Neuropeptide Receptor Positive Allosteric Modulation in Epilepsy: Galanin Modulation Revealed

Total Page:16

File Type:pdf, Size:1020Kb

Neuropeptide Receptor Positive Allosteric Modulation in Epilepsy: Galanin Modulation Revealed COMMENTARY Neuropeptide receptor positive allosteric modulation in epilepsy: Galanin modulation revealed Daniel Hoyer1 Neuropsychiatry/Neuroscience Research, WSJ-386/745, Novartis Institutes for Biomedical Research Basel, CH 4002 Basel, Switzerland he therapeutic potential of neu- Galanin and Epilepsy neuropeptide receptor agonists are largely ropeptides has long been recog- There is ample evidence that galanin, like peptide analogs. Lu et al. describe an LMW T nized; some successes have been NPY or somatostatin, plays a role in epi- PAM that is reasonably potent at and se- registered, for example, in the lepsy, because when applied to the brain/ lective for a neuropeptide receptor. There tachykinin, endothelin, somatostatin, an- hippocampus these neuropeptides or are a few examples of allosteric modulators giotensin, calcitonin gene-related peptide their analogs have antiepileptic properties at corticotropin-releasing factor, CGRP, (CGRP), orexin, or glucagon-like peptide (2–4). Further, knockout or knockdown of GLP1, or ghrelin receptors, primarily 1 (GLP1) fields (1), primarily in peripheral their receptors result in increased sensi- antagonists (6). When it comes to agonists, applications. However, given the nature tivity to seizures, as reported for NPY2, almost no neuropeptide receptor PAMs of receptor–ligand interactions at the sst2/sst4 receptors (depending on species), are described. More generally, LMW, cen- orthosteric site of G protein-coupled re- and GalR1/GalR2. GalR1 and GalR2 are trally acting, bioavailable, and metaboli- ceptors (GPCRs), which seem to involve expressed in the hippocampus, whereas cally stable agonists acting at neuropeptide multiple pharmacophores that are hard to GalR3 is not. It is known that galanin, receptors are rare. L-692,429 is a ghrelin display on low-molecular-weight (LMW) receptor PAM, one of the very few peptide ligands, it is not surprising that the number Lu et al. describe an LMW receptor PAMs to be described (7). Addi- of such ligands, especially agonists, is very tionally, LMW ago-allosteric compounds limited, particularly considering brain- PAM that is reasonably acting on GLP1 receptors exist (8). Ghrelin penetrant ones. This is not because large and GLP1 are not commonly accepted as pharmaceutical companies have neglected potent at and selective neuropeptides, although this may change as modern high throughput screening tech- central effects are being investigated. Al- fi nology to nd such drug candidates. Many for a neuropeptide losteric modulation of GPCRs offers great synthetic neuropeptide analogs exist, but opportunities, especially for receptors for most of them are peptides, with inherent receptor. which the chemical space is limited (e.g., limitations in biovailability, plasma half- glutamate, GABAB, calcium sensing, and life/proteolytic degradation, brain pene- now neuropeptides); it is also expected that NPY, or somatostatin are released dur- tration, and cost of goods. More recently, orthosteric and allosteric sites are very dif- alternate approaches in the GPCR field ing epileptogenesis, and one may argue ferent (although ago-allosteric ligands emerged with negative or positive alloste- that such a release may contribute to may act on both), and it seems that allo- ric modulators (PAM), acting as antago- seizures; however, this is not so, because steric sites with a receptor subfamily are nists or agonists, but only in the presence mice constitutively overexpressing the rather divergent, a good basis for selectivity. of the endogenous agonist. Galanin, which neuropeptides or subjected to lentivirus- An allosteric modulator is expected to be acts on three GPCRs (GalR1–3), has long or recombinant adeno-associated virus- largely devoid of (side) effects (if selective been proposed to play a role in seizures; mediated overexpression are also less however, with few exceptions, the tools susceptible to seizures (2, 5). Lu et al. (2) enough) because it is inoperant in the ab- described are still peptides or peptidomi- now make the point that increased galanin sence of the endogenous ligand (6, 9). In metics. Now, Lu et al. (2) report in PNAS release plays a protective role, although not addition, it is not expected to induce de- on the discovery of CYM2503, an LMW, enough galanin is released to provide full sensitization, an issue with some neuro- potent, selective, and in vivo active GalR2 protection. However, the GalR2 PAM peptides. Negative allosteric modulators PAM, effective in several modalities (which per se has no activating effect), by (antagonists) are in clinical development linked to epilepsy in rodents. modulating endogenous peptide induces (10), and a few are on the market (9); it re- This is a major contribution to the a large degree of protection in different mains to be seen whether neuropeptide re- galanin and GPCR fields in that a PAM models of seizure and epileptogenesis. ceptor PAMs will be as successful, but the of GalR2 is described: CYM2503 does not Their article also suggests that targeting data presented here are very promising. Fi- interact with the orthosteric site of GlaR2 receptor may suffice, although pre- nally, whether a GalR2 PAM has peripheral GalR2 and is devoid of effect on GalR1 vious work also suggested a role for GalR1. effects is to be investigated. Interestingly, binding or function. CYM2503 is effective It remains to be seen whether a com- Fallarino et al. (11) report on the effects of in vivo in various models of mouse or bined effect at GalR1 and GalR2 may im- an MgluR4 PAM in animal models of mul- rat seizures and epileptogenesis and is as prove the outcome, although this may be tiple sclerosis [experimental autoimmune fi active as a reference antiepileptic, making dif cult to achieve; it is assumed that allo- encephalomyelitis (EAE)]; that article also at least two points. (i) GalR2 PAM steric sites among GPCR subfamily mem- concludes that endogenous glutamate is re- seems to be a valid principle in the treat- bers are highly different. In other words, to leased as a protective mechanism in rodent ment of seizures/epileptogenesis. (ii) The achieve a PAM for both GalR1 and GalR2 EAE models and that an mGluR4 PAM, just study validates the fact that galanin, seems rather unrealistic. which like other neuropeptides [somato- statin, neuropeptide Y (NPY)] is released Allosteric Modulators of Peptide Author contributions: D.H. wrote the paper. after seizure induction and can indeed Receptors The author declares no conflict of interest. be modulated to provide protection The present work highlights another major See companion article on page 15229. against seizures. advantage of the PAM approach, because 1E-mail: [email protected]. www.pnas.org/cgi/doi/10.1073/pnas.1010365107 PNAS | August 24, 2010 | vol. 107 | no. 34 | 14943–14944 Downloaded by guest on September 28, 2021 like the GalR2 PAM by Lu et al. (2), is able brain-penetrant galanin receptor agonist galanin peptide analogs (e.g., NAX5055) that to confer additional protection. (12–17). Numerous attempts in that direction have antiepileptic effects when applied sys- This represents the near-culmination of a were made: galmic and galnon are two LMW temically have since been described; how- long search. Tamas Bartfai received galanin nonpeptide GalR agonists; however, their ever, NAX5055 has a relatively short half-life fi from Viktor Mutt in late 1986 and since af nity for GalRs and a number of other in vivo and, being a peptide, will lack bio- then has published more than 140 papers on GPCRs is in the high micromolar range, thus galanin, galanin receptors, and their func- a more selective compound was needed. In- availability (18); furthermore, cost of goods is tional and pathophysiological roles, with terestingly, although galnon is unselective, it not to be neglected with peptides. Thus, emphasis on epilepsy, pain, and other CNS is a good tool for screening modulators or CYM2503 seems to be very close to the goal disorders, the ultimate aim being to discover antagonists at recombinant cell lines, set by Bartfai and his collaborators, and an LMW, systemically active, selective and galnon being more stable than galanin. A few many others, some time ago. 1. Hökfelt T, Bartfai T, Bloom F (2003) Neuropeptides: Op- retagogues act both as ghrelin receptor agonist and as 13. Fisone G, et al. (1989) N-terminal galanin-(1-16) frag- portunities for drug discovery. Lancet Neurol 2:463–472. positive or negative allosteric modulators of ghrelin ment is an agonist at the hippocampal galanin recep- 2. Lu X, et al. (2010) GalR2 positive allosteric modulator signaling. Mol Endocrinol 19:2400–2411. tor. Proc Natl Acad Sci USA 86:9588–9591. exhibits anticonvulsant effects in animal models. Proc 8. Knudsen LB, et al. (2007) Small-molecule agonists for 14. Bartfai T, et al. (1991) M-15: High-affinity chimeric pep- Natl Acad Sci USA 107:15229–15234. the glucagon-like peptide 1 receptor. Proc Natl Acad tide that blocks the neuronal actions of galanin in the 3. Vezzani A, et al. (1994) Enhanced neuropeptide Y re- Sci USA 104:937–942. hippocampus, locus coeruleus, and spinal cord. Proc lease in the hippocampus is associated with chronic 9. Conn PJ, Christopoulos A, Lindsley CW (2009) Alloste- Natl Acad Sci USA 88:10961–10965. seizure susceptibility in kainic acid treated rats. Brain ric modulators of GPCRs: A novel approach for the 15. Bartfai T, et al. (2004) Galmic, a nonpeptide galanin recep- Res 660:138–143. treatment of CNS disorders. Nat Rev Drug Discov 8: tor agonist, affects behaviors in seizure, pain, and forced- 4. Vezzani A, Hoyer D (1999) Brain somatostatin: A can- 41–54. swim tests. Proc Natl Acad Sci USA 101:10470–10475. didate inhibitory role in seizures and epileptogenesis. 10. Berry-Kravis E, et al. (2009) A pilot open label, single 16. Crawley JN, Robinson JK, Langel U, Bartfai T (1993) Eur J Neurosci 11:3767–3776. dose trial of fenobam in adults with fragile X syn- Galanin receptor antagonists M40 and C7 block galanin- 5.
Recommended publications
  • Bone-Specific Master Transcription Factor Runx2 Regulates Signaling and Metabolism Related Programs in Osteoprogenitors
    ISSN 0233-7657. Biopolymers and Cell. 2010. Vol. 26. N 4 Bone-specific master transcription factor Runx2 regulates signaling and metabolism related programs in osteoprogenitors N. M. Teplyuk1, 2, V. I. Teplyuk2 1University of Massachusetts Medical School 55, Lake Ave North, 01655, Worcester, MA, USA 2Institute of Molecular Biology and Genetics of National Academy of Sciences of Ukraine 150, Zabolotnogo str., Kiev, Ukraine, 03680 [email protected] Aim. Runx2 (AML3) transcription factor is the key regulator of osteoblastic lineage progression and is indispensable for the formation of mineral bones. Runx2 expression increases during differentiation of osteoblasts to induce osteoblast-specific genes necessary for the production and deposition of bone mineral matrix. However, Runx2 is also expressed at a lower level in early osteoprogenitors, where its function is less understood. Here we study how Runx2 determines the early stages of osteoblastic commitment using the model system of Runx2 re-introduction in mouse calvaria cells with Runx2 null background. Method. Affymetrix analysis, Western blot analysis and quantitative real-time reverse transcriptase PCR (qRT-PCR) analysis were employed. Results. Gene expression profiling by Affymetrix microarrays revealed that along with the induction of extracellular matrix and bone mineral deposition related phenotypic markers, Runx2 regulates several cell programs related to signaling and metabolism in the early osteoprogenitors. Particularly, Runx2 regulates transcription of genes involved in G-protein coupled signaling network, FGF and BMP/TGF beta signaling pathways and in biogenesis and metabolism pathways of steroid hormones. Conclusion. The data indicate that the lineage specific program, regulated by the master regulatory transcription factor, includes the regulation of cellular signaling and metabolism which may allow the committed cell to react and behave differently in the same microenvironment.
    [Show full text]
  • The Cardiopulmonary Effects of the Calcitonin Gene- Related Peptide Family Kalsitonin-Geni İle İlişkili Peptit Ailesinin Kardiyopulmoner Etkileri
    Turk J Pharm Sci 2020;17(3):349-356 DOI: 10.4274/tjps.galenos.2019.47123 REVIEW The Cardiopulmonary Effects of the Calcitonin Gene- related Peptide Family Kalsitonin-Geni İle İlişkili Peptit Ailesinin Kardiyopulmoner Etkileri Gökçen TELLİ1, Banu Cahide TEL1, Bülent GÜMÜŞEL2* 1Hacettepe University Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey 2Lokman Hekim University Faculty of Pharmacy, Department of Pharmacology, Ankara, Turkey ABSTRACT Cardiopulmonary diseases are very common among the population. They are high-cost diseases and there are still no definitive treatments. The roles of members of the calcitonin-gene related-peptide (CGRP) family in treating cardiopulmonary diseases have been studied for many years and promising results obtained. Especially in recent years, two important members of the family, adrenomedullin and adrenomedullin2/intermedin, have been considered new treatment targets in cardiopulmonary diseases. In this review, the roles of CGRP family members in cardiopulmonary diseases are investigated based on the studies performed to date. Key words: CGRP family, cardiopulmonary diseases, adrenomedullin, adrenomedullin2/intermedin, pulmonary hypertension ÖZ Kardiyopulmoner hastalıklar toplumda sık görülen, tedavi maliyeti oldukça yüksek ve halen kesin bir tedavisi bulunmayan hastalıklardır. Kalsitonin- geni ile ilişkili peptit (CGRP) ailesinin üyelerinin bir çok kardiyopulmoner hastalıktaki rolleri uzun yıllardır çalışılmakta ve umut vadeden sonuçlar elde edilmektedir. Özellikle son yıllarda CGRP ailesine
    [Show full text]
  • One-Month of High-Intensity Exercise Did Not Change the Food Intake And
    This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/ licenses/ by-nc-nd/3.0), which permits copy and redistribute the material in any medium or format, provided the original work is properly cited. 8 ENDOCRINE REGULATIONS, Vol. 53, No. 1, 8–13, 2019 doi:10.2478/enr-2019-0002 One-month of high-intensity exercise did not change the food intake and the hypothalamic arcuate nucleus proopiomelanocortin and neuropeptide Y expression levels in male Wistar rats Nazli Khajehnasiri1, Homayoun Khazali2, Farzam Sheikhzadeh3, Mahnaz Ghowsi4 1Department of Biological Sciences, Faculty of Basic Sciences, Higher Education Institute of Rab-Rashid, Tabriz, Iran; 2Department of Animal Sciences and Biotechnology, Faculty of Biological Sciences and Technology, Shahid Beheshti University, Tehran, Iran; 3Department of Animal Sciences, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran; 4Department of Biology, Faculty of Sciences, Razi University, Kermanshah, Iran E-mail: [email protected] Objective. The hypothalamic arcuate nucleus proopiomelanocortin (POMC) and neuropeptide Y (NPY) circuitries are involved in the inhibition and stimulation of the appetite, respectively. The aim of this study was to investigate the effects of one-month lasting high-intensity exercise on the POMC mRNA and NPY mRNA expression in the above-mentioned brain structure and appetite and food intake levels. Methods. Fourteen male Wistar rats (250±50 g) were used and kept in the well-controlled con- ditions (22±2 °C, 50±5% humidity, and 12 h dark/light cycle) with food and water ad libitum. The rats were divided into two groups (n=7): 1) control group (C, these rats served as controls) and 2) exercised group (RIE, these rats performed a high-intensity exercise for one month (5 days per week) 40 min daily with speed 35 m/min.
    [Show full text]
  • CCK-8S) of Protein Phosphorylation in the Neostriatum (Forskonln/N-Methyl-D-Aspartic Acid/Glutamate) GRETCHEN L
    Proc. Natl. Acad. Sci. USA Vol. 90, pp. 11277-11281, December 1993 Neurobiology Regulation by the neuropeptide cholecystokinin (CCK-8S) of protein phosphorylation in the neostriatum (forskonln/N-methyl-D-aspartic acid/glutamate) GRETCHEN L. SNYDER*, GILBERTO FISONE*, PATRIZIA MORINOt, VIDAR GUNDERSEN*, OLE PETTER OTTERSEN*, TOMAS HOKFELTt, AND PAUL GREENGARD*§ *Laboratory of Molecular and Cellular Neuroscience, Rockefeller University, New York, NY 10021; tDepartment of Histology and Neurobiology, Karolinska Institute, S-10401, Stockholm, Sweden; and *Department of Anatomy, University of Oslo, Blindern, N-0317 Oslo, Norway Contributed by Tomas Hokfelt, August 16, 1993 ABSTRACT Despite physiological evidence that cholecys- rons, apparently through a mechanism that involves the tokinin (CCK) is an excitatory neurotransmitter in the brain, release of an excitatory neurotransmitter and activation of little is known about its mechanism of action. CCK immuno- NMDA receptors. reactivity in the brain, including projections to the striatum, is primarily attributable to the sulfated octapeptide CCK-8S. We report here that CCK-8S abolishes cAMP-dependent phos- MATERIALS AND METHODS phorylation ofa dopamine- and cAMP-regulated 32-kDa phos- Materials. RPMI 1640 balanced salt solution, bovine serum phoprotein (DARPP-32) in striatal neurons. The effect of albumin, and 3-isobutylmethylxanthine were obtained from CCK-8S is prevented by antagonists of CCKB and N-methyl- Sigma; forskolin was from Calbiochem; NMDA and (+)-MK- D-aspartate receptors. Our results support a model in which 801 hydrogen maleate (MK-801) were from Research Bio- CCK-8S, originating from CCK or CCK/glutamate cortico- chemicals; CCK-8S was from Bachem; CI-988 was from J. striatal neurons, promotes the release of an excitatory neuro- Hughes; cAMP, RIA, and ECL Western blotting detection transmitter that causes the dephosphorylation and inactivation kits were from Amersham; and goat anti-mouse horseradish of DARPP-32, a potent protein phosphatase inhibitor, thereby peroxidase-linked antibody was from Pierce.
    [Show full text]
  • Galanin Stimulates Cortisol Secretion from Human Adrenocortical Cells
    859-864 9/11/07 11:36 Page 859 INTERNATIONAL JOURNAL OF MOLECULAR MEDICINE 20: 859-864, 2007 859 Galanin stimulates cortisol secretion from human adrenocortical cells through the activation of galanin receptor subtype 1 coupled to the adenylate cyclase-dependent signaling cascade ANNA S. BELLONI1, LUDWIK K. MALENDOWICZ2, MARCIN RUCINSKI2, DIEGO GUIDOLIN1 and GASTONE G. NUSSDORFER1 1Department of Human Anatomy and Physiology, School of Medicine, University of Padua, I-35121 Padua, Italy; 2Department of Histology and Embryology, Poznan School of Medicine, PL-60781 Poznan, Poland Received September 10, 2007; Accepted October 5, 2007 Abstract. Previous studies showed that galanin receptors are Introduction expressed in the rat adrenal, and galanin modulates gluco- corticoid secretion in this species. Hence, we investigated the Galanin is a regulatory peptide (30 amino acid residues in expression of the various galanin receptor subtypes (GAL-R1, humans) originally isolated from pig intestine (1) which is GAL-R2 and GAL-R3) in the human adrenocortical cells, and widely distributed in the central and peripheral nervous the possible involvement of galanin in the control of cortisol system, where it acts as a neurotransmitter/neuromodulator. secretion. Reverse transcription-polymerase chain reaction In the gut, galanin modulates insulin release and intestine detected the expression of GAL-R1 (but not GAL-R2 and contractility (2,3). Galanin acts through three distinct subtypes GAL-R3) in the inner zones of the human adrenal cortex. The of G protein-coupled receptors, referred to as GAL-R1, GAL-R2 galanin concentration dependently enhanced basal, but not and GAL-R3 (4). ACTH-stimulated secretion of cortisol from dispersed inner Evidence suggests that galanin is involved in the functional adrenocortical cells (maximal effective concentration, 10-8 M).
    [Show full text]
  • Characterization of a High-Affinity Galanin Receptor in the Rat
    Proc. Natl. Acad. Sci. USA Vol. 90, pp. 4231-4235, May 1993 Neurobiology Characterization of a high-affinity galanin receptor in the rat anterior pituitary: Absence of biological effect and reduced membrane binding of the antagonist M15 differentiate it from the brain/gut receptor (galanin fragment/hemolytic plaque technique/prolactin) DAVID WYNICK*, DAVID M. SMITH*, MOHAMMAD GHATEI*, KAREN AKINSANYA*, RANJEV BHOGAL*, PAUL PURKISSt, PETER BYFIELDt, NOBORU YANAIHARAt, AND STEPHEN R. BLOOM* *Department of Medicine, Hammersmith Hospital, London W12 ONN, United Kingdom; tHaemostasis Research Group, Clinical Research Centre, Harrow, Middlesex, HAl 3UJ, United Kingdom; and tDepartment of Bio-organic Chemistry, University of Shizuoka, Shizuoka, Japan Communicated by L. L. Iversen, December 30, 1992 (received for review November 24, 1992) ABSTRACT Structure-activity studies demonstrate that anterior pituitary, where it has been shown to be estrogen galanin fragments 1-15 and 2-29 are fully active, whereas inducible (5). fragment 3-29 has been reported to be inactive, in a number Various studies have demonstrated effects of galanin on ofdifferent in vivo models. M15, a chimeric peptide comprising basal and stimulated release of prolactin (6, 7), growth galanin 1-13 and substance P 5-11, has recently been found to hormone (8-11), and luteinizing hormone (12, 13) either from be a potent galanin antagonist. Direct effects of galanin at the dispersed pituitary cells or at the hypothalamic level modu- level of the pituitary have been defined, yet, paradoxically, a lating dopamine, somatostatin (SRIF; somatotropin release- number of studies have been unable to demonstrate galanin inhibiting factor), and gonadotropin-releasing hormone binding to an anterior pituitary receptor.
    [Show full text]
  • Dietary Fat Alters the Response of Hypothalamic Neuropeptide Y to Subsequent Energy Intake in Broiler Chickens Xiao J
    © 2017. Published by The Company of Biologists Ltd | Journal of Experimental Biology (2017) 220, 607-614 doi:10.1242/jeb.143792 RESEARCH ARTICLE Dietary fat alters the response of hypothalamic neuropeptide Y to subsequent energy intake in broiler chickens Xiao J. Wang, Shao H. Xu, Lei Liu, Zhi G. Song, Hong C. Jiao and Hai Lin* ABSTRACT Several studies have shown that these hypothalamic neuropeptides Dietary fat affects appetite and appetite-related peptides in birds and are sensitive to body energy status and critical to whole-body mammals; however, the effect of dietary fat on appetite is still unclear metabolic adjustment. NPY reduces energy expenditure by in chickens faced with different energy statuses. Two experiments decreasing adipose tissue thermogenesis (Egawa et al., 1991; Patel were conducted to investigate the effects of dietary fat on food intake et al., 2006; Chao et al., 2011; Zhang et al., 2014). The obesity of and hypothalamic neuropeptides in chickens subjected to two ob/ob mice is attenuated when NPY is knocked out (Erickson et al., fi feeding states or two diets. In Experiment 1, chickens were fed a 1996; Segal-Lieberman et al., 2003). NPY de ciency attenuates high-fat (HF) or low-fat (LF) diet for 35 days, and then subjected to fed responses to a palatable high fat diet in mice (Hollopeter et al., 1998; (HF-fed, LF-fed) or fasted (HF-fasted, LF-fasted) conditions for 24 h. Patel et al., 2006). Animals in which POMC neurons have been In Experiment 2, chickens that were fed a HF or LF diet for 35 days knocked out are obese and hyperphagic (Butler and Cone, 2002; were fasted for 24 h and then re-fed with HF (HF-RHF, LF-RHF) or LF Mencarelli et al., 2012; Diané et al., 2014; Raffan et al., 2016).
    [Show full text]
  • Neurotransmitter and Neuropeptide Regulation of Mast Cell Function
    Xu et al. Journal of Neuroinflammation (2020) 17:356 https://doi.org/10.1186/s12974-020-02029-3 REVIEW Open Access Neurotransmitter and neuropeptide regulation of mast cell function: a systematic review Huaping Xu1, Xiaoyun Shi2, Xin Li3, Jiexin Zou4, Chunyan Zhou5, Wenfeng Liu5, Huming Shao5, Hongbing Chen5 and Linbo Shi4* Abstract The existence of the neural control of mast cell functions has long been proposed. Mast cells (MCs) are localized in association with the peripheral nervous system (PNS) and the brain, where they are closely aligned, anatomically and functionally, with neurons and neuronal processes throughout the body. They express receptors for and are regulated by various neurotransmitters, neuropeptides, and other neuromodulators. Consequently, modulation provided by these neurotransmitters and neuromodulators allows neural control of MC functions and involvement in the pathogenesis of mast cell–related disease states. Recently, the roles of individual neurotransmitters and neuropeptides in regulating mast cell actions have been investigated extensively. This review offers a systematic review of recent advances in our understanding of the contributions of neurotransmitters and neuropeptides to mast cell activation and the pathological implications of this regulation on mast cell–related disease states, though the full extent to which such control influences health and disease is still unclear, and a complete understanding of the mechanisms underlying the control is lacking. Future validation of animal and in vitro models also is needed, which incorporates the integration of microenvironment-specific influences and the complex, multifaceted cross-talk between mast cells and various neural signals. Moreover, new biological agents directed against neurotransmitter receptors on mast cells that can be used for therapeutic intervention need to be more specific, which will reduce their ability to support inflammatory responses and enhance their potential roles in protecting against mast cell–related pathogenesis.
    [Show full text]
  • Differential Gene Expression in Oligodendrocyte Progenitor Cells, Oligodendrocytes and Type II Astrocytes
    Tohoku J. Exp. Med., 2011,Differential 223, 161-176 Gene Expression in OPCs, Oligodendrocytes and Type II Astrocytes 161 Differential Gene Expression in Oligodendrocyte Progenitor Cells, Oligodendrocytes and Type II Astrocytes Jian-Guo Hu,1,2,* Yan-Xia Wang,3,* Jian-Sheng Zhou,2 Chang-Jie Chen,4 Feng-Chao Wang,1 Xing-Wu Li1 and He-Zuo Lü1,2 1Department of Clinical Laboratory Science, The First Affiliated Hospital of Bengbu Medical College, Bengbu, P.R. China 2Anhui Key Laboratory of Tissue Transplantation, Bengbu Medical College, Bengbu, P.R. China 3Department of Neurobiology, Shanghai Jiaotong University School of Medicine, Shanghai, P.R. China 4Department of Laboratory Medicine, Bengbu Medical College, Bengbu, P.R. China Oligodendrocyte precursor cells (OPCs) are bipotential progenitor cells that can differentiate into myelin-forming oligodendrocytes or functionally undetermined type II astrocytes. Transplantation of OPCs is an attractive therapy for demyelinating diseases. However, due to their bipotential differentiation potential, the majority of OPCs differentiate into astrocytes at transplanted sites. It is therefore important to understand the molecular mechanisms that regulate the transition from OPCs to oligodendrocytes or astrocytes. In this study, we isolated OPCs from the spinal cords of rat embryos (16 days old) and induced them to differentiate into oligodendrocytes or type II astrocytes in the absence or presence of 10% fetal bovine serum, respectively. RNAs were extracted from each cell population and hybridized to GeneChip with 28,700 rat genes. Using the criterion of fold change > 4 in the expression level, we identified 83 genes that were up-regulated and 89 genes that were down-regulated in oligodendrocytes, and 92 genes that were up-regulated and 86 that were down-regulated in type II astrocytes compared with OPCs.
    [Show full text]
  • I AMYLIN MEDIATES BRAINSTEM
    AMYLIN MEDIATES BRAINSTEM CONTROL OF HEART RATE IN THE DIVING REFLEX A Dissertation Submitted to The Temple University Graduate Board In Partial Fulfillment of the Requirements for the Degree of Doctor of Philosophy By Fan Yang May, 2012 Examination committee members: Dr. Nae J Dun (advisor), Dept. of Pharmacology, Temple University Dr. Alan Cowan, Dept. of Pharmacology, Temple University Dr. Lee-Yuan Liu-Chen, Dept. of Pharmacology, Temple University Dr. Gabriela Cristina Brailoiu, Dept. of Pharmacology, Temple University Dr. Parkson Lee-Gau Chong, Dept. of Biochemistry, Temple University Dr. Hreday Sapru (external examiner), Depts. of Neurosciences, Neurosurgery & Pharmacology/Physiology, UMDNJ-NJMS. i © 2012 By Fan Yang All Rights Reserved ii ABSTRACT AMYLIN’S ROLE AS A NEUROPEPTIDE IN THE BRAINSTEM Fan Yang Doctor of Philosophy Temple University, 2012 Doctoral Advisory Committee Chair: Nae J Dun, Ph.D. Amylin, or islet amyloid polypeptide is a 37-amino acid member of the calcitonin peptide family. Amylin role in the brainstem and its function in regulating heart rates is unknown. The diving reflex is a powerful autonomic reflex, however no neuropeptides have been described to modulate its function. In this thesis study, amylin expression in the brainstem involving pathways between the trigeminal ganglion and the nucleus ambiguus was visualized and characterized using immunohistochemistry. Its functional role in slowing heart rate and also its involvement in the diving reflex were elucidated using stereotaxic microinjection, whole-cel patch-clamp, and a rat diving model. Immunohistochemical and tract tracing studies in rats revealed amylin expression in trigeminal ganglion cells, which also contained vesicular glutamate transporter 2 positive.
    [Show full text]
  • A G-Protein-Coupled Receptor Mediates Neuropeptide-Induced
    bioRxiv preprint doi: https://doi.org/10.1101/801225; this version posted October 10, 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. A G-protein-coupled receptor mediates neuropeptide-induced oocyte maturation in the jellyfish Clytia Gonzalo Quiroga Artigas1#, Pascal Lapébie1, Lucas Leclère1, Philip Bauknecht 2, Julie Uveira1, Sandra Chevalier1, Gáspár Jékely2,3, Tsuyoshi Momose1 and Evelyn Houliston1* 1. Sorbonne University, CNRS, Villefranche-sur-mer Developmental Biology Laboratory (LBDV), 06230 Villefranche-sur-mer, France 2. Max Planck Institute for Developmental Biology, Spemannstraße 35, 72076 Tübingen, Germany. 3. Living Systems Institute, University of Exeter, Stocker Road, EX4 4QD, Exeter, UK # current address: GQA: The Whitney Laboratory for Marine Bioscience, University of Florida, St. Augustine, FL, USA *Corresponding author: E. Houliston [email protected] Short title: The Clytia oocyte maturation hormone receptor 1 bioRxiv preprint doi: https://doi.org/10.1101/801225; this version posted October 10, 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. Abstract The reproductive hormones that trigger oocyte meiotic maturation and release from the ovary vary greatly between animal species. Identification of receptors for these Maturation Inducing Hormones (MIHs), and understanding how they initiate the largely conserved maturation process, remain important challenges.
    [Show full text]
  • Nitric Oxide As a Second Messenger in Parathyroid Hormone-Related Protein Signaling
    433 Nitric oxide as a second messenger in parathyroid hormone-related protein signaling L Kalinowski, L W Dobrucki and T Malinski Department of Chemistry and Biochemistry, Ohio University, Athens, Ohio, USA (Requests for offprints should be addressed to T Malinski, Department of Chemistry and Biochemistry, Ohio University, Biochemistry Research Laboratories 136, Athens, Ohio 45701–2979, USA; Email: [email protected]) (L Kalinowski was on sabbatical leave from the Department of Clinical Biochemistry, Medical University of Gdansk and Laboratory of Cellular and Molecular Nephrology, Medical Research Center of the Polish Academy of Science, Poland) Abstract Parathyroid hormone (PTH)-related protein (PTHrP) is competitive PTH/PTHrP receptor antagonists, 10 µmol/l produced in smooth muscles and endothelial cells and [Leu11,-Trp12]-hPTHrP(7–34)amide and 10 µmol/l is believed to participate in the local regulation of vascu- [Nle8,18,Tyr34]-bPTH(3–34)amide, were equipotent in lar tone. No direct evidence for the activation of antagonizing hPTH(1–34)-stimulated NO release; endothelium-derived nitric oxide (NO) signaling pathway [Leu11,-Trp12]-hPTHrP(7–34)amide was more potent by PTHrP has been found despite attempts to identify it. than [Nle8,18,Tyr34]-bPTH(3–34)amide in inhibiting Based on direct in situ measurements, it is reported here for hPTHrP(1–34)-stimulated NO release. The PKC inhibi- the first time that the human PTH/PTHrP receptor tor, H-7 (50 µmol/l), did not change hPTH(1–34)- and analogs, hPTH(1–34) and hPTHrP(1–34), stimulate NO hPTHrP(1–34)-stimulated NO release, whereas the release from a single endothelial cell.
    [Show full text]