Regulatory Mechanisms Underlying Corticotropin-Releasing Factor Gene Expression in the Hypothalamus

Total Page:16

File Type:pdf, Size:1020Kb

Regulatory Mechanisms Underlying Corticotropin-Releasing Factor Gene Expression in the Hypothalamus Endocrine Journal 2009, 56 (3), 335-344 REVIEW Regulatory Mechanisms Underlying Corticotropin-Releasing Factor Gene Expression in the Hypothalamus KAZUNORI KAGEYAMA AND TOSHIHIRO SUDA Department of Endocrinology and Metabolism, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Aomori 036-8562, Japan Abstract. The hypothalamic-pituitary-adrenal (HPA) axis is activated under various stressors. Corticotropin-releasing factor (CRF) plays a central role in controlling stress response, and regulating the HPA axis. CRF, produced in the hypothalamic paraventricular nucleus (PVN), stimulates adrenocorticotropic hormone (ACTH) production via CRF receptor type 1 (CRF1 receptor) from the corticotrophs of the anterior pituitary (AP). Cyclic AMP (cAMP)-protein kinase A (PKA) pathway takes a main role in stimulating CRF gene transcription. Forskolin and pituitary adenylate cyclase- activating polypeptide (PACAP) stimulate adenylate cyclase, intracellular cAMP production, and then CRF and arginine vasopressin (AVP) gene expression in hypothalamic 4B cells. Interleukin (IL)-6, produced in the PVN, both directly and indirectly stimulates CRF and AVP gene expression. Estradiol may enhance the activation of CRF gene expression in response to stress. The HPA axis is regulated by a negative feedback mechanism, because glucocorticoids inhibit both CRF production in the hypothalamic PVN and ACTH production in the pituitary. Hypothalamic parvocellular neurons in the PVN are known to express glucocorticoid receptors, and glucocorticoids are able to regulate CRF gene transcription and expression levels directly in the PVN. Glucocorticoids-dependent repression of cAMP-stimulated CRF promoter activity is mainly localized to promoter sequences between –278 and –233 bp. Both negative glucocorticoid regulatory element (nGRE) and serum response element (SRE) are involved in the repression of the CRF gene in the hypothalamic cells. Key words: Corticotropin-releasing factor, Hypothalamus, Stress, Cyclic AMP, Receptor (Endocrine Journal 56: 335-344, 2009) THE HYPOTHALAMIC-PITUItaRY-ADRENAL Stimulation of CRF gene in the hypothalamus (HPA) axis is activated under various stressors [1]. Corticotropin-releasing factor (CRF) plays a central Involvement of cAMP on CRF gene transcription role in controlling stress response, and regulating the HPA axis. CRF, produced in the hypothalamic paraven- Cyclic AMP (cAMP)-protein kinase A (PKA) path- tricular nucleus (PVN), stimulates adrenocorticotropic way takes a main role in stimulating CRF synthesis hormone (ACTH) production via CRF receptor type 1 [3-5]. There are several candidates for activating CRF (CRF1 receptor) from the corticotrophs of the anterior neurons. For example, pituitary adenylate cyclase-ac- pituitary (AP). ACTH then stimulates glucocorticoid tivating polypeptide (PACAP), a member of the secre- release from the adrenal glands [2]. Glucocorticoid in tin/glucagon/vasoactive intestinal peptide (VIP) fam- turn inhibits both CRF production in the hypothalamic ily, is one of the putative hormones. Both PACAP and PVN and ACTH production in the pituitary. the PACAP-selective PACAP receptor type 1 (PAC1 receptor) are known to be highly expressed in the hy- pothalamus, including the parvocellular and magno- cellular subdivisions of the PVN, and the supraoptic Received: March 9, 2009 nucleus (SON) [6, 7]. PACAP has shown to stimu- Accepted: March 11, 2009 late cAMP production in the AP [8]. PACAP also in- Correspondence to: Kazunori KAGEYAMA, M.D., Ph.D., Department of Endocrinology and Metabolism, Hirosaki creases CRF mRNA levels in the parvocellular region University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, of the PVN, suggesting that PACAP is involved in the Aomori 036-8562, Japan positive regulation of CRF gene expression [9]. 336 KAGEYAMA et al. 1000 Computer analysis of the proximal CRF promot- ** er reveals several possible binding sites for transcrip- ** ** tional factors, such as cAMP-response element (CRE), 800 activator protein 1 (AP-1) protein (Fos/Jun) binding n sites, half glucocorticoid regulatory element (GRE), ) and half estrogen-responsive element (ERE) [10]. 600 Forskolin or PACAP stimulates adenylate cyclase, ** ** and then increases intracellular cAMP levels in hypo- (% of control 400 thalamic 4B cells (Fig. 1) [11]. Forskolin increases cAMP productio CRF transcriptional activity in the hypothalamic cells (Fig. 2), which corresponds with previous studies us- 200 * ing other cells [4, 5, 12]. The forskolin-stimulated ac- tivity of CRF gene transcription is reduced in 4B cells 0 transfected with a mutant construct, CRF-233Mtluc, C 0.01 0.1 1101 10 100 1000 in which the CRE element (TGACGTCA) is mutat- Fsk (µM) PACAP (nM) ed (TGGATCCA), or a deletion mutant construct of Fig. 1. Effects of PACAP on cAMP production in 4B cells. the CRF gene promoter, CRF-220luc (Fig. 3) [13]. *P < 0.05, **P < 0.005 (compared with control (C)). Cells Therefore, the forskolin-induced CRF gene transcrip- were pre-incubated for 20 min with medium containing tion is mainly mediated by CRE, which includes -220 0.1 mM 3-isobutyl-1-methylxanthine, followed by to –233 bp, on the CRF 5’-promoter region in hypo- the addition of forskolin (Fsk) or PACAP. The level thalamic cells. of intracellular cAMP was measured by cAMP EIA. The PKA pathway is mainly involved in CRF gene (Reproduction from Ref. [11] with permission of the publisher.) Copyright 2007, Society for Endocrinology. regulation in the hypothalamic cells, because a PKA inhibitor strongly blocks the forskolin-induced CRF promoter activity [14]. This result supports our previ- ous report on the involvement of the PKA pathway in sion via the cAMP-PKA pathway in hypothalamic CRF synthesis and release in hypothalamic explants 4B cells (Fig. 4). PACAP is detected in nerve termi- [3]. Activation of the PKA pathway leads to binding nals that innervate AVP-containing neurons in the rat of CRE-binding protein (CREB) to the CRE on the hypothalamus [18], and its receptor mRNA is highly CRF promoter in hypothalamic cells as well as in hu- expressed in AVP-containing neurons [18]. The 4B man placental cells [15]. Protein kinase C (PKC) and cells also express PAC1 and VIP-preferring receptors p38 mitogen-activated protein (MAP) kinase are also (VPAC receptors). Therefore, activation of PAC1 or involved in the regulation of the forskolin-induced VPAC receptors by PACAP induces production of in- CRF gene expression, because both SB203580, an in- tracellular cAMP, activating the transcription of AVP hibitor highly specific for p38 kinase, and bisindo- gene in hypothalamic cells. Taken together, PACAP- lylmaleimide I (BIM), a PKC inhibitor, suppress for- induced intracellular cAMP production is involved skolin-induced CRF promoter activity [14]. At the in stimulating the transcription of both CRF and AVP same time, PD98059, a selective MAP kinase kinase genes via the PKA pathway in the hypothalamic cells. (MEK)-extracellular signal-related kinases (ERK) in- hibitor, and SP600125, a Jun amino-terminal kinas- Involvement of IL-6 on CRF gene transcription es (JNK) 1/2/3 inhibitor, does not show inhibitory ef- fects. Therefore, PKA, PKC, and p38 MAP kinase Interleukin (IL)-6 shows a variety of biological ac- pathways are involved in forskolin-induced activation tivities. Plasma IL-6 levels rise in response to both of CRF gene transcription in hypothalamic 4B cells. immune activation and non-immune stress [19, 20]. PACAP regulates the HPA axis by stimulation of IL-6 prompts lymphocytic proliferation and differ- CRF gene expression in the hypothalamus (Fig. 4) and entiation [21] and induces production of acute-phase through direct effects on pituitary corticotrophs [14, proteins in the liver [22]. In addition, IL-6 stimulates 16, 17]. PACAP also stimulates both arginine vaso- the HPA axis [23], resulting in increasing glucocorti- pressin (AVP) gene transcription and mRNA expres- coid levels, which in turn suppress IL-6 production [24, REGULATION OF CRF IN THE HYPOTHALAMUS 337 2000 2000 * * 1500 1500 ) ) 1000 1000 (% of control (% of control CRF 5’-Promoter Activity CRF 5’-Promoter Activity 500 500 0 0 0612 18 24 C 0.01 0.1 110 Time (h) Fsk (µM) Fig. 2. Effects of forskolin on CRF 5’-promoter activity in 4B cells. Control cells treated with medium alone are indicated as (C). *P < 0.05 (compared with control [C]). Time-dependent changes in forskolin-induced CRF 5’-promoter activity (left panel). Cells were incubated with medium containing 10mM forskolin. Dose-dependent changes in forskolin-induced CRF 5’-promoter activity (right panel). Cells were incubated for 2 h with medium containing 0.01 to 10mM forskolin (Fsk). (Reproduction from Ref. [13] with permission of the publisher.) Copyright 2008, Editrice Kurtis srl. -907 -233 -220 0+170 Control PACAP (2 h) PACAP (6 h) PACAP (24 h) (A) CRF Promoter CRE CRF (B) 2000 AVP 1500 B2MG ) + 300 * CRF 1000 + ** * AVP (% of control ) 200 * * CRF 5’-Promoter Activity 500 ** mRNA ** * (% of control 0 100 C Fsk C Fsk C Fsk C Fsk CRF-907luc CRF-233 luc CRF-233 Mt luc CRF-220 luc 0 Fig. 3. Effects of CRE deletion on forskolin-induced CRF 0 6 12 18 24 Time (h) 5’-promoter activity in 4B cells. (A) Schematic representation of CRE in the CRF promoter. (B) Fig. 4. Effects of PACAP on CRF and AVP mRNA levels. 4B Cells were transfected with full-length (CRF-907luc), Cells were stimulated with 100 nM PACAP for 2, 6, deleted (CRF-220luc or CRF-233luc), or mutant (CRF- and 24 h. Expression of CRF and AVP mRNA was 233Mtluc) promoter constructs, and then incubated for examined by RT-PCR. *P < 0.05, **P < 0.005 (compared 2h with 10mM forskolin alone (Fsk) or vehicle (C). *P with control). (Reproduction from Ref. [11] with < 0.05 (compared with forskolin alone [Fsk] in CRF- permission of the publisher.) Copyright 2007, Society 907luc transfected cells). +P < 0.05 (compared with each for Endocrinology. control). (Reproduction from Ref. [13] with permission of the publisher.) Copyright 2008, Editrice Kurtis srl. 338 KAGEYAMA et al. 800 M4BHypo AP C1 C2 C3 ** ERα 600 ** ) ** ++ ERβ 400 * (% of control # B2MG CRF 5’ Promoter Activity 200 Fig.
Recommended publications
  • Roles for Androgens in Mediating the Sex Differences of Neuroendocrine and Behavioral Stress Responses Damian G
    Zuloaga et al. Biology of Sex Differences (2020) 11:44 https://doi.org/10.1186/s13293-020-00319-2 REVIEW Open Access Roles for androgens in mediating the sex differences of neuroendocrine and behavioral stress responses Damian G. Zuloaga1, Ashley L. Heck2, Rose M. De Guzman1 and Robert J. Handa2* Abstract Estradiol and testosterone are powerful steroid hormones that impact brain function in numerous ways. During development, these hormones can act to program the adult brain in a male or female direction. During adulthood, gonadal steroid hormones can activate or inhibit brain regions to modulate adult functions. Sex differences in behavioral and neuroendocrine (i.e., hypothalamic pituitary adrenal (HPA) axis) responses to stress arise as a result of these organizational and activational actions. The sex differences that are present in the HPA and behavioral responses to stress are particularly important considering their role in maintaining homeostasis. Furthermore, dysregulation of these systems can underlie the sex biases in risk for complex, stress-related diseases that are found in humans. Although many studies have explored the role of estrogen and estrogen receptors in mediating sex differences in stress-related behaviors and HPA function, much less consideration has been given to the role of androgens. While circulating androgens can act by binding and activating androgen receptors, they can also act by metabolism to estrogenic molecules to impact estrogen signaling in the brain and periphery. This review focuses on androgens as an important hormone for modulating the HPA axis and behaviors throughout life and for setting up sex differences in key stress regulatory systems that could impact risk for disease in adulthood.
    [Show full text]
  • Cionin, a Vertebrate Cholecystokinin/Gastrin
    www.nature.com/scientificreports OPEN Cionin, a vertebrate cholecystokinin/gastrin homolog, induces ovulation in the ascidian Ciona intestinalis type A Tomohiro Osugi, Natsuko Miyasaka, Akira Shiraishi, Shin Matsubara & Honoo Satake* Cionin is a homolog of vertebrate cholecystokinin/gastrin that has been identifed in the ascidian Ciona intestinalis type A. The phylogenetic position of ascidians as the closest living relatives of vertebrates suggests that cionin can provide clues to the evolution of endocrine/neuroendocrine systems throughout chordates. Here, we show the biological role of cionin in the regulation of ovulation. In situ hybridization demonstrated that the mRNA of the cionin receptor, Cior2, was expressed specifcally in the inner follicular cells of pre-ovulatory follicles in the Ciona ovary. Cionin was found to signifcantly stimulate ovulation after 24-h incubation. Transcriptome and subsequent Real-time PCR analyses confrmed that the expression levels of receptor tyrosine kinase (RTK) signaling genes and a matrix metalloproteinase (MMP) gene were signifcantly elevated in the cionin-treated follicles. Of particular interest is that an RTK inhibitor and MMP inhibitor markedly suppressed the stimulatory efect of cionin on ovulation. Furthermore, inhibition of RTK signaling reduced the MMP gene expression in the cionin-treated follicles. These results provide evidence that cionin induces ovulation by stimulating MMP gene expression via the RTK signaling pathway. This is the frst report on the endogenous roles of cionin and the induction of ovulation by cholecystokinin/gastrin family peptides in an organism. Ascidians are the closest living relatives of vertebrates in the Chordata superphylum, and thus they provide important insights into the evolution of peptidergic systems in chordates.
    [Show full text]
  • Glucagon-Like Peptide-1 Reduces Pancreatic Β-Cell Mass Through
    www.nature.com/scientificreports OPEN Glucagon-like peptide-1 reduces pancreatic β-cell mass through hypothalamic neural pathways in Received: 3 October 2016 Accepted: 30 May 2017 high-fat diet-induced obese rats Published: xx xx xxxx Hisae Ando, Koro Gotoh, Kansuke Fujiwara, Manabu Anai, Seiichi Chiba, Takayuki Masaki, Tetsuya Kakuma & Hirotaka Shibata We examined whether glucagon-like peptide-1 (GLP-1) affectsβ -cell mass and proliferation through neural pathways, from hepatic afferent nerves to pancreatic efferent nerves via the central nervous system, in high-fat diet (HFD)-induced obese rats. The effects of chronic administration of GLP-1 (7–36) and liraglutide, a GLP-1 receptor agonist, on pancreatic morphological alterations, c-fos expression and brain-derived neurotrophic factor (BDNF) content in the hypothalamus, and glucose metabolism were investigated in HFD-induced obese rats that underwent hepatic afferent vagotomy (VgX) and/ or pancreatic efferent sympathectomy (SpX). Chronic GLP-1 (7–36) administration to HFD-induced obese rats elevated c-fos expression and BDNF content in the hypothalamus, followed by a reduction in pancreatic β-cell hyperplasia and insulin content, thus resulting in improved glucose tolerance. These responses were abolished by VgX and SpX. Moreover, administration of liraglutide similarly activated the hypothalamic neural pathways, thus resulting in a more profound amelioration of glucose tolerance than native GLP-1 (7–36). These data suggest that GLP-1 normalizes the obesity-induced compensatory increase in β-cell mass and glucose intolerance through a neuronal relay system consisting of hepatic afferent nerves, the hypothalamus, and pancreatic efferent nerves. β-cell mass, which is determined by the product of the number and size of pancreatic β-cells, is tightly con- trolled to maintain glucose levels within a normal range.
    [Show full text]
  • Neuropeptidergic Signaling Partitions Arousal Behaviors in Zebrafish
    3142 • The Journal of Neuroscience, February 26, 2014 • 34(9):3142–3160 Behavioral/Cognitive Neuropeptidergic Signaling Partitions Arousal Behaviors in Zebrafish Ian G. Woods,1,2 David Schoppik,2 Veronica J. Shi,2 Steven Zimmerman,2 Haley A. Coleman,1 Joel Greenwood,3 Edward R. Soucy,3 and Alexander F. Schier2,3 1Department of Biology, Ithaca College, Ithaca, New York 14850, and 2Department of Molecular and Cellular Biology and 3Center for Brain Science, Harvard University, Cambridge, Massachusetts 02138 Animals modulate their arousal state to ensure that their sensory responsiveness and locomotor activity match environmental demands. Neuropeptides can regulate arousal, but studies of their roles in vertebrates have been constrained by the vast array of neuropeptides and their pleiotropic effects. To overcome these limitations, we systematically dissected the neuropeptidergic modulation of arousal in larval zebrafish. We quantified spontaneous locomotor activity and responsiveness to sensory stimuli after genetically induced expression of seven evolutionarily conserved neuropeptides, including adenylate cyclase activating polypeptide 1b (adcyap1b), cocaine-related and amphetamine-related transcript (cart), cholecystokinin (cck), calcitonin gene-related peptide (cgrp), galanin, hypocretin, and nocicep- tin. Our study reveals that arousal behaviors are dissociable: neuropeptide expression uncoupled spontaneous activity from sensory responsiveness, and uncovered modality-specific effects upon sensory responsiveness. Principal components analysis and phenotypic clustering revealed both shared and divergent features of neuropeptidergic functions: hypocretin and cgrp stimulated spontaneous locomotor activity, whereas galanin and nociceptin attenuated these behaviors. In contrast, cart and adcyap1b enhanced sensory respon- siveness yet had minimal impacts on spontaneous activity, and cck expression induced the opposite effects. Furthermore, hypocretin and nociceptin induced modality-specific differences in responsiveness to changes in illumination.
    [Show full text]
  • Neuropeptidergic Modulation of Gnrh Neuronal Activity and Gnrh Secretion Controlling Reproduction: Insights from Recent Mouse Studies
    Cell and Tissue Research (2019) 375:179–191 https://doi.org/10.1007/s00441-018-2893-z REVIEW Neuropeptidergic modulation of GnRH neuronal activity and GnRH secretion controlling reproduction: insights from recent mouse studies Daniel J. Spergel1 Received: 1 April 2018 /Accepted: 6 July 2018 /Published online: 4 August 2018 # Springer-Verlag GmbH Germany, part of Springer Nature 2018 Abstract Gonadotropin-releasing hormone (GnRH) secretion from GnRH neurons and its modulation by neuropeptides are essential for mammalian reproduction. Here, I review the neuropeptides that have been shown to act directly and that may also act indirectly, on GnRH neurons, the reproduction-related processes with which the neuropeptides may be associated or the physiological information they may convey, as well as their cognate receptors, signaling pathways and roles in the modulation of GnRH 2+ neuronal firing, [Ca ]i, GnRH secretion and reproduction. The review focuses on recent research in mice, which offer the most tractable experimental system for studying mammalian GnRH neurons. Keywords GnRH neuron . Neuropeptide . Receptor . Signaling . Reproduction Introduction Leshan et al. 2009; Roa and Herbison 2012; Liu et al. 2014; Cimino et al. 2016; Hellier et al. 2018; Phumsatitpong and Gonadotropin-releasing hormone (GnRH; also known as Moenter 2018). The cell bodies of GnRH neurons, which re- GnRH1 or LHRH) neurons comprise a scattered group of ceive neuropeptidergic inputs from neurons in the hypothalamus ~ 600–800 cells in the mouse brain, with only ~ 70–200 of those and other brain areas (Turi et al. 2003;Yipetal.2015), are cells being required for reproductive function, which form the distributed along a continuum in the medial and lateral preoptic final common pathway for the central control of reproduction area (POA) of the hypothalamus, the horizontal and vertical (Wray et al.
    [Show full text]
  • Oxytocin Modulation of Neural Circuits for Social Behavior
    Oxytocin Modulation of Neural Circuits for Social Behavior Bianca J. Marlin,1,2 Robert C. Froemke3,4,5 1 Department of Neuroscience, Columbia University, New York, New York 10032 2 Howard Hughes Medical Institute, College of Physicians and Surgeons, Columbia University, New York, New York 10032 3 Department of Otolaryngology, Skirball Institute for Biomolecular Medicine, Neuroscience Institute, New York University School of Medicine, New York, New York 4 Department of Neuroscience and Physiology Skirball Institute for Biomolecular Medicine, Neuroscience Institute New York University School of Medicine, New York, New York 5 Center for Neural Science, New York University, New York, New York Received 23 June 2016; revised 7 September 2016; accepted 12 September 2016 ABSTRACT: Oxytocin is a hypothalamic neuro- on social behavior and circuit plasticity in various brain peptide that has gained attention for the effects on social regions shown to be enriched for oxytocin receptors. We behavior. Recent findings shed new light on the mecha- provide a framework that highlights current circuit-level nisms of oxytocin in synaptic plasticity and adaptively mechanisms underlying the widespread action of oxyto- modifying neural circuits for social interactions such as cin. VC 2016 Wiley Periodicals, Inc. Develop Neurobiol 77: 169–189, 2017 conspecific recognition, pair bonding, and maternal care. Keywords: cortex; inhibition; neural circuits; oxytocin; Here, we review several of these newer studies on oxyto- synaptic plasticity cin in the context of previous findings, with an emphasis INTRODUCTION encoding of behaviorally important experiences. One neuromodulator in particular, the peptide hormone Neural circuits are plastic, changing in response to oxytocin, has been implicated in an array of social sensory stimuli and environmental interactions.
    [Show full text]
  • The Melanin-Concentrating Hormone (MCH) System: a Tale of Two Peptides
    fnins-13-01280 November 23, 2019 Time: 16:1 # 1 REVIEW published: 26 November 2019 doi: 10.3389/fnins.2019.01280 The Melanin-Concentrating Hormone (MCH) System: A Tale of Two Peptides Giovanne B. Diniz1,2 and Jackson C. Bittencourt1,3* 1 Departamento de Anatomia, Instituto de Ciências Biomedicas, Universidade de São Paulo, São Paulo, Brazil, 2 Department of Neurosurgery, Yale School of Medicine, New Haven, CT, United States, 3 Nucleo de Neurociencias e Comportamento, Instituto de Psicologia, Universidade de São Paulo, São Paulo, Brazil The melanin-concentrating hormone (MCH) system is a robust integrator of exogenous and endogenous information, modulating arousal and energy balance in mammals. Its predominant function in teleosts, however, is to concentrate melanin in the scales, contributing to the adaptive color change observed in several teleost species. These contrasting functions resulted from a gene duplication that occurred after the teleost divergence, which resulted in the generation of two MCH-coding genes in this clade, which acquired distinctive sequences, distribution, and functions, examined in detail here. We also describe the distribution of MCH immunoreactivity and gene expression in a large number of species, in an attempt to identify its core elements. While initially originated as a periventricular peptide, with an intimate relationship with the Edited by: third ventricle, multiple events of lateral migration occurred during evolution, making Dan Larhammar, the ventrolateral and dorsolateral hypothalamus the predominant sites of MCH in Uppsala University, Sweden teleosts and mammals, respectively. Substantial differences between species can Reviewed by: Hervé Tostivint, be identified, likely reflecting differences in habitat and behavior. This observation Muséum National d’Histoire Naturelle, aligns well with the idea that MCH is a major integrator of internal and external France information, ensuring an appropriate response to ensure the organism’s homeostasis.
    [Show full text]
  • Enhancement of Brain Melanocortin Signaling In
    ENHANCEMENT OF BRAIN MELANOCORTIN SIGNALING IN LEAN, ACTIVE RATS A dissertation submitted to Kent State University in partial fulfillment of the requirements for the degree of Doctor of Philosophy By Charu Shukla April 2014 Dissertation written by Charu Shukla M.S., University of Alabama at Birmingham, USA, 2009 M.Sc., Panjab University, India, 2007 B.Sc., CCS University, India 2005 Approved by Dr. Colleen M. Novak , Chair, Doctoral Dissertation Committee Dr. Gail C. Fraizer, Member, Doctoral Dissertation Committee Dr. Edgar E. Kooijman, Member, Doctoral Dissertation Committee Dr. Aaron M. Jasnow, Member, Doctoral Dissertation Committee Dr. Soumitra Basu, Member, Doctoral Dissertation Committee Accepted by Dr. Laura G. Leff, Acting Chair, Department of Biological Sciences Dr. Jan Crowther, Associate Dean, College of Arts and Sciences ii TABLE OF CONTENTS Page Appendix 1: List of Figures ............................................................................................. vi Appendix 2: List of Tables ............................................................................................... xi Appendix 3: List of Abbreviations used ....................................................................... xii Acknowledgements………………………………………………………………......... xv CHAPTER 1 : Introduction ................................................................................................. 1 1.1 Obesity ................................................................................................................... 1 1.2 Physical Activity
    [Show full text]
  • The Preproglucagon Derived Peptides and Energy
    THE PREPROGLUCAGON DERIVED PEPTIDES AND ENERGY HOMEOSTASIS A thesis submitted for the degree of Doctor of Philosophy Jennifer Parker 2013 Department of Medicine Imperial College London Abstract Obesity is a major contributor to the development of chronic diseases, and there is a paucity of effective treatments. Recent studies suggest that co-agonists at the glucagon-like peptide-1 (GLP-1) and glucagon receptor efficaciously reduce body weight and improve glucose homeostasis. This thesis explores the effects of glucagon, GLP-1 and the endogenous GLP-1/glucagon receptor co-agonist oxyntomodulin, on appetite and glucose homeostasis and their mechanisms. As expected, peripheral injection of GLP-1 or glucagon to fasted mice transiently reduced food intake. Interestingly, subanorectic doses of GLP-1 and glucagon potently inhibited food intake when combined. Agonists at the GLP-1 (GLPAg) and glucagon (GCGAg) receptors designed in this laboratory were found to have receptor affinities comparable with those of GLP-1 and GCG, but with a prolonged duration of action. When administered chronically, individually and in combination, to an obese mouse model, the combination of these peptides appeared to cause superior reduction in body weight and improvement in glucose tolerance compared to the individual peptides. The receptors and central appetite regulating centres involved in the response to anorectic doses of GLP-1, glucagon and oxyntomodulin were investigated. The pattern of c-fos immunoreactivity in response to glucagon was examined for the first time and appeared indistinguishable from that induced by GLP-1. Oxyntomodulin appeared to induce greater c-fos activation in the nucleus tractus solitarius (NTS) than either glucagon or GLP-1 at equivalently anorectic doses.
    [Show full text]
  • Identification of Putative Neuropeptidergic Signaling Systems
    Invertebrate Neuroscience (2020) 20:2 https://doi.org/10.1007/s10158-020-0235-9 SHORT COMMUNICATION Identifcation of putative neuropeptidergic signaling systems in the spiny lobster, Panulirus argus Andrew E. Christie1 Received: 8 November 2019 / Accepted: 4 January 2020 / Published online: 24 January 2020 © Springer-Verlag GmbH Germany, part of Springer Nature 2020 Abstract Members of the decapod infraorder Achelata, specifcally species from the genus Panulirus, have storied histories as models for investigating the basic principles governing the generation, maintenance, and modulation of rhythmic motor behavior, including modulation by locally released and circulating peptides. Despite their contributions to our understanding of pepti- dergic neuromodulation, little is known about the identity of the native neuropeptides and neuronal peptide receptors present in these crustaceans. Here, a Panulirus argus nervous system-specifc transcriptome was used to help fll this void, provid- ing insight into the neuropeptidome and neuronal peptide receptome of this species. A neuropeptidome consisting of 266 distinct peptides was predicted using the P. argus assembly, 128 having structures placing them into a generally recognized arthropod peptide family: agatoxin-like peptide, allatostatin A (AST-A), allatostatin B, allatostatin C, bursicon, CCHamide, crustacean cardioactive peptide, crustacean hyperglycemic hormone/molt-inhibiting hormone, diuretic hormone 31 (DH31), ecdysis-triggering hormone (ETH), FMRFamide-like peptide (FLP), glycoprotein hormone (GPH), GSEFLamide, inotocin, leucokinin, myosuppressin, natalisin, neuroparsin, neuropeptide F, orcokinin, orcomyotropin, periviscerokinin, pigment- dispersing hormone, pyrokinin, red pigment-concentrating hormone, RYamide, short neuropeptide F (sNPF), SIFamide, sulfakinin, tachykinin-related peptide (TRP), and trissin. Twenty-fve putative neuronal receptors, encompassing 15 peptide groups, were also identifed from the P.
    [Show full text]
  • Molecular Evolution of Peptidergic Signaling Systems in Bilaterians
    Molecular evolution of peptidergic signaling systems in bilaterians Olivier Mirabeau1 and Jean-Stéphane Joly Unité propre de Recherche 3294, Neurobiology and Development Research Unit, Institut Fessard, Centre National de la Recherche Scientifique and Institut National de la Recherche Agronomique, 91198 Gif-sur-Yvette, France Edited by John Gerhart, University of California, Berkeley, CA, and approved April 12, 2013 (received for review November 25, 2012) Peptide hormones and their receptors are widespread in metazo- atode Caenorhabditis elegans (12), and the mosquito Anopheles ans, but the knowledge we have of their evolutionary relationships gambiae (13). In these species, original genome-wide searches remains unclear. Recently, accumulating genome sequences from have revealed the existence of a large number of GPCRs that many different species have offered the opportunity to reassess resembled vertebrate GPCRs (11), but comparatively few ver- the relationships between protostomian and deuterostomian pepti- tebrate-type peptides (11, 12, 14). dergic systems (PSs). Here we used sequences of all human rhodopsin Before the genomic era, some researchers had postulated a and secretin-type G protein-coupled receptors as bait to retrieve deep orthology between PSs from distant animals on the basis of potential homologs in the genomes of 15 bilaterian species, in- peptide primary sequence similarity (15), functional analogies cluding nonchordate deuterostomian and lophotrochozoan spe- (16), and immunoreactivity of invertebrate tissues to mammalian cies. Our phylogenetic analysis of these receptors revealed 29 well- hormone antibodies (17), but the idea that it could be a general supported subtrees containing mixed sets of protostomian and feature of PSs remained controversial. Now, with the accumu- deuterostomian sequences.
    [Show full text]
  • Neuropeptide Relay Between Sifa Signaling Controls the Experience-Dependent Mating
    bioRxiv preprint doi: https://doi.org/10.1101/819045; this version posted October 25, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Neuropeptide relay between SIFa signaling controls the experience-dependent mating duration of male Drosophila Kyle Wong1, Justine Schweizer1, Khoi-Nguyen Ha Nguyen1, Shatha Atieh1, and Woo Jae Kim1,* 1Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada *Correspondence: [email protected] bioRxiv preprint doi: https://doi.org/10.1101/819045; this version posted October 25, 2019. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Summary Drosophila melanogaster is a suitable model for investigating how neuropeptides influence animal behaviours and physiology. We previously reported that two behavioural paradigms control mating duration of male Drosophila, called Longer-Mating-Duration (LMD) and Shorter-Mating-Duration (SMD) that are induced through socio-sexual environment prior to copulation. Understanding the molecular and cellular mechanisms by which males exhibit plasticity to different social cues remains poorly understood. Here, we show that SIFa modulates the neural circuitry for both LMD and SMD. Neuropeptide-to-neuropeptide communication, so called ‘neuropeptide relay’ plays a key role to mediate this control. We identified that 7 neuropeptides expressed in SIFa Receptor-positive cells are functionally important to regulate either LMD and/or SMD.
    [Show full text]