G Protein-Coupled Receptors at the Crossroad Between Physiologic and Pathologic Angiogenesis: Old Paradigms and Emerging Concepts

Total Page:16

File Type:pdf, Size:1020Kb

G Protein-Coupled Receptors at the Crossroad Between Physiologic and Pathologic Angiogenesis: Old Paradigms and Emerging Concepts 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 and developmental process tightly regulated by a highly coordinated balance between pro- and anti-angiogenic factors. Alterations in this fine-tuned equilibrium could arguably lead to the increased production of pro-angiogenic mediators and the loss of inhibitory factors, which ultimately drive an aberrant vascularization [16–18]. For example, during cancer progression, an excessive vascular growth with abnormal vessel remodeling and tortuosity is actively stimulated by the release in the tumor microenvironment of angiogenic mediators such as vascular endothelial growth factor (VEGF) [19–21]. Several 7TMRs are reported to trigger VEGF release to boost the formation of new blood vessels, particularly during cancer evolution toward an aggressive and metastatic cancer phenotype [22,23], suggesting that GPCRs may serve as novel drug candidates in combination therapies aimed at combatting tumor angiogenesis. In addition to their role in the production of angiogenic factors, 7TMRs also function as receptor for angiogenic ligands,
Recommended publications
  • Molecular Signatures of G-Protein-Coupled Receptors A
    REVIEW doi:10.1038/nature11896 Molecular signatures of G-protein-coupled receptors A. J. Venkatakrishnan1, Xavier Deupi2, Guillaume Lebon1,3,4,5, Christopher G. Tate1, Gebhard F. Schertler2,6 & M. Madan Babu1 G-protein-coupled receptors (GPCRs) are physiologically important membrane proteins that sense signalling molecules such as hormones and neurotransmitters, and are the targets of several prescribed drugs. Recent exciting developments are providing unprecedented insights into the structure and function of several medically important GPCRs. Here, through a systematic analysis of high-resolution GPCR structures, we uncover a conserved network of non-covalent contacts that defines the GPCR fold. Furthermore, our comparative analysis reveals characteristic features of ligand binding and conformational changes during receptor activation. A holistic understanding that integrates molecular and systems biology of GPCRs holds promise for new therapeutics and personalized medicine. ignal transduction is a fundamental biological process that is comprehensively, and in the process expand the current frontiers of required to maintain cellular homeostasis and to ensure coordi- GPCR biology. S nated cellular activity in all organisms. Membrane proteins at the In this analysis, we objectively compare known structures and reveal cell surface serve as the communication interface between the cell’s key similarities and differences among diverse GPCRs. We identify a external and internal environments. One of the largest and most diverse consensus structural scaffold of GPCRs that is constituted by a network membrane protein families is the GPCRs, which are encoded by more of non-covalent contacts between residues on the transmembrane (TM) than 800 genes in the human genome1. GPCRs function by detecting a helices.
    [Show full text]
  • Endothelial-Protective Effects of a G-Protein-Biased Sphingosine-1 Phosphate Receptor-1 Agonist, SAR247799, in Type-2 Diabetes R
    medRxiv preprint doi: https://doi.org/10.1101/2020.05.15.20103101; this version posted May 20, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted medRxiv a license to display the preprint in perpetuity. All rights reserved. No reuse allowed without permission. 1 Endothelial-protective effects of a G-protein-biased sphingosine-1 phosphate receptor-1 agonist, SAR247799, in type-2 diabetes rats and a randomized placebo-controlled patient trial. Luc Bergougnan1, Grit Andersen2, Leona Plum-Mörschel3, Maria Francesca Evaristi1, Bruno Poirier1, Agnes Tardat4, Marcel Ermer3, Theresa Herbrand2, Jorge Arrubla2, Hans Veit Coester2, Roberto Sansone5, Christian Heiss6, Olivier Vitse4, Fabrice Hurbin4, Rania Boiron1, Xavier Benain4, David Radzik1, Philip Janiak1, Anthony J Muslin7, Lionel Hovsepian1, Stephane Kirkesseli1, Paul Deutsch8, Ashfaq A Parkar8 1 Sanofi R&D, 1 Avenue Pierre Brossolette, 91385 Chilly Mazarin, France; 2 Profil Institut für Stoffwechselforschung GmbH, Hellersbergstraße 9, 41460 Neuss, Germany; 3 Profil Mainz GmbH & Co. KG, Malakoff-Passage, Rheinstraße 4C, Eingang via Templerstraße, D-55116 Mainz, Germany; 4 Sanofi R&D, 371 Rue du Professeur Blayac, 34080 Montpellier, France; 5 University Hospital Düsseldorf, Division of Cardiology, Pulmonary diseases and Vascular medicine, 40225 Düsseldorf, Germany; 6 Department of Clinical and Experimental Medicine, University of Surrey, Stag Hill, Guildford GU2 7XH, UK; 7 Sanofi US Services, 640 Memorial Drive, Cambridge MA 02139, USA; 8 Sanofi US Services, 55 Corporate Drive, Bridgewater, NJ 08807, USA. The authors confirm that the Principal Investigators for the clinical study were Grit Anderson and Leona Plum- Mörschel and that they had direct clinical responsibility for patients at the Neuss and Mainz sites, respectively.
    [Show full text]
  • 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]
  • 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]
  • CXCL13/CXCR5 Interaction Facilitates VCAM-1-Dependent Migration in Human Osteosarcoma
    International Journal of Molecular Sciences Article CXCL13/CXCR5 Interaction Facilitates VCAM-1-Dependent Migration in Human Osteosarcoma 1, 2,3,4, 5 6 7 Ju-Fang Liu y, Chiang-Wen Lee y, Chih-Yang Lin , Chia-Chia Chao , Tsung-Ming Chang , Chien-Kuo Han 8, Yuan-Li Huang 8, Yi-Chin Fong 9,10,* and Chih-Hsin Tang 8,11,12,* 1 School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei City 11031, Taiwan; [email protected] 2 Department of Orthopaedic Surgery, Chang Gung Memorial Hospital, Puzi City, Chiayi County 61363, Taiwan; [email protected] 3 Department of Nursing, Division of Basic Medical Sciences, and Chronic Diseases and Health Promotion Research Center, Chang Gung University of Science and Technology, Puzi City, Chiayi County 61363, Taiwan 4 Research Center for Industry of Human Ecology and Research Center for Chinese Herbal Medicine, Chang Gung University of Science and Technology, Guishan Dist., Taoyuan City 33303, Taiwan 5 School of Medicine, China Medical University, Taichung 40402, Taiwan; [email protected] 6 Department of Respiratory Therapy, Fu Jen Catholic University, New Taipei City 24205, Taiwan; [email protected] 7 School of Medicine, Institute of Physiology, National Yang-Ming University, Taipei City 11221, Taiwan; [email protected] 8 Department of Biotechnology, College of Health Science, Asia University, Taichung 40402, Taiwan; [email protected] (C.-K.H.); [email protected] (Y.-L.H.) 9 Department of Sports Medicine, College of Health Care, China Medical University, Taichung 40402, Taiwan 10 Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yunlin 65152, Taiwan 11 Department of Pharmacology, School of Medicine, China Medical University, Taichung 40402, Taiwan 12 Chinese Medicine Research Center, China Medical University, Taichung 40402, Taiwan * Correspondence: [email protected] (Y.-C.F.); [email protected] (C.-H.T.); Tel.: +886-4-2205-2121-7726 (C.-H.T.); Fax: +886-4-2233-3641 (C.-H.T.) These authors contributed equally to this work.
    [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]
  • Reck and Gpr124 Activate Canonical Wnt Signaling To
    RECK AND GPR124 ACTIVATE CANONICAL WNT SIGNALING TO CONTROL MAMMALIAN CENTRAL NERVOUS SYSTEM ANGIOGENESIS AND BLOOD-BRAIN BARRIER REGULATION by Chris Moonho Cho A dissertation submitted to Johns Hopkins University in conformity with the requirements for the degree of Doctor of Philosophy Baltimore, Maryland July 2018 © Chris Moonho Cho 2018 All rights reserved Abstract Canonical Wnt signaling plays a pivotal role in promoting central nervous system (CNS) angiogenesis and blood-brain barrier (BBB) formation and maintenance. Specifically, Wnt7a and Wnt7b are required for vascular development in the forebrain and ventral spinal cord. Yet, how these two ligands – among the 19 mammalian Wnts – are selectively communicated to Frizzled receptors expressed on endothelial cells (ECs) remains largely unclear. In this thesis, we propose a novel paradigm for Wnt specificity. We have identified two EC surface proteins – orphan receptor Gpr124, and more recently, GPI-anchored Reck (reversion-inducing cysteine-rich protein with Kazal motifs) – as essential receptor co-factors that assemble into a multi-protein complex with Wnt7a/7b and Frizzled for the development of the mammalian neurovasculature. Specifically, we show that EC-specific reduction in Reck impairs CNS angiogenesis and that EC-specific postnatal loss of Reck, combined with loss of Norrin, impairs BBB maintenance. We identify the critical domains of both Reck and Gpr124 that are required for Wnt activity, and demonstrate that these regions are important for ii direct binding and complex formation. Importantly, weakening this interaction by targeted mutagenesis reduces Reck-Gpr124 stimulation of Wnt7a signaling in cell culture and impairs CNS angiogenesis. Finally, a soluble Gpr124 probe binds specifically to cells expressing Frizzled (Fz), Wnt7a or Wnt7b, and Reck; and a soluble Reck probe binds specifically to cells expressing Fz, Wnt7a or Wnt7b, and Gpr124.
    [Show full text]
  • Neutrophil Chemoattractant Receptors in Health and Disease: Double-Edged Swords
    Cellular & Molecular Immunology www.nature.com/cmi REVIEW ARTICLE Neutrophil chemoattractant receptors in health and disease: double-edged swords Mieke Metzemaekers1, Mieke Gouwy1 and Paul Proost 1 Neutrophils are frontline cells of the innate immune system. These effector leukocytes are equipped with intriguing antimicrobial machinery and consequently display high cytotoxic potential. Accurate neutrophil recruitment is essential to combat microbes and to restore homeostasis, for inflammation modulation and resolution, wound healing and tissue repair. After fulfilling the appropriate effector functions, however, dampening neutrophil activation and infiltration is crucial to prevent damage to the host. In humans, chemoattractant molecules can be categorized into four biochemical families, i.e., chemotactic lipids, formyl peptides, complement anaphylatoxins and chemokines. They are critically involved in the tight regulation of neutrophil bone marrow storage and egress and in spatial and temporal neutrophil trafficking between organs. Chemoattractants function by activating dedicated heptahelical G protein-coupled receptors (GPCRs). In addition, emerging evidence suggests an important role for atypical chemoattractant receptors (ACKRs) that do not couple to G proteins in fine-tuning neutrophil migratory and functional responses. The expression levels of chemoattractant receptors are dependent on the level of neutrophil maturation and state of activation, with a pivotal modulatory role for the (inflammatory) environment. Here, we provide an overview
    [Show full text]
  • Original Article Expression of Chemokine Receptor CXCR5 in Gastric Cancer and Its Clinical Significance
    Int J Clin Exp Pathol 2016;9(7):7202-7208 www.ijcep.com /ISSN:1936-2625/IJCEP0023559 Original Article Expression of chemokine receptor CXCR5 in gastric cancer and its clinical significance Qing Sun*, Lujun Chen*, Bin Xu, Qi Wang, Xiao Zheng, Peng Du, Dachuan Zhang, Changping Wu, Jingting Jiang Department of Tumor Biological Treatment, The Third Affiliated Hospital, Soochow University, Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu, China. *Equal contributors. Received January 8, 2016; Accepted March 22, 2016; Epub July 1, 2016; Published July 15, 2016 Abstract: The increased expression of chemokine receptor CXCR5 in cancers has been demonstrated. In order to characterize the expression pattern of CXCR5 in cell lines and tissues of gastric cancer and to assess clinical implications, the expression of CXCR5 mRNA in gastric cancer tissues and adjacent tissues was evaluated by real- time RT-PCR. Meanwhile, the expression of CXCR5 in cell lines of human gastric cancer was also analyzed by flow cytometry. Tissue microarray and immunohistochemistry were used to detect the protein expression of CXCR5 in human gastric cancer tissues and adjacent normal tissues. Flow cytometry results revealed the positive expression of CXCR5 in human gastric cancer cell lines such as BGC-823, SGC-7901 and HGC-27 cells. The immunohistochem- istry results showed higher expression of CXCR5 in 52.87% of gastric cancer tissues. The expression of CXCR5 in patients with tumor size less than 2.8 cm subgroup was significantly lower than that in patients with tumor size larger than 2.8 cm subgroup (P = 0.0456). There was no significant correlation between the expression of CXCR5 and other clinical parameters in gastric cancer.
    [Show full text]
  • Constitutive Activation of G Protein-Coupled Receptors and Diseases: Insights Into Mechanisms of Activation and Therapeutics
    Pharmacology & Therapeutics 120 (2008) 129–148 Contents lists available at ScienceDirect Pharmacology & Therapeutics journal homepage: www.elsevier.com/locate/pharmthera Associate editor: S. Enna Constitutive activation of G protein-coupled receptors and diseases: Insights into mechanisms of activation and therapeutics Ya-Xiong Tao ⁎ Department of Anatomy, Physiology and Pharmacology, 212 Greene Hall, College of Veterinary Medicine, Auburn University, Auburn, AL 36849, USA article info abstract The existence of constitutive activity for G protein-coupled receptors (GPCRs) was first described in 1980s. In Keywords: 1991, the first naturally occurring constitutively active mutations in GPCRs that cause diseases were reported G protein-coupled receptor Disease in rhodopsin. Since then, numerous constitutively active mutations that cause human diseases were reported Constitutively active mutation in several additional receptors. More recently, loss of constitutive activity was postulated to also cause Inverse agonist diseases. Animal models expressing some of these mutants confirmed the roles of these mutations in the Mechanism of activation pathogenesis of the diseases. Detailed functional studies of these naturally occurring mutations, combined Transgenic model with homology modeling using rhodopsin crystal structure as the template, lead to important insights into the mechanism of activation in the absence of crystal structure of GPCRs in active state. Search for inverse Abbreviations: agonists on these receptors will be critical for correcting the diseases cause by activating mutations in GPCRs. ADRP, autosomal dominant retinitis pigmentosa Theoretically, these inverse agonists are better therapeutics than neutral antagonists in treating genetic AgRP, Agouti-related protein AR, adrenergic receptor diseases caused by constitutively activating mutations in GPCRs. CAM, constitutively active mutant © 2008 Elsevier Inc.
    [Show full text]
  • Differential Effects of Mineralocorticoid and Angiotensin II on Incentive and Mesolimbic Activity Laura A
    Bryn Mawr College Scholarship, Research, and Creative Work at Bryn Mawr College Psychology Faculty Research and Scholarship Psychology 2016 Differential effects of mineralocorticoid and angiotensin II on incentive and mesolimbic activity Laura A. Grafe Bryn Mawr College, [email protected] Loretta M. Flanagan-Cato Let us know how access to this document benefits ouy . Follow this and additional works at: https://repository.brynmawr.edu/psych_pubs Part of the Psychology Commons Custom Citation Grafe, Laura A. and Loretta M. Flanagan-Cato. 2016. "Differential effects of mineralocorticoid and angiotensin II on incentive and mesolimbic activity." Hormones and Behavior 79: 28–36. This paper is posted at Scholarship, Research, and Creative Work at Bryn Mawr College. https://repository.brynmawr.edu/psych_pubs/71 For more information, please contact [email protected]. 1 2 DIFFERENTIAL EFFECTS OF MINERALOCORTICOID 3 AND ANGIOTENSIN II ON INCENTIVE AND MESOLIMBIC ACTIVITY 4 5 Laura A. Grafea and Loretta M. Flanagan-Catoa,b,c 6 7 Neuroscience Graduate Groupa, Department of Psychologyb, and the Mahoney Institute of 8 Neurological Sciencesc, University of Pennsylvania, 9 Philadelphia Pennsylvania, USA 10 11 Abbreviated title: Aldo and AngII-induced motivation for sodium 12 13 14 Corresponding Author: 15 L.M. Flanagan-Cato 16 [email protected] 17 3720 Walnut Street, Philadelphia, PA, 19104 18 19 Laura A. Grafe co-designed the research, executed the experiments, analyzed the data, and co- 20 wrote the paper. Loretta M. Flanagan-Cato co-designed research, analyzed the data, and co- 21 wrote the paper. 22 23 ABSTRACT 24 The controls of thirst and sodium appetite are mediated in part by the hormones 25 aldosterone and angiotensin II (AngII).
    [Show full text]