G Protein-Coupled Receptors at the Crossroad Between Physiologic

Total Page:16

File Type:pdf, Size:1020Kb

G Protein-Coupled Receptors at the Crossroad Between Physiologic G protein-coupled receptors at the crossroad between physiologic and pathologic angiogenesis : old paradigms and emerging concepts De Francesco, EM, Sotgia, F, Clarke, RB, Lisanti, MP and Maggiolini, M http://dx.doi.org/10.3390/ijms18122713 Title G protein-coupled receptors at the crossroad between physiologic and pathologic angiogenesis : old paradigms and emerging concepts Authors De Francesco, EM, Sotgia, F, Clarke, RB, Lisanti, MP and Maggiolini, M Type Article URL This version is available at: http://usir.salford.ac.uk/id/eprint/44805/ Published Date 2017 USIR is a digital collection of the research output of the University of Salford. Where copyright permits, full text material held in the repository is made freely available online and can be read, downloaded and copied for non-commercial private study or research purposes. Please check the manuscript for any further copyright restrictions. For more information, including our policy and submission procedure, please contact the Repository Team at: [email protected]. International Journal of Molecular Sciences Review G Protein-Coupled Receptors at the Crossroad between Physiologic and Pathologic Angiogenesis: Old Paradigms and Emerging Concepts Ernestina M. De Francesco 1,2, Federica Sotgia 3, Robert B. Clarke 2, Michael P. Lisanti 3 and Marcello Maggiolini 1,* ID 1 Department of Pharmacy, Health and Nutrition Sciences, University of Calabria via Savinio, 87036 Rende, Italy; [email protected] 2 Breast Cancer Now Research Unit, Division of Cancer Sciences, Manchester Cancer Research Centre, University of Manchester, Wilmslow Road, Manchester M20 4GJ, UK; [email protected] 3 Translational Medicine, School of Environment and Life Sciences, Biomedical Research Centre, University of Salford, Greater Manchester M5 4WT, UK; [email protected] (F.S.); [email protected] (M.P.L.) * Correspondence: [email protected]; Tel.: +39-0984-493076 Received: 30 October 2017; Accepted: 11 December 2017; Published: 14 December 2017 Abstract: G protein-coupled receptors (GPCRs) have been implicated in transmitting signals across the extra- and intra-cellular compartments, thus allowing environmental stimuli to elicit critical biological responses. As GPCRs can be activated by an extensive range of factors including hormones, neurotransmitters, phospholipids and other stimuli, their involvement in a plethora of physiological functions is not surprising. Aberrant GPCR signaling has been regarded as a major contributor to diverse pathologic conditions, such as inflammatory, cardiovascular and neoplastic diseases. In this regard, solid tumors have been demonstrated to activate an angiogenic program that relies on GPCR action to support cancer growth and metastatic dissemination. Therefore, the manipulation of aberrant GPCR signaling could represent a promising target in anticancer therapy. Here, we highlight the GPCR-mediated angiogenic function focusing on the molecular mechanisms and transduction effectors driving the patho-physiological vasculogenesis. Specifically, we describe evidence for the role of heptahelic receptors and associated G proteins in promoting angiogenic responses in pathologic conditions, especially tumor angiogenesis and progression. Likewise, we discuss opportunities to manipulate aberrant GPCR-mediated angiogenic signaling for therapeutic benefit using innovative GPCR-targeted and patient-tailored pharmacological strategies. Keywords: GPCR; tumor angiogenesis; tumor microenvironment; VEGF; HIF-1; GPER; SDF-1; sphingosine-1P 1. Introduction Over the past decade, the discovery and study of G-protein coupled receptors (GPCRs) has unveiled novel molecular mechanisms through which extracellular signals promote changes in cell functions. Encoded by more than 900 genes in the human genome, GPCRs belong to one of the largest families of membrane proteins involved in the detection of a wide panel of extracellular stimuli, including photons and ions, as well as peptides, proteins, hormones and phospholipids [1,2]. Members belonging to the GPCR superfamily regulate a broad spectrum of physiological functions, such as vision, smell, and taste, as well as neurological, cardiovascular, endocrine, and reproductive functions [1–3]. GPCRs signal through their association with G-proteins, which are membrane bound heterotrimers consisting of a guanine diphosphate (GDP)-bound α subunit with GTPase activity, Int. J. Mol. Sci. 2017, 18, 2713; doi:10.3390/ijms18122713 www.mdpi.com/journal/ijms Int. J. Mol. Sci. 2017, 18, 2713 2 of 26 Int. J. Mol. Sci. 2017, 18, 2713 2 of 27 GPCRs signal through their association with G-proteins, which are membrane bound heterotrimers consisting of a guanine diphosphate (GDP)-bound α subunit with GTPase activity, βγ α and a Gβγ functional monomer [[1–3].1–3]. Based on the sequence of the α subunit, G-proteins are defineddefined α α α α inin fourfour classes classes (G (Gs,αs, G Gq,α Gq, Gi,α andi, and G 12), Gα each12), each coupling coupling to more to thanmore one than receptor one receptor subtypes, subtypes, although withalthough different with affinitydifferent [2 ].affinity GPCRs [2]. are GPCRs classified are classified based on theirbased phylogenetic on their phylogenetic origin and origin sequence and homologysequence homology (Figure1). (Figure 1). Figure 1.1.G-protein G-protein coupled coupled receptors receptors (GPCRs) (GPCRs) classification. classification. The International The International Union of Pharmacology Union of (IUPHAR)Pharmacology classification (IUPHAR) (left classification column) applies (left tocolumn both vertebrates) applies to and both invertebrates vertebrates (Classand invertebrates D and E are unique(Class D to and invertebrates). E are unique The to invertebrates). GRAPH system The (middle GRAPH column) system applies (middle specifically column) applies to vertebrates. specifically to vertebrates. Typically, uponupon ligandligand binding,binding, thethe receptorreceptor undergoesundergoes aa conformationalconformational changechange transmitted to thethe heterotrimer, whose whose tri-dimensional tri-dimensional rearrangem rearrangementent determines determines the the exchange exchange of ofGTP GTP for for GDP GDP on α βγ onthe theα subunitsubunit and and the thedissociation dissociation of the of theGβγ G monomer.monomer. Overlapping Overlapping with with GTP GTP recruitment, recruitment, the α βγ thetransition transition from from“off” “off”to “on” to state “on” allows state the allows Gα and the G βγ andsubunit G tosubunit interact towith interact and regulate with and the regulateactivity of the a activity vast repertoire of a vast of repertoire transduction of transduction effectors, effectors,such as calcium, such as calcium,adenylyl adenylylcyclase, cyclase,phospholipase phospholipase C, phosphodiesterases, C, phosphodiesterases, and protein and kinases protein [4,5]. kinases These [4,5 ].second These messengers second messengers activate activateadditional additional intracellular intracellular pathways pathways such suchas the as themitogen-activated mitogen-activated protein protein kinase kinase (MAPK), phosphoinositide-3 kinase (PI3K)-Akt, small GTP-bindingGTP-binding proteins (Ras and Rho GTPases),GTPases), whichwhich ultimately engageengage transcription transcription factors, factors, leading leading to geneto gene expression expression changes changes [5,6]. [5,6]. Additionally, Additionally, many sevenmany trans-membraneseven trans-membrane receptors receptors (7TMRs) (7TMRs) are orphan are GPCRsorphan withGPCRs unidentified with unidentified ligands and ligands potential and ligand-independentpotential ligand-independent properties, properties, suggesting suggesting that GPCR-mediated that GPCR-mediated action may action include may unexpected include biologicalunexpected functions, biological conceivably functions, unrelated conceivably with unre the classicallated with paradigm the classical of a signal paradigm conveyed of a through signal theconveyed cell membrane through [7the]. In cell addition membrane to their [7]. participation In addition in to an their extraordinary participation number in an of extraordinary physiological processes,number of some physiological of which yetprocesses, to be defined, some GPCRsof which are yet implicated to be defined, in the onsetGPCRs and are evolution implicated of several in the pathologicalonset and evolution conditions, of includingseveral pathological inflammatory conditions, and degenerative including diseases, inflammatory metabolic and imbalances, degenerative and cancerdiseases, [8– metabolic11]. Not surprisingly, imbalances, the and manipulation cancer [8–11]. of aberrant Not surprisingly, GPCR transduction the mani pathwayspulation of holds aberrant great therapeuticGPCR transduction potential pathways [12]. Indeed, holds GPCR-targeting great therapeuti agents,c potential totaling [12]. almostIndeed, one GPCR-targeting third of all currently agents, marketedtotaling almost drugs, one are third excellent of all candidates currently formarketed the treatment drugs, are of a excellent widespread candidates array of for diseases the treatment [12–14]. of a widespread array of diseases [12–14]. Int. J. Mol. Sci. 2017, 18, 2713 3 of 27 For instance, GPCRs as drug targets to halt the anomalous formation of new blood vessels may be a strategy to normalize the dysregulated vascularization, which distinguishes various ischemic and inflammatory conditions and cancer [15]. Neoangiogenesis is an essential physiological
Recommended publications
  • Sample Lab Report
    3030 Venture Lane, Suite 108 ● Melbourne, Florida 32934 ● Phone 321-253-5197 ● Fax 321-253-5199 PATIENT: DOE, JAMES ACCESSION NO: CLINICIAN/ PATIENT ID: REQUESTING DATE OF BIRTH: 1/5/1986 DOCTOR: GENDER: MALE DATE COLLECTED: 5/26/2015 DATE OF REPORT: 6/15/2015 RESULTS PRIMARY TUMOR TYPE RIGHT TESTICLE BIOLOGICALLY IMPORTANT ONCOGENES DETECTED GENE IMPLICATION TP53 TP53 mutations may be an important driver of tumorigenesis and / or a reason for treatment resistance in a some patients. PTEN Responsible for uncontrolled growth. MDM2 Causes p53 inactivation. Associated with cancer growth and progression. TGFB1 TGFB appears to promote tumor progession by stimulating invasion and metastasis. TUBB2A Microtubules are the key components of the cytoskeleton of eukaryotic cells and have an important role in various cellular functions such as intracellular migration and transport, cell shape maintenance, polarity, cell signaling and mitosis. c-JUN Proto-oncogene PHB2 Prohibitins play a crucial role in adhesion processes in the cell and thereby sustaining cancer cell propagation and survival. Clinical Impression: Low Aggressive Potential Additional Genes Detected: ABCG2, ARHGAP5, ATF4, BIRC5, BNIP3, CAPNS1, CARD17, CCNB1, CD24, CDC20, CDK18, CKS2, DCN, DEPDC1, FTL, FZD5, FZD9, GAPDH, GNB2, GPR126, H2AFZ, HDAC1, HMGN2, ID1, IFITM1, JUNB, KPNA2, KRT18, LDHA, LTF, MAD2L1, MAP2K1, MAP2K2, MAP2K4, MAPRE1, MAS1, NME1, NME3, NPM1, PA2G4, PABPC1, PFDN4, PGAM1, PGK1, PHB, PIK3CB, PKM, PPIA, PPIH, PRKX, PRNP, PTMA, RAC1, RAC2, RALBP1, RAP1A, RBBP4, RHOB, RHOC,
    [Show full text]
  • Lysophosphatidic Acid and Its Receptors: Pharmacology and Therapeutic Potential in Atherosclerosis and Vascular Disease
    JPT-107404; No of Pages 13 Pharmacology & Therapeutics xxx (2019) xxx Contents lists available at ScienceDirect Pharmacology & Therapeutics journal homepage: www.elsevier.com/locate/pharmthera Lysophosphatidic acid and its receptors: pharmacology and therapeutic potential in atherosclerosis and vascular disease Ying Zhou a, Peter J. Little a,b, Hang T. Ta a,c, Suowen Xu d, Danielle Kamato a,b,⁎ a School of Pharmacy, University of Queensland, Pharmacy Australia Centre of Excellence, Woolloongabba, QLD 4102, Australia b Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China c Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, St Lucia, QLD 4072, Australia d Aab Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA article info abstract Available online xxxx Lysophosphatidic acid (LPA) is a collective name for a set of bioactive lipid species. Via six widely distributed G protein-coupled receptors (GPCRs), LPA elicits a plethora of biological responses, contributing to inflammation, Keywords: thrombosis and atherosclerosis. There have recently been considerable advances in GPCR signaling especially Lysophosphatidic acid recognition of the extended role for GPCR transactivation of tyrosine and serine/threonine kinase growth factor G-protein coupled receptors receptors. This review covers LPA signaling pathways in the light of new information. The use of transgenic and Atherosclerosis gene knockout animals, gene manipulated cells, pharmacological LPA receptor agonists and antagonists have Gproteins fi β-arrestins provided many insights into the biological signi cance of LPA and individual LPA receptors in the progression Transactivation of atherosclerosis and vascular diseases.
    [Show full text]
  • Edinburgh Research Explorer
    Edinburgh Research Explorer International Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list Citation for published version: Davenport, AP, Alexander, SPH, Sharman, JL, Pawson, AJ, Benson, HE, Monaghan, AE, Liew, WC, Mpamhanga, CP, Bonner, TI, Neubig, RR, Pin, JP, Spedding, M & Harmar, AJ 2013, 'International Union of Basic and Clinical Pharmacology. LXXXVIII. G protein-coupled receptor list: recommendations for new pairings with cognate ligands', Pharmacological reviews, vol. 65, no. 3, pp. 967-86. https://doi.org/10.1124/pr.112.007179 Digital Object Identifier (DOI): 10.1124/pr.112.007179 Link: Link to publication record in Edinburgh Research Explorer Document Version: Publisher's PDF, also known as Version of record Published In: Pharmacological reviews Publisher Rights Statement: U.S. Government work not protected by U.S. copyright General rights Copyright for the publications made accessible via the Edinburgh Research Explorer is retained by the author(s) and / or other copyright owners and it is a condition of accessing these publications that users recognise and abide by the legal requirements associated with these rights. Take down policy The University of Edinburgh has made every reasonable effort to ensure that Edinburgh Research Explorer content complies with UK legislation. If you believe that the public display of this file breaches copyright please contact [email protected] providing details, and we will remove access to the work immediately and investigate your claim. Download date: 02. Oct. 2021 1521-0081/65/3/967–986$25.00 http://dx.doi.org/10.1124/pr.112.007179 PHARMACOLOGICAL REVIEWS Pharmacol Rev 65:967–986, July 2013 U.S.
    [Show full text]
  • Supplementary Data
    Supplemental Data A novel mouse model of X-linked nephrogenic diabetes insipidus: Phenotypic analysis and therapeutic implications Jian Hua Li, Chung-Lin Chou, Bo Li, Oksana Gavrilova, Christoph Eisner, Jürgen Schnermann, Stasia A. Anderson, Chu-Xia Deng, Mark A. Knepper, and Jürgen Wess Supplemental Methods Metabolic cage studies. Animals were maintained in mouse metabolic cages (Hatteras Instruments, Cary, NC) under controlled temperature and light conditions (12 hr light and dark cycles). Mice received a fixed daily ration of 6.5 g of gelled diet per 20 g of body weight per day. The gelled diet was composed of 4 g of Basal Diet 5755 (Test Diet, Richmond, IN), 2.5 ml of deionized water, and 65 mg agar. Preweighted drinking water was provided ad libitum during the course of the study. Mice were acclimated in the metabolic cages for 1-2 days. Urine was collected under mineral oil in preweighted collection vials for successive 24 hr periods. Analysis of GPCR expression in mouse IMCD cells via TaqMan real-time qRT-PCR. Total RNA prepared from mouse IMCD tubule suspensions was reverse transcribed as described under Experimental Procedures. Tissues from ten 10-week old C57BL/6 WT mice were collected and pooled for each individual experiment. cDNA derived from 640 ng of RNA was mixed with an equal volume of TaqMan gene expression 2 x master mix (Applied Biosystems, Foster City, CA). 100 μl-aliquots of this mixture (corresponding to 80 ng of RNA) were added to each of the 8 fill ports of a 384-well plate of a mouse GPCR array panel (Applied Biosystems).
    [Show full text]
  • A Computational Approach for Defining a Signature of Β-Cell Golgi Stress in Diabetes Mellitus
    Page 1 of 781 Diabetes A Computational Approach for Defining a Signature of β-Cell Golgi Stress in Diabetes Mellitus Robert N. Bone1,6,7, Olufunmilola Oyebamiji2, Sayali Talware2, Sharmila Selvaraj2, Preethi Krishnan3,6, Farooq Syed1,6,7, Huanmei Wu2, Carmella Evans-Molina 1,3,4,5,6,7,8* Departments of 1Pediatrics, 3Medicine, 4Anatomy, Cell Biology & Physiology, 5Biochemistry & Molecular Biology, the 6Center for Diabetes & Metabolic Diseases, and the 7Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202; 2Department of BioHealth Informatics, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46202; 8Roudebush VA Medical Center, Indianapolis, IN 46202. *Corresponding Author(s): Carmella Evans-Molina, MD, PhD ([email protected]) Indiana University School of Medicine, 635 Barnhill Drive, MS 2031A, Indianapolis, IN 46202, Telephone: (317) 274-4145, Fax (317) 274-4107 Running Title: Golgi Stress Response in Diabetes Word Count: 4358 Number of Figures: 6 Keywords: Golgi apparatus stress, Islets, β cell, Type 1 diabetes, Type 2 diabetes 1 Diabetes Publish Ahead of Print, published online August 20, 2020 Diabetes Page 2 of 781 ABSTRACT The Golgi apparatus (GA) is an important site of insulin processing and granule maturation, but whether GA organelle dysfunction and GA stress are present in the diabetic β-cell has not been tested. We utilized an informatics-based approach to develop a transcriptional signature of β-cell GA stress using existing RNA sequencing and microarray datasets generated using human islets from donors with diabetes and islets where type 1(T1D) and type 2 diabetes (T2D) had been modeled ex vivo. To narrow our results to GA-specific genes, we applied a filter set of 1,030 genes accepted as GA associated.
    [Show full text]
  • HER2 Gene Signatures: (I) Novel and (Ii) Established by Desmedt Et Al 2008 (31)
    Table S1: HER2 gene signatures: (i) Novel and (ii) Established by Desmedt et al 2008 (31). Pearson R [neratinib] is the correlation with neratinib response using a pharmacogenomic model of breast cancer cell lines (accessed online via CellMinerCDB). *indicates significantly correlated genes. n/a = data not available in CellMinerCDB Genesig N Gene ID Pearson R [neratinib] p-value (i) Novel 20 ERBB2 0.77 1.90E-08* SPDEF 0.45 4.20E-03* TFAP2B 0.2 0.24 CD24 0.38 0.019* SERHL2 0.41 0.0097* CNTNAP2 0.12 0.47 RPL19 0.29 0.073 CAPN13 0.51 1.00E-03* RPL23 0.22 0.18 LRRC26 n/a n/a PRODH 0.42 9.00E-03* GPRC5C 0.44 0.0056* GGCT 0.38 1.90E-02* CLCA2 0.31 5.70E-02 KDM5B 0.33 4.20E-02* SPP1 -0.25 1.30E-01 PHLDA1 -0.54 5.30E-04* C15orf48 0.06 7.10E-01 SUSD3 -0.09 5.90E-01 SERPINA1 0.14 4.10E-01 (ii) Established 24 ERBB2 0.77 1.90E-08* PERLD1 0.77 1.20E-08* PSMD3 0.33 0.04* PNMT 0.33 4.20E-02* GSDML 0.4 1.40E-02* CASC3 0.26 0.11 LASP1 0.32 0.049* WIPF2 0.27 9.70E-02 EPN3 0.42 8.50E-03* PHB 0.38 0.019* CLCA2 0.31 5.70E-02 ORMDL2 0.06 0.74 RAP1GAP 0.53 0.00059* CUEDC1 0.09 0.61 HOXC11 0.2 0.23 CYP2J2 0.45 0.0044* HGD 0.14 0.39 ABCA12 0.07 0.67 ATP2C2 0.42 0.0096* ITGA3 0 0.98 CEACAM5 0.4 0.012* TMEM16K 0.15 0.37 NR1D1 n/a n/a SNX7 -0.28 0.092 FJX1 -0.26 0.12 KCTD9 -0.11 0.53 PCTK3 -0.04 0.83 CREG1 0.17 0.3 Table S2: Up-regulated genes from the top 500 DEGs for each comparison by WAD score METABRIC METABRIC METABRIC TCGA ERBB2amp ERBB2mut oncERBB2mut HER2+ ERBB2 PIP ANKRD30A ERBB2 GRB7 CYP4Z1 CYP4Z1 SCGB2A2 PGAP3 PROM1 LRRC26 SPDEF GSDMB CD24 PPP1R1B FOXA1
    [Show full text]
  • Table 1A SIRT1 Differential Binding Gene List Down
    Rp1 Rb1cc1 Pcmtd1 Mybl1 Sgk3 Cspp1 Arfgef1 Cpa6 Kcnb2 Stau2 Jph1 Paqr8 Kcnq5 Rims1 Smap1 Bai3 Prim2 Bag2 Zfp451 Dst Uggt1 4632411B12Rik Fam178b Tmem131 Inpp4a 2010300C02Rik Rev1 Aff3 Map4k4 Il1r1 Il1rl2 Tgfbrap1 Col3a1 Wdr75 Tmeff2 Hecw2 Boll Plcl1 Satb2 Aox4 Mpp4 Gm973 Carf Nbeal1 Pard3b Ino80d Adam23 Dytn Pikfyve Atic Fn1 Smarcal1 Tns1 Arpc2 Pnkd Ctdsp1 Usp37 Acsl3 Cul3 Dock10 Col4a4 Col4a3 Mff Wdr69 Pid1 Sp110 Sp140 Itm2c 2810459M11Rik Dis3l2 Chrng Gigyf2 Ugt1a7c Ugt1a6b Hjurp A730008H23Rik Trpm8 Kif1a D1Ertd622e Pam Cntnap5b Rnf152 Phlpp1 Clasp1 Gli2 Dpp10 Tmem163 Zranb3 Pfkfb2 Pigr Rbbp5 Sox13 Ppfia4 Rabif Kdm5b Ppp1r12b Lgr6 Pkp1 Kif21b Nr5a2 Dennd1b Trove2 Pdc Hmcn1 Ncf2 Nmnat2 Lamc2 Cacna1e Xpr1 Tdrd5 Fam20b Ralgps2 Sec16b Astn1 Pappa2 Tnr Klhl20 Dnm3 Bat2l2 4921528O07Rik Scyl3 Dpt Mpzl1 Creg1 Cd247 Gm4846 Lmx1a Pbx1 Ddr2 Cd244 Cadm3 Fmn2 Grem2 Rgs7 Fh1 Wdr64 Pld5 Cep170 Akt3 Pppde1 Smyd3 Parp1 Enah Capn8 Susd4 Mosc1 Rrp15 Gpatch2 Esrrg Ptpn14 Smyd2 Tmem206 Nek2 Traf5 Hhat Cdnf Fam107b Camk1d Cugbp2 Gata3 Itih5 Sfmbt2 Itga8 Pter Rsu1 Ptpla Slc39a12 Cacnb2 Nebl Pip4k2a Abi1 Tbpl2 Pnpla7 Kcnt1 Camsap1 Nacc2 Gpsm1 Sec16a Fam163b Vav2 Olfm1 Tsc1 Med27 Rapgef1 Pkn3 Zer1 Prrx2 Gpr107 Ass1 Nup214 Bat2l Dnm1 Ralgps1 Fam125b Mapkap1 Hc Ttll11 Dennd1a Olfml2a Scai Arhgap15 Gtdc1 Mbd5 Kif5c Lypd6b Lypd6 Fmnl2 Arl6ip6 Galnt13 Acvr1 Ccdc148 Dapl1 Tanc1 Ly75 Gcg Kcnh7 Cobll1 Scn3a Scn9a Scn7a Gm1322 Stk39 Abcb11 Slc25a12 Metapl1 Pdk1 Rapgef4 B230120H23Rik Gpr155 Wipf1 Pde11a Prkra Gm14461 Pde1a Calcrl Olfr1033 Mybpc3 F2 Arhgap1 Ambra1 Dgkz Creb3l1
    [Show full text]
  • Synaptamide Activates the Adhesion GPCR GPR110 (ADGRF1) Through GAIN Domain Binding
    ARTICLE https://doi.org/10.1038/s42003-020-0831-6 OPEN Synaptamide activates the adhesion GPCR GPR110 (ADGRF1) through GAIN domain binding Bill X. Huang1, Xin Hu2, Heung-Sun Kwon1, Cheng Fu1, Ji-Won Lee1, Noel Southall2, Juan Marugan2 & ✉ Hee-Yong Kim1 1234567890():,; Adhesion G protein-coupled receptors (aGPCR) are characterized by a large extracellular region containing a conserved GPCR-autoproteolysis-inducing (GAIN) domain. Despite their relevance to several disease conditions, we do not understand the molecular mechanism by which aGPCRs are physiologically activated. GPR110 (ADGRF1) was recently deorphanized as the functional receptor of N-docosahexaenoylethanolamine (synaptamide), a potent synap- togenic metabolite of docosahexaenoic acid. Thus far, synaptamide is the first and only small- molecule endogenous ligand of an aGPCR. Here, we demonstrate the molecular basis of synaptamide-induced activation of GPR110 in living cells. Using in-cell chemical cross-linking/ mass spectrometry, computational modeling and mutagenesis-assisted functional assays, we discover that synaptamide specifically binds to the interface of GPR110 GAIN subdomains through interactions with residues Q511, N512 and Y513, causing an intracellular conforma- tional change near TM6 that triggers downstream signaling. This ligand-induced GAIN-tar- geted activation mechanism provides a framework for understanding the physiological function of aGPCRs and therapeutic targeting in the GAIN domain. 1 Laboratory of Molecular Signaling, National Institute on Alcohol Abuse
    [Show full text]
  • The Adhesion G Protein-Coupled Receptor GPR56 Is a Cell-Autonomous Regulator of Oligodendrocyte Development
    ARTICLE Received 27 May 2014 | Accepted 14 Dec 2014 | Published 21 Jan 2015 DOI: 10.1038/ncomms7121 OPEN The adhesion G protein-coupled receptor GPR56 is a cell-autonomous regulator of oligodendrocyte development Stefanie Giera1,*, Yiyu Deng1,*,w, Rong Luo1,*, Sarah D. Ackerman2, Amit Mogha2, Kelly R. Monk2,3, Yanqin Ying1, Sung-Jin Jeong1,w, Manabu Makinodan4,5, Allison R. Bialas4,5, Bernard S. Chang6, Beth Stevens4,5, Gabriel Corfas4,5,w & Xianhua Piao1 Mutations in GPR56, a member of the adhesion G protein-coupled receptor family, cause a human brain malformation called bilateral frontoparietal polymicrogyria (BFPP). Magnetic resonance imaging (MRI) of BFPP brains reveals myelination defects in addition to brain malformation. However, the cellular role of GPR56 in oligodendrocyte development remains unknown. Here, we demonstrate that loss of Gpr56 leads to hypomyelination of the central nervous system in mice. GPR56 levels are abundant throughout early stages of oligodendrocyte development, but are downregulated in myelinating oligodendrocytes. Gpr56-knockout mice manifest with decreased oligodendrocyte precursor cell (OPC) proliferation and diminished levels of active RhoA, leading to fewer mature oligodendrocytes and a reduced number of myelinated axons in the corpus callosum and optic nerves. Conditional ablation of Gpr56 in OPCs leads to a reduced number of mature oligodendrocytes as seen in constitutive knockout of Gpr56. Together, our data define GPR56 as a cell-autonomous regulator of oligodendrocyte development. 1 Division of Newborn Medicine, Department of Medicine, Boston Children’s Hospital and Harvard Medical School, Boston, Massachusetts 02115, USA. 2 Department of Developmental Biology, Washington University School of Medicine, St Louis, Missouri 63110, USA.
    [Show full text]
  • G Protein-Coupled Receptors
    S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2015/16: G protein-coupled receptors. British Journal of Pharmacology (2015) 172, 5744–5869 THE CONCISE GUIDE TO PHARMACOLOGY 2015/16: G protein-coupled receptors Stephen PH Alexander1, Anthony P Davenport2, Eamonn Kelly3, Neil Marrion3, John A Peters4, Helen E Benson5, Elena Faccenda5, Adam J Pawson5, Joanna L Sharman5, Christopher Southan5, Jamie A Davies5 and CGTP Collaborators 1School of Biomedical Sciences, University of Nottingham Medical School, Nottingham, NG7 2UH, UK, 2Clinical Pharmacology Unit, University of Cambridge, Cambridge, CB2 0QQ, UK, 3School of Physiology and Pharmacology, University of Bristol, Bristol, BS8 1TD, UK, 4Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK, 5Centre for Integrative Physiology, University of Edinburgh, Edinburgh, EH8 9XD, UK Abstract The Concise Guide to PHARMACOLOGY 2015/16 provides concise overviews of the key properties of over 1750 human drug targets with their pharmacology, plus links to an open access knowledgebase of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. The full contents can be found at http://onlinelibrary.wiley.com/doi/ 10.1111/bph.13348/full. G protein-coupled receptors are one of the eight major pharmacological targets into which the Guide is divided, with the others being: ligand-gated ion channels, voltage-gated ion channels, other ion channels, nuclear hormone receptors, catalytic receptors, enzymes and transporters. These are presented with nomenclature guidance and summary information on the best available pharmacological tools, alongside key references and suggestions for further reading.
    [Show full text]
  • G Protein‐Coupled Receptors
    S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2019/20: G protein-coupled receptors. British Journal of Pharmacology (2019) 176, S21–S141 THE CONCISE GUIDE TO PHARMACOLOGY 2019/20: G protein-coupled receptors Stephen PH Alexander1 , Arthur Christopoulos2 , Anthony P Davenport3 , Eamonn Kelly4, Alistair Mathie5 , John A Peters6 , Emma L Veale5 ,JaneFArmstrong7 , Elena Faccenda7 ,SimonDHarding7 ,AdamJPawson7 , Joanna L Sharman7 , Christopher Southan7 , Jamie A Davies7 and CGTP Collaborators 1School of Life Sciences, University of Nottingham Medical School, Nottingham, NG7 2UH, UK 2Monash Institute of Pharmaceutical Sciences and Department of Pharmacology, Monash University, Parkville, Victoria 3052, Australia 3Clinical Pharmacology Unit, University of Cambridge, Cambridge, CB2 0QQ, UK 4School of Physiology, Pharmacology and Neuroscience, University of Bristol, Bristol, BS8 1TD, UK 5Medway School of Pharmacy, The Universities of Greenwich and Kent at Medway, Anson Building, Central Avenue, Chatham Maritime, Chatham, Kent, ME4 4TB, UK 6Neuroscience Division, Medical Education Institute, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, UK 7Centre for Discovery Brain Sciences, University of Edinburgh, Edinburgh, EH8 9XD, UK Abstract The Concise Guide to PHARMACOLOGY 2019/20 is the fourth in this series of biennial publications. The Concise Guide provides concise overviews of the key properties of nearly 1800 human drug targets with an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (www.guidetopharmacology.org), which provides more detailed views of target and ligand properties. Although the Concise Guide represents approximately 400 pages, the material presented is substantially reduced compared to information and links presented on the website.
    [Show full text]
  • Gpr126 Functions in Schwann Cells to Control Differentiation and Myelination Via G-Protein Activation Amit Mogha Washington University School of Medicine in St
    Washington University School of Medicine Digital Commons@Becker Open Access Publications 11-2013 Gpr126 functions in schwann cells to control differentiation and myelination via G-protein activation Amit Mogha Washington University School of Medicine in St. Louis Andrew E. Benesh Washington University School of Medicine in St. Louis Chinmoy Patra Max Planck Institute for Heart and Lung Research Felix B. Engel University of Erlangen-Nurnberg Torsten Schoneberg University of Leipzig See next page for additional authors Follow this and additional works at: https://digitalcommons.wustl.edu/open_access_pubs Recommended Citation Mogha, Amit; Benesh, Andrew E.; Patra, Chinmoy; Engel, Felix B.; Schoneberg, Torsten; Liebscher, Ines; and Monk, Kelly R., ,"Gpr126 functions in schwann cells to control differentiation and myelination via G-protein activation." The ourJ nal of Neuroscience.33,46. 17976-17985. (2013). https://digitalcommons.wustl.edu/open_access_pubs/1894 This Open Access Publication is brought to you for free and open access by Digital Commons@Becker. It has been accepted for inclusion in Open Access Publications by an authorized administrator of Digital Commons@Becker. For more information, please contact [email protected]. Authors Amit Mogha, Andrew E. Benesh, Chinmoy Patra, Felix B. Engel, Torsten Schoneberg, Ines Liebscher, and Kelly R. Monk This open access publication is available at Digital Commons@Becker: https://digitalcommons.wustl.edu/open_access_pubs/1894 17976 • The Journal of Neuroscience, November 13, 2013 • 33(46):17976–17985 Development/Plasticity/Repair Gpr126 Functions in Schwann Cells to Control Differentiation and Myelination via G-Protein Activation Amit Mogha,1 Andrew E. Benesh,1 Chinmoy Patra,3 Felix B. Engel,3,4 Torsten Scho¨neberg,5 Ines Liebscher,5 and Kelly R.
    [Show full text]