and inositol phosphate assays were performed to determine the response to the three native lamprey peptides NPY, PYY and PMY. The three peptides have similar potencies in the nanomolar range for Y1. No obvious response to the three peptides was detected for Y2. Synteny analysis supports identification of the previously cloned receptor as Y4. No additional NPY receptor genes could be identified in the presently available lamprey genome assemblies. Thus, four NPY-family receptors have been identified in lampreys, orthologs of the same subtypes as in humans (Y1, Y2, Y4 and Y5), whereas many other vertebrate lineages have retained additional ancestral subtypes.
Collapse
Affiliation(s)
- Bo Xu
- Department of Neuroscience, Science for Life Laboratory, Uppsala University, Box 593, SE-75124 Uppsala, Sweden
| | - David Lagman
- Department of Neuroscience, Science for Life Laboratory, Uppsala University, Box 593, SE-75124 Uppsala, Sweden
| | - Görel Sundström
- Department of Neuroscience, Science for Life Laboratory, Uppsala University, Box 593, SE-75124 Uppsala, Sweden
| | - Dan Larhammar
- Department of Neuroscience, Science for Life Laboratory, Uppsala University, Box 593, SE-75124 Uppsala, Sweden.
| |
Collapse
|
86
|
Zieba J, Low JK, Purtell L, Qi Y, Campbell L, Herzog H, Karl T. Behavioural characteristics of the Prader-Willi syndrome related biallelic Snord116 mouse model. Neuropeptides 2015; 53:71-7. [PMID: 26259850 DOI: 10.1016/j.npep.2015.06.009] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 05/04/2015] [Accepted: 06/30/2015] [Indexed: 10/23/2022]
Abstract
Prader-Willi syndrome (PWS) is the predominant genetic cause of obesity in humans and is associated with several behavioural phenotypes such as altered motoric function, reduced activity, and learning disabilities. It can include mood instability and, in some cases, psychotic episodes. Recently, the Snord116 gene has been associated with the development of PWS, however, it's contribution to the behavioural aspects of the disease are unknown. Here we show that male and female mice lacking Snord116 on both alleles exhibit normal motor behaviours and exploration but do display task-dependent alterations to locomotion and anxiety-related behaviours. Sociability is well developed in Snord116 deficient mice as are social recognition memory, spatial working memory, and fear-associated behaviours. No sex-specific effects were found. In conclusion, the biallelic Snord116 deficiency mouse model exhibits particular endophenotypes with some relevance to PWS, suggesting partial face validity for the syndrome.
Collapse
Affiliation(s)
- Jerzy Zieba
- Neuroscience Research Australia, Randwick, NSW 2031, Australia; Schizophrenia Research Institute, Darlinghurst, NSW 2010, Australia
| | - Jac Kee Low
- Neuroscience Research Australia, Randwick, NSW 2031, Australia
| | - Louise Purtell
- Neuroscience Division, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Yue Qi
- Neuroscience Division, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Lesley Campbell
- Neuroscience Division, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, Darlinghurst, NSW 2010, Australia
| | - Tim Karl
- Neuroscience Research Australia, Randwick, NSW 2031, Australia; Schizophrenia Research Institute, Darlinghurst, NSW 2010, Australia; School of Medical Sciences, University of New South Wales, NSW 2052, Australia.
| |
Collapse
|
87
|
Celik O, Aydin S, Celik N, Yilmaz M. Peptides: Basic determinants of reproductive functions. Peptides 2015; 72:34-43. [PMID: 26074346 DOI: 10.1016/j.peptides.2015.05.016] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Revised: 05/28/2015] [Accepted: 05/29/2015] [Indexed: 12/22/2022]
Abstract
Mammalian reproduction is a costly process in terms of energy consumption. The critical information regarding metabolic status is signaled to the hypothalamus mainly through peripheral peptides from the adipose tissue and gastrointestinal tract. Changes in energy stores produce fluctuations in leptin, insulin, ghrelin and glucose signals that feedback mainly to the hypothalamus to regulate metabolism and fertility. In near future, possible effects of the nutritional status on GnRH regulation can be evaluated by measuring serum or tissue levels of leptin and ghrelin in patiens suffering from infertility. The fact that leptin and ghrelin are antagonistic in their effects on GnRH neurons, their respective agonistic and antagonistic roles make them ideal candidates to use instead of GnRH agonist and antagonist. Similarly, kisspeptin expressing neurons are likely to mediate the well-established link between energy balance and reproductive functions. Exogenous kisspeptin can be used for physiological ovarian hyperstimulation for in-vitro fertilization. Moreover, kisspeptin antagonist therapy can be used for the treatment of postmenapousal women, precocious puberty, PCOS, endometriosis and uterine fibroids. In this review, we will analyze the central mechanisms involved in the integration of metabolic information and their contribution to the control of the reproductive function. Particular attention will be paid to summarize the participation of leptin, kisspeptin, ghrelin, NPY, orexin, urocortin, VIP, insulin, galanin, galanin like peptide, oxytocin, agouti gene-related peptide, and POMC neurons in this process and their possible interactions to contribute to the metabolic control of reproduction.
Collapse
Affiliation(s)
- Onder Celik
- Private Clinic, Department of Obstetrics and Gynecology, İzmir, Turkey
| | - Suleyman Aydin
- Firat University, School of Medicine, Department of Medical Biochemistry (Firat Hormones Research Group), 23119 Elazig, Turkey.
| | - Nilufer Celik
- Behcet Uz Children's Hospital, Department of Biochemistry, İzmir, Turkey
| | - Musa Yilmaz
- Firat University, School of Medicine, Department of Medical Biochemistry (Firat Hormones Research Group), 23119 Elazig, Turkey
| |
Collapse
|
88
|
Fukuoka T, Noguchi K. A potential anti-allodynic mechanism of GDNF following L5 spinal nerve ligation; Mitigation of NPY up-regulation in the touch sense pathway. Neuroscience 2015. [PMID: 26215916 DOI: 10.1016/j.neuroscience.2015.07.059] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Intrathecal delivery of glial cell line-derived neurotrophic factor (GDNF) reverses mechanical allodynia after 5th lumbar (L5) spinal nerve ligation (SNL). However, the molecular mechanism behind this process is not fully understood. Following sciatic nerve injury, primary afferent neurons in the injured dorsal root ganglion (DRG) begin to express neuropeptide Y (NPY) that is absent in normal DRG. The aim of the current study was to determine the relationship of this de novo expression of NPY and the anti-allodynic effect of GDNF. Following L5 SNL, 73% of neurons began to express NPY mRNA in the ipsilateral L5 DRG and robust NPY-immunoreactive fibers appeared in the ipsilateral GN where the touch-sense mediating A-fiber primary afferents from the hindpaw terminate. Seven-daylong intrathecal infusion of GDNF at the L5 DRG level, starting on day three when mechanical allodynia had fully developed, reversed once-established these changes. The GN neurons normally expressed NPY Y1 receptor, but not Y2, Y4, or Y5 receptors, and L5 SNL did not change the expression pattern. Bolus intracisternal injection of BIBP3226, a Y1 receptor antagonist, dose-dependently reversed mechanical allodynia. We demonstrated that GDNF reversed once-established mechanical allodynia as well as NPY induction in the touch-sense processing pathway. NPY could facilitate touch-sense processing by Y1 receptor in the gracile nucleus after peripheral nerve injury. GDNF may exert anti-allodynic effects through mitigation of this NPY up-regulation. The effectiveness of delayed treatment further indicates the therapeutic potential of GDNF on neuropathic pain.
Collapse
Affiliation(s)
- T Fukuoka
- Department of Anatomy & Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan; Pain Mechanism Research Group, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan.
| | - K Noguchi
- Department of Anatomy & Neuroscience, Hyogo College of Medicine, Nishinomiya, Hyogo, Japan
| |
Collapse
|
89
|
Al-Barazanji K, McNulty J, Binz J, Generaux C, Benson W, Young A, Chen L. Synergistic Effects of a GPR119 Agonist with Metformin on Weight Loss in Diet-Induced Obese Mice. J Pharmacol Exp Ther 2015; 353:496-504. [PMID: 25770135 DOI: 10.1124/jpet.115.222828] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/12/2015] [Indexed: 12/29/2022] Open
Abstract
G protein-coupled receptor 119 (GPR119) is a G protein-coupled receptor expressed predominantly in pancreatic β-cells and gastrointestinal enteroendocrine cells. Metformin is a first-line treatment of type 2 diabetes, with minimal weight loss in humans. In this study, we investigated the effects of GSK2041706 [2-([(1S)-1-(1-[3-(1-methylethyl)-1,2,4-oxadiazol-5-yl]-4-piperidinyl)ethyl]oxy)-5-[4-(methylsulfonyl)phenyl]pyrazine], a GPR119 agonist, and metformin as monotherapy or in combination on body weight in a diet-induced obese (DIO) mouse model. Relative to vehicle controls, 14-day treatment with GSK2041706 (30 mg/kg b.i.d.) or metformin at 30 and 100 mg/kg b.i.d. alone caused a 7.4%, 3.5%, and 4.4% (all P < 0.05) weight loss, respectively. The combination of GSK2041706 with metformin at 30 or 100 mg/kg resulted in a 9.5% and 16.7% weight loss, respectively. The combination of GSK2041706 and metformin at 100 mg/kg caused a significantly greater weight loss than the projected additive weight loss of 11.8%. This body weight effect was predominantly due to a loss of fat. Cumulative food intake was reduced by 17.1% with GSK2041706 alone and 6.6% and 8.7% with metformin at 30 and 100 mg/kg, respectively. The combination of GSK2041706 with metformin caused greater reductions in cumulative food intake (22.2% at 30 mg/kg and 37.5% at 100 mg/kg) and higher fed plasma glucagon-like peptide 1 and peptide tyrosine tyrosine levels and decreased plasma insulin and glucose-dependent insulinotropic polypeptide levels compared with their monotherapy groups. In addition, we characterized the effect of GSK2041706 and metformin as monotherapy or in combination on neuronal activation in the appetite regulating centers in fasted DIO mice. In conclusion, our data demonstrate the beneficial effects of combining a GPR119 agonist with metformin in the regulation of body weight in DIO mice.
Collapse
Affiliation(s)
- Kamal Al-Barazanji
- Enteroendocrine Drug Performance Unit, GlaxoSmithKline Research and Development, Research Triangle Park, North Carolina
| | - Judi McNulty
- Enteroendocrine Drug Performance Unit, GlaxoSmithKline Research and Development, Research Triangle Park, North Carolina
| | - Jane Binz
- Enteroendocrine Drug Performance Unit, GlaxoSmithKline Research and Development, Research Triangle Park, North Carolina
| | - Claudia Generaux
- Enteroendocrine Drug Performance Unit, GlaxoSmithKline Research and Development, Research Triangle Park, North Carolina
| | - William Benson
- Enteroendocrine Drug Performance Unit, GlaxoSmithKline Research and Development, Research Triangle Park, North Carolina
| | - Andrew Young
- Enteroendocrine Drug Performance Unit, GlaxoSmithKline Research and Development, Research Triangle Park, North Carolina
| | - Lihong Chen
- Enteroendocrine Drug Performance Unit, GlaxoSmithKline Research and Development, Research Triangle Park, North Carolina
| |
Collapse
|
90
|
Huang Y, Thathiah A. Regulation of neuronal communication by G protein-coupled receptors. FEBS Lett 2015; 589:1607-19. [PMID: 25980603 DOI: 10.1016/j.febslet.2015.05.007] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 05/05/2015] [Accepted: 05/05/2015] [Indexed: 02/06/2023]
Abstract
Neuronal communication plays an essential role in the propagation of information in the brain and requires a precisely orchestrated connectivity between neurons. Synaptic transmission is the mechanism through which neurons communicate with each other. It is a strictly regulated process which involves membrane depolarization, the cellular exocytosis machinery, neurotransmitter release from synaptic vesicles into the synaptic cleft, and the interaction between ion channels, G protein-coupled receptors (GPCRs), and downstream effector molecules. The focus of this review is to explore the role of GPCRs and G protein-signaling in neurotransmission, to highlight the function of GPCRs, which are localized in both presynaptic and postsynaptic membrane terminals, in regulation of intrasynaptic and intersynaptic communication, and to discuss the involvement of astrocytic GPCRs in the regulation of neuronal communication.
Collapse
Affiliation(s)
- Yunhong Huang
- VIB Center for the Biology of Disease, Leuven, Belgium; Center for Human Genetics (CME) and Leuven Institute for Neurodegenerative Diseases (LIND), University of Leuven (KUL), Leuven, Belgium.
| | - Amantha Thathiah
- VIB Center for the Biology of Disease, Leuven, Belgium; Center for Human Genetics (CME) and Leuven Institute for Neurodegenerative Diseases (LIND), University of Leuven (KUL), Leuven, Belgium.
| |
Collapse
|
91
|
Henry KE, Elfers CT, Burke RM, Chepurny OG, Holz GG, Blevins JE, Roth CL, Doyle RP. Vitamin B12 conjugation of peptide-YY(3-36) decreases food intake compared to native peptide-YY(3-36) upon subcutaneous administration in male rats. Endocrinology 2015; 156:1739-49. [PMID: 25658456 PMCID: PMC4398759 DOI: 10.1210/en.2014-1825] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Challenges to peptide-based therapies include rapid clearance, ready degradation by hydrolysis/proteolysis, and poor intestinal uptake and/or a need for blood brain barrier transport. This work evaluates the efficacy of conjugation of vitamin B12 (B12) on sc administered peptide tyrosine tyrosine (PYY)(3-36) function. In the current experiments, a B12-PYY(3-36) conjugate was tested against native PYY(3-36), and an inactive conjugate B12-PYYC36 (null control) in vitro and in vivo. In vitro experiments demonstrated similar agonism for the neuropeptide Y2 receptor by the B12-PYY(3-36) conjugate (EC50 26.5 nM) compared with native PYY(3-36) (EC50 16.0 nM), with the null control having an EC50 of 1.8 μM. In vivo experiments were performed in young adult male Sprague Dawley rats (9 wk). Daily treatments were delivered sc in five 1-hour pulses, each pulse delivering 5-10 nmol/kg, by implanted microinfusion pumps. Increases in hindbrain Fos expression were comparable 90 minutes after B12-PYY(3-36) or PYY3-36 injection relative to saline or B12-PYYC36. Food intake was reduced during a 5-day treatment for both B12-PYY(3-36)- (24%, P = .001) and PYY(3-36)-(13%, P = .008) treated groups relative to baseline. In addition, reduction of food intake after the three dark cycle treatment pulses was more consistent with B12-PYY(3-36) treatment (-26%, -29%, -27%) compared with the PYY(3-36) treatment (-3%, -21%, -16%), and B12-PYY(3-36) generated a significantly longer inhibition of food intake vs. PYY(3-36) treatment after the first two pulses (P = .041 and P = .036, respectively). These findings demonstrate a stronger, more consistent, and longer inhibition of food intake after the pulses of B12-PYY(3-36) conjugate compared with the native PYY(3-36).
Collapse
Affiliation(s)
- Kelly E Henry
- Department of Chemistry (K.E.H., R.M.B., R.P.D.), Center for Science and Technology, Syracuse University, Syracuse, New York 13244; Center for Integrative Brain Research (C.T.E., C.L.R.), Division of Endocrinology, Seattle Children's Research Institute, Seattle, Washington 98101; Departments of Medicine (O.G.C., G.G.H., R.P.D.) and Pharmacology (G.G.H.), State University of New York, Upstate Medical University, Syracuse, New York 13210; Research and Development Service (J.E.B.), Veterans Affairs Puget Sound Health Care System, Seattle, Washington 98108; Department of Medicine (J.E.B.), Division of Metabolism, Endocrinology, and Nutrition, University of Washington, Seattle, Washington 98195; and Division of Endocrinology (C.L.R.), Department of Pediatrics, University of Washington, Seattle, Washington 98105
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Mörl K, Beck-Sickinger AG. Intracellular Trafficking of Neuropeptide Y Receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2015; 132:73-96. [PMID: 26055055 DOI: 10.1016/bs.pmbts.2015.02.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The multireceptor multiligand system of neuropeptide Y receptors and their ligands is involved in the regulation of a multitude of physiological and pathophysiological processes. Specific expression patterns, ligand-binding modes, and signaling properties contribute to the complex network regulating distinct cellular responses. Intracellular trafficking processes are important key steps that are regulated in context with accessory proteins. These proteins exert their influence by interacting directly or indirectly with the receptors, causing modification of the receptors, or operating as scaffolds for the assembly of larger signaling complexes. On the intracellular receptor faces, sequence-specific motifs have been identified that play an important role in this process. Interestingly, it is also possible to influence the receptor internalization by modification of the peptide ligand.
Collapse
Affiliation(s)
- Karin Mörl
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Universität Leipzig, Leipzig, Germany.
| | - Annette G Beck-Sickinger
- Faculty of Biosciences, Pharmacy and Psychology, Institute of Biochemistry, Universität Leipzig, Leipzig, Germany
| |
Collapse
|
93
|
Geloso MC, Corvino V, Di Maria V, Marchese E, Michetti F. Cellular targets for neuropeptide Y-mediated control of adult neurogenesis. Front Cell Neurosci 2015; 9:85. [PMID: 25852477 PMCID: PMC4360818 DOI: 10.3389/fncel.2015.00085] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 02/23/2015] [Indexed: 12/14/2022] Open
Abstract
Neuropeptides are emerging as key regulators of stem cell niche activities in health and disease, both inside and outside the central nervous system (CNS). Among them, neuropeptide Y (NPY), one of the most abundant neuropeptides both in the nervous system and in non-neural districts, has become the focus of much attention for its involvement in a wide range of physiological and pathological conditions, including the modulation of different stem cell activities. In particular, a pro-neurogenic role of NPY has been evidenced in the neurogenic niche, where a direct effect on neural progenitors has been demonstrated, while different cellular types, including astrocytes, microglia and endothelial cells, also appear to be responsive to the peptide. The marked modulation of the NPY system during several pathological conditions that affect neurogenesis, including stress, seizures and neurodegeneration, further highlights the relevance of this peptide in the regulation of adult neurogenesis. In view of the considerable interest in understanding the mechanisms controlling neural cell fate, this review aims to summarize and discuss current data on NPY signaling in the different cellular components of the neurogenic niche in order to elucidate the complexity of the mechanisms underlying the modulatory properties of this peptide.
Collapse
Affiliation(s)
- Maria Concetta Geloso
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Valentina Corvino
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Valentina Di Maria
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Elisa Marchese
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| | - Fabrizio Michetti
- Institute of Anatomy and Cell Biology, Università Cattolica del Sacro Cuore Rome, Italy
| |
Collapse
|
94
|
Fernando HA, Zibellini J, Hsu MS, Seimon RV, Nguyen AD, Sainsbury A. The neuropeptide Y-ergic system: potential therapeutic target against bone loss with obesity treatments. Expert Rev Endocrinol Metab 2015; 10:177-191. [PMID: 30293515 DOI: 10.1586/17446651.2015.1001741] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Obesity is no longer considered to provide protection against osteoporosis. Moreover, treatments for obesity are now suspected of reducing bone mass. With the escalating incidence of obesity, combined with increases in the frequency, duration and intensity of interventions used to combat it, we face a potential increase in health burden due to osteoporotic fractures. The neuropeptide Y-ergic system offers a potential target for the prevention and anabolic treatment of bone loss in obesity, due to its dual role in the regulation of energy homeostasis and bone mass. Although the strongest stimulation of bone mass by this system appears to occur via indirect hypothalamic pathways involving Y2 receptors (one of the five types of receptors for neuropeptide Y), Y1 receptors on osteoblasts (bone-forming cells) induce direct effects to enhance bone mass. This latter pathway may offer a suitable target for anti-osteoporotic treatment while also minimizing the risk of adverse side effects.
Collapse
Affiliation(s)
- Hamish A Fernando
- a 1 The Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, Sydney Medical School, The University of Sydney, Camperdown NSW 2006, Australia
| | - Jessica Zibellini
- a 1 The Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, Sydney Medical School, The University of Sydney, Camperdown NSW 2006, Australia
| | - Michelle Sh Hsu
- a 1 The Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, Sydney Medical School, The University of Sydney, Camperdown NSW 2006, Australia
| | - Radhika V Seimon
- a 1 The Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, Sydney Medical School, The University of Sydney, Camperdown NSW 2006, Australia
| | - Amy D Nguyen
- b 2 Neuroscience Program, Garvan Institute of Medical Research, University of New South Wales, Darlinghurst, Australia
| | - Amanda Sainsbury
- a 1 The Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, Sydney Medical School, The University of Sydney, Camperdown NSW 2006, Australia
| |
Collapse
|
95
|
Bari A, Dec A, Lee AW, Lee J, Song D, Dale E, Peterson J, Zorn S, Huang X, Campbell B, Robbins TW, West AR. Enhanced inhibitory control by neuropeptide Y Y5 receptor blockade in rats. Psychopharmacology (Berl) 2015; 232:959-73. [PMID: 25194952 DOI: 10.1007/s00213-014-3730-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Accepted: 08/24/2014] [Indexed: 12/25/2022]
Abstract
RATIONALE The neuropeptide Y (NPY) system acts in synergy with the classic neurotransmitters to regulate a large variety of functions including autonomic, affective, and cognitive processes. Research on the effects of NPY in the central nervous system has focused on food intake control and affective processes, but growing evidence of NPY involvement in attention-deficit/hyperactivity disorder (ADHD) and other psychiatric conditions motivated the present study. OBJECTIVES We tested the effects of the novel and highly selective NPY Y5 receptor antagonist Lu AE00654 on impulsivity and the underlying cortico-striatal circuitry in rats to further explore the possible involvement of the NPY system in pathologies characterized by inattention and impulsive behavior. RESULTS A low dose of Lu AE00654 (0.03 mg/kg) selectively facilitated response inhibition as measured by the stop-signal task, whereas no effects were found at higher doses (0.3 and 3 mg/kg). Systemic administration of Lu AE00654 also enhanced the inhibitory influence of the dorsal frontal cortex on neurons in the caudate-putamen, this fronto-striatal circuitry being implicated in the executive control of behavior. Finally, by locally injecting a Y5 agonist, we observed reciprocal activation between dorsal frontal cortex and caudate-putamen neurons. Importantly, the effects of the Y5 agonist were attenuated by pretreatment with Lu AE00654, confirming the presence of Y5 binding sites modulating functional interactions within frontal-subcortical circuits. CONCLUSIONS These results suggest that the NPY system modulates inhibitory neurotransmission in brain areas important for impulse control, and may be relevant for the treatment of pathologies such as ADHD and drug abuse.
Collapse
Affiliation(s)
- A Bari
- Behavioral and Clinical Neuroscience Institute and Department of Psychology, University of Cambridge, Cambridge, CB2 3EB, UK,
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Loh K, Herzog H, Shi YC. Regulation of energy homeostasis by the NPY system. Trends Endocrinol Metab 2015; 26:125-35. [PMID: 25662369 DOI: 10.1016/j.tem.2015.01.003] [Citation(s) in RCA: 206] [Impact Index Per Article: 20.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 01/11/2015] [Accepted: 01/12/2015] [Indexed: 01/01/2023]
Abstract
Obesity develops when energy intake exceeds energy expenditure over time. Numerous neurotransmitters, hormones, and factors have been implicated to coordinately control energy homeostasis, centrally and peripherally. However, the neuropeptide Y (NPY) system has emerged as the one with the most critical functions in this process. While NPY centrally promotes feeding and reduces energy expenditure, peptide YY (PYY) and pancreatic polypeptide (PP), the other family members, mediate satiety. Importantly, recent research has uncovered additional functions for these peptides that go beyond the simple feeding/satiety circuits and indicate a more extensive function in controlling energy homeostasis. In this review, we will discuss the actions of the NPY system in the regulation of energy balance, with a particular focus on energy expenditure.
Collapse
Affiliation(s)
- Kim Loh
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, 2010, Australia; Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia
| | - Herbert Herzog
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, 2010, Australia; Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia.
| | - Yan-Chuan Shi
- Neuroscience Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, 2010, Australia; Faculty of Medicine, UNSW Australia, Sydney, 2052, Australia.
| |
Collapse
|
97
|
Singewald N, Schmuckermair C, Whittle N, Holmes A, Ressler KJ. Pharmacology of cognitive enhancers for exposure-based therapy of fear, anxiety and trauma-related disorders. Pharmacol Ther 2014; 149:150-90. [PMID: 25550231 PMCID: PMC4380664 DOI: 10.1016/j.pharmthera.2014.12.004] [Citation(s) in RCA: 275] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2014] [Accepted: 12/24/2014] [Indexed: 12/20/2022]
Abstract
Pathological fear and anxiety are highly debilitating and, despite considerable advances in psychotherapy and pharmacotherapy they remain insufficiently treated in many patients with PTSD, phobias, panic and other anxiety disorders. Increasing preclinical and clinical evidence indicates that pharmacological treatments including cognitive enhancers, when given as adjuncts to psychotherapeutic approaches [cognitive behavioral therapy including extinction-based exposure therapy] enhance treatment efficacy, while using anxiolytics such as benzodiazepines as adjuncts can undermine long-term treatment success. The purpose of this review is to outline the literature showing how pharmacological interventions targeting neurotransmitter systems including serotonin, dopamine, noradrenaline, histamine, glutamate, GABA, cannabinoids, neuropeptides (oxytocin, neuropeptides Y and S, opioids) and other targets (neurotrophins BDNF and FGF2, glucocorticoids, L-type-calcium channels, epigenetic modifications) as well as their downstream signaling pathways, can augment fear extinction and strengthen extinction memory persistently in preclinical models. Particularly promising approaches are discussed in regard to their effects on specific aspects of fear extinction namely, acquisition, consolidation and retrieval, including long-term protection from return of fear (relapse) phenomena like spontaneous recovery, reinstatement and renewal of fear. We also highlight the promising translational value of the preclinial research and the clinical potential of targeting certain neurochemical systems with, for example d-cycloserine, yohimbine, cortisol, and L-DOPA. The current body of research reveals important new insights into the neurobiology and neurochemistry of fear extinction and holds significant promise for pharmacologically-augmented psychotherapy as an improved approach to treat trauma and anxiety-related disorders in a more efficient and persistent way promoting enhanced symptom remission and recovery.
Collapse
Affiliation(s)
- N Singewald
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria.
| | - C Schmuckermair
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - N Whittle
- Department of Pharmacology and Toxicology, Institute of Pharmacy and CMBI, Leopold-Franzens University of Innsbruck, Innrain 80-82, A-6020 Innsbruck, Austria
| | - A Holmes
- Laboratory of Behavioral and Genomic Neuroscience, National Institute on Alcohol Abuse and Alcoholism, NIH, Bethesda, MD, USA
| | - K J Ressler
- Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
98
|
Parks GS, Wang L, Wang Z, Civelli O. Identification of neuropeptide receptors expressed by melanin-concentrating hormone neurons. J Comp Neurol 2014; 522:3817-33. [PMID: 24978951 PMCID: PMC4167928 DOI: 10.1002/cne.23642] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Revised: 06/17/2014] [Accepted: 06/24/2014] [Indexed: 01/13/2023]
Abstract
Melanin-concentrating hormone (MCH) is a 19-amino-acid cyclic neuropeptide that acts in rodents via the MCH receptor 1 (MCHR1) to regulate a wide variety of physiological functions. MCH is produced by a distinct population of neurons located in the lateral hypothalamus (LH) and zona incerta (ZI), but MCHR1 mRNA is widely expressed throughout the brain. The physiological responses and behaviors regulated by the MCH system have been investigated, but less is known about how MCH neurons are regulated. The effects of most classical neurotransmitters on MCH neurons have been studied, but those of most neuropeptides are poorly understood. To gain insight into how neuropeptides regulate the MCH system, we investigated which neuropeptide receptors are expressed by MCH neurons by using double in situ hybridization. In all, 20 receptors, selected based on either a suspected interaction with the MCH system or demonstrated high expression levels in the LH and ZI, were tested to determine whether they are expressed by MCH neurons. Overall, 11 neuropeptide receptors were found to exhibit significant colocalization with MCH neurons: nociceptin/orphanin FQ opioid receptor (NOP), MCHR1, both orexin receptors (ORX), somatostatin receptors 1 and 2 (SSTR1, SSTR2), kisspeptin recepotor (KissR1), neurotensin receptor 1 (NTSR1), neuropeptide S receptor (NPSR), cholecystokinin receptor A (CCKAR), and the κ-opioid receptor (KOR). Among these receptors, six have never before been linked to the MCH system. Surprisingly, several receptors thought to regulate MCH neurons displayed minimal colocalization with MCH, suggesting that they may not directly regulate the MCH system.
Collapse
Affiliation(s)
- Gregory S. Parks
- Department of Pharmacology, University of California Irvine, Irvine, California 92697
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, 92697
| | - Lien Wang
- Department of Pharmacology, University of California Irvine, Irvine, California 92697
| | - Zhiwei Wang
- Department of Pharmacology, University of California Irvine, Irvine, California 92697
| | - Olivier Civelli
- Department of Pharmacology, University of California Irvine, Irvine, California 92697
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, California, 92697
- Department of Pharmaceutical Sciences, University of California Irvine, Irvine, California, 92697
| |
Collapse
|
99
|
Neuropeptide Y receptor gene expression in the primate amygdala predicts anxious temperament and brain metabolism. Biol Psychiatry 2014; 76:850-7. [PMID: 24342924 PMCID: PMC4022724 DOI: 10.1016/j.biopsych.2013.11.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Revised: 10/15/2013] [Accepted: 11/02/2013] [Indexed: 12/21/2022]
Abstract
BACKGROUND Anxious temperament (AT) is identifiable early in life and predicts the later development of anxiety disorders and depression. Neuropeptide Y (NPY) is a putative endogenous anxiolytic neurotransmitter that adaptively regulates responses to stress and might confer resilience to stress-related psychopathology. With a well-validated nonhuman primate model of AT, we examined expression of the NPY system in the central nucleus (Ce) of the amygdala, a critical neural substrate for extreme anxiety. METHODS In 24 young rhesus monkeys, we measured Ce messenger RNA (mRNA) levels of all members of the NPY system that are detectable in the Ce with quantitative real time polymerase chain reaction. We then examined the relationship between these mRNA levels and both AT expression and brain metabolism. RESULTS Lower mRNA levels of neuropeptide Y receptor 1 (NPY1R) and NPY5R but not NPY or NPY2R in the Ce predicted elevated AT; mRNA levels for NPY1R and NPY5R in the motor cortex were not related to AT. In situ hybridization analysis provided for the first time a detailed description of NPY1R and NPY5R mRNA distribution in the rhesus amygdala and associated regions. Lastly, mRNA levels for these two receptors in the Ce predicted metabolic activity in several regions that have the capacity to regulate the Ce. CONCLUSIONS Decreased NPY signaling in the Ce might contribute to the altered metabolic activity that is a component of the neural substrate underlying AT. This suggests that enhancement of NPY signaling might reduce the risk to develop psychopathology.
Collapse
|
100
|
Baldock PA, Lin S, Zhang L, Karl T, Shi Y, Driessler F, Zengin A, Hörmer B, Lee NJ, Wong IPL, Lin EJD, Enriquez RF, Stehrer B, During MJ, Yulyaningsih E, Zolotukhin S, Ruohonen ST, Savontaus E, Sainsbury A, Herzog H. Neuropeptide y attenuates stress-induced bone loss through suppression of noradrenaline circuits. J Bone Miner Res 2014; 29:2238-49. [PMID: 24535841 DOI: 10.1002/jbmr.2205] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2013] [Revised: 02/09/2014] [Accepted: 02/12/2014] [Indexed: 12/23/2022]
Abstract
Chronic stress and depression have adverse consequences on many organ systems, including the skeleton, but the mechanisms underlying stress-induced bone loss remain unclear. Here we demonstrate that neuropeptide Y (NPY), centrally and peripherally, plays a critical role in protecting against stress-induced bone loss. Mice lacking the anxiolytic factor NPY exhibit more anxious behavior and elevated corticosterone levels. Additionally, following a 6-week restraint, or cold-stress protocol, Npy-null mice exhibit three-fold greater bone loss compared to wild-type mice, owing to suppression of osteoblast activity. This stress-protective NPY pathway acts specifically through Y2 receptors. Centrally, Y2 receptors suppress corticotropin-releasing factor expression and inhibit activation of noradrenergic neurons in the paraventricular nucleus. In the periphery, they act to control noradrenaline release from sympathetic neurons. Specific deletion of arcuate Y2 receptors recapitulates the Npy-null stress response, coincident with elevated serum noradrenaline. Importantly, specific reintroduction of NPY solely in noradrenergic neurons of otherwise Npy-null mice blocks the increase in circulating noradrenaline and the stress-induced bone loss. Thus, NPY protects against excessive stress-induced bone loss, through Y2 receptor-mediated modulation of central and peripheral noradrenergic neurons.
Collapse
Affiliation(s)
- P A Baldock
- Neurological Disease Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, Australia; Osteoporosis and Bone Biology Division, Garvan Institute of Medical Research, St Vincent's Hospital, Sydney, Australia; Faculty of Medicine, University of New South Wales, Sydney, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|