Expression and Localization of Somatostatin Receptor Types 3, 4 and 5 in the Wild-Type, SSTR1 and SSTR1/SSTR2 Knockout Mouse Cochlea

Total Page:16

File Type:pdf, Size:1020Kb

Expression and Localization of Somatostatin Receptor Types 3, 4 and 5 in the Wild-Type, SSTR1 and SSTR1/SSTR2 Knockout Mouse Cochlea Cell Tissue Res (2014) 358:717–727 DOI 10.1007/s00441-014-1977-7 REGULAR ARTICLE Expression and localization of somatostatin receptor types 3, 4 and 5 in the wild-type, SSTR1 and SSTR1/SSTR2 knockout mouse cochlea Vesna Radojevic & Daniel Bodmer Received: 5 November 2013 /Accepted: 24 July 2014 /Published online: 23 August 2014 # Springer-Verlag Berlin Heidelberg 2014 Abstract Somatostatin (SST) is a peptide hormone that ex- actions of SST. Five distinct SST receptor types (SSTR1-5) erts inhibitory effects mediated through binding to specific have been identified (Patel 1999). cell surface G protein-coupled receptors, of which five distinct The distribution of mRNA expression for all five SST recep- subtypes (SSTR1-SSTR5) have been characterized. Our study tor types has been investigated extensively using different tech- performed on mouse cochlear hair cells shows the expression niques in rats and humans (Harrington et al. 1995;Kongetal. and localization of the three receptors (SSTR3-SSTR5) in 1994; Schindler et al. 1995). Published studies are in agreement wild-type (WT), single-knockout (SSTR1 KO) and double- and suggest that all SSTR types are variably expressed wide- knockout SSTR1/SSTR2 (DKO) mice. Similar SSTRs ex- spread throughout the brain. In rats, SSTR1 mRNA is the most pression were observed in the inner hair cells (IHC), outer abundant, followed by SSTR2, SSTR5, SSTR3 and SSTR4 hair cells (OHC) and supporting cells of cultivated P7 mouse (Patel 1999;Brederetal.1992; Kong et al. 1994;Thossetal. organ of Corti (OC) explants as well as in cultivated cochlear 1995;Beaudetetal.1995). SSTR3 immunoreactivity is widely neuroepithelial supporting cells (NEsc). We found differences distributed throughout the human brain and is particularly in the expression of SSTR3-5 in WT, SSTR1 KO and DKO enriched in the cortex, hippocampus, amygdala, hypothalamus mouse cochlea, which might be explained as a compensatory and cerebellum (Hervieu and Emson 1999; Schindler et al. effect in the cochlea after the loss of SSTR1 and/or SSTR2. 1995). Immunohistochemistry of adult rat brain sections showed that SSTR4 is distributed in both the cell body and neuronal Keywords Cochlea . Hair cells . Knockout mice . Organ of processes in the cerebral cortex, hippocampus and several Corti . Somatostatin . Somatostatin receptor nuclei of the brainstem (Selmer et al. 2000). Schreff et al. (2000) showed that SSTR4 is strictly somatodendritic and most likely functions in a postsynaptic manner. SSTR4 may have a previously unappreciated function during neuronal degenera- Introduction tion/regeneration. SSTR5 is prominently expressed in the basal forebrain, suggesting that it is involved in the mediation of the SST, a regulatory peptide with two bioactive forms, SST-14 effects of SST on the sleep-wake cycle through its association and SST-28, is produced in neuroendocrine cells in the brain with cortically projecting subcortical systems (Stroh et al. 1999). and periphery and acts on a number of tissue targets to Most SSTR subtypes have been shown to modulate voltage- modulate neurotransmission, cell secretion and cell prolifera- gated Ca2+ channels and glutamate receptor channels in several tion (Reichlin 1983; Epelbaum et al. 1994; Patel 1999). A organ systems (Cervia and Bagnoli 2007). Excessive Ca2+ family of transmembrane receptors mediates the physiological influx into cells is thought to be a major contributor to cell death in ischemic and excitotoxic models of neuronal injury (Smaili V. Radojevic (*) : D. Bodmer Department of Biomedicine, University Hospital Basel, Petersgraben et al. 2009). SSTRs are also involved in the regulation of ion 2+ + 4, 4031 Basel, Switzerland channels, the inhibition of Ca channels and activation of K e-mail: [email protected] channels, involved in the release of several neurotransmitters and the modulation of neurotransmission (Patel 1999; Petrucci V. Radojevic : D. Bodmer Clinic for Otolaryngology, Head and Neck Surgery, University et al. 2001; Johnson et al. 2001). All five subtypes of SSTRs are Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland expressed in the mammalian retina and are distributed to specific 718 Cell Tissue Res (2014) 358:717–727 cell populations, including subtypes of photoreceptor, bipolar, RNA extraction amacrine and ganglion cells (Thermos 2003;KeandZhong 2007; Vasilaki and Thermos 2009). Studies have shown that The SG, brain (Br) and cell lysate (Jayanthi et al. 2005)from SST analogues may be effective therapeutic agents in retinal P7 mice, the OC from WT mouse pups (P0–P21) and OC of diseases (Vasilaki and Thermos 2009). DKO mouse pups by age P14 and P21 were placed separately However, in contrast to the situation in the retina, little in RNAlater (Qiagen, Hombrechtikon, Switzerland). RNA information is available on the expression or function of isolation of the brains and inner ear components was per- somatostatin and its receptors in the inner ear. Tachibana formed using the RNAeasy Minikit (Qiagen) and an Ultra- et al. (1979) reported on somatostatin-like immunoreactivity Turrax T8 tissue homogenizer (IKA-Werke, Staufen, in the medial geniculate body, cochlear nucleus, inferior Germany) according to the manufacturer’s instructions, in- colliculus, auditory cortex and in the cochlea. Also, cluding DNase treatment. The quantity and quality of isolated somatostatin-like immunoreactivity could be observed in the RNA was determined with a NanoDrop ND 1000 (NanoDrop cochlear nuclei of postnatal rats and it was suggested that Technologies, DE, USA). The 260-/280-nm ratio of all sam- somatostatin might be important for the development of the ples was between 1.8 and 2.1. auditory system (Takatsuki et al. 1982). Our previous study demonstrated that SSTR1 and SSTR2 Reverse transcription polymerase chain reaction mRNA and protein are expressed in the mouse cochlea and localized in IHC and OHC and in supporting cells of the OC Total RNA (500 ng) was reverse transcribed into cDNA using (Radojevic et al. 2011). The present study is the first attempt to the first strand cDNA synthesis kit for reverse transcription determine SSTR3, -4 and -5 expression in the cochlea from polymerase chain reaction (RT-PCR) (Roche Applied Biosci- WT and DKO mice. ence, Rotkreuz, Switzerland) according to the supplier’sin- structions. PCR was performed using the PCR Master Mix (Roche Applied Bioscience) with primers specific for SSTR3, SSTR4 and SSTR5. β-actin primers were used as a positive Materials and methods control for cDNA synthesis. The primers were designed to cross at least one exon junction for the specific amplification Animals of cDNA and to recognize contamination with genomic DNA. The primer sequences were: Experiments were performed on mouse cochleae from WT C57BL/6 mice of both sexes. The patterns of SSTR3-5 ex- SSTR1 forward (fwd): 5’-CAGGGTAGCCATTCTC pression were also observed at postnatal ages (n=10 per ATC-3’, postnatal day for mRNA isolation). Immunohistochemistry SSTR1 reverse (rev): 5’-AGCGTGGAAGTGACCAGA was performed on four mice per postnatal day. The procedures − − AAG-3’, used to generate SSTR1 / (homozygous SSTR1 KO)/ − − SSTR2 (fwd): 5’-CGCATGGTGTCCATCGTAGT-3’, C57BL6J mice (Kreienkamp et al. 1999) and SSTR2 / (ho- SSTR2 (rev): 5’-GGATTGTGAATTGTCTGCCTTGA-3’, mozygous SSTR2 KO)/C57BL6J mice have been described SSTR3 (fwd): 5’-CTGGCTGTCAGTGGCATCTT-3’, previously (Zheng et al. 1997). SSTR1 KO mice were crossed SSTR3 (rev): 5’-CACATGAGATCCGAAGGGC-3’, with SSTR2 KO mice to produce DKO mice. Age-matched SSTR4 fwd: 5’-CAGCGGCATGGTCACTATC-3’, WT mice were produced from C57BL6J mice used to stabilize SSTR4 rev: 5’-CCGTCCACGCTAAGCACTG-3’, the genetic backgrounds of SSTR1 KO and SSTR2 KO mice SSTR5 fwd: 5’-TTCCTGGCAACGCAGAATG-3’, (Bodmer et al. 2012). SSTR5 rev: 5’-CAAGAGCGGCAGAGACATGAG-3’, Animal care and use were conducted in conformity with β-actin fwd, 5’-ACGGTCAGGTCATCACTATCGG the European Communities Council Directive of 24 Novem- CA-3’, ber 1986 (86/609/EEC) and reviewed and permitted by the β-actin rev, 5’-ATCCTGTCAGCAATGCCTGGGT-3’ Kantonales Veterinäramt, Basel, Switzerland. (Microsynth, St. Gallen, Switzerland). Tissue extraction and culture PCR reactions were performed in an Eppendorf Mastercycler (Eppendorf, Hamburg, Germany). For SSTR3, The mice were decapitated and cochlear microdissections SSTR4 and SSTR5, an initial denaturing step at 94 °C for 30 s were performed under a light microscope to isolate the OC was followed by 30 PCR cycles consisting in denaturation at and the spiral ganglion (SG). Brain tissue was removed from 94 °C for 15 s, annealing at 60 °C for 30 s and extension at the same animals. During microdissection, the different tis- 72 °C for 15 s. The PCR was completed with a final extension sues were maintained in ice-cold PBS. step at 72 °C for 3 min. β-actin was subjected to 30 PCR Cell Tissue Res (2014) 358:717–727 719 cycles consisting in denaturation at 94 °C for 30 s, annealing SSTR5-fwd: 5′-GGCTTCCACACCTAGCTGG-3′, at 58 °C for 45 s and extension at 72 °C for 90 s. The PCR was SSTR5-rev: 5′-AGCACAGGCACTAATACCGC- completed with a final extension step at 72° for 5 min. PCR 3′(Microsynth). products were stained with SYBR Green I (Molecular Probes, OR, USA), separated by electrophoresis on a 2 % agarose gel Each reaction contained 300 nM of primer. The reaction and visualized under ultraviolet light. Omission of RT-PCR occurred at 95 °C for 10 min, followed by 40 cycles of 95 °C was used as the no template control. for 15 s and 60 °C for 60 s. We calculated the relative quantities of specifically amplified cDNA using the compar- Real-time PCR ative threshold cycle method. GAPDH was used as an endog- enous reference (Microsynth) (Bodmer et al. 2012;Halletal. Total RNA (500 ng) was reverse transcribed into cDNA using 2007; Chen and Ruan 2009). Template-free and reverse the First Strand cDNA synthesis kit (Roche Applied Biosci- transcription-free controls excluded non-specific amplifica- ences, Foster City, USA) according to the manufacturer’s tion and DNA contamination. instructions.
Recommended publications
  • Strategies to Increase ß-Cell Mass Expansion
    This electronic thesis or dissertation has been downloaded from the King’s Research Portal at https://kclpure.kcl.ac.uk/portal/ Strategies to increase -cell mass expansion Drynda, Robert Lech Awarding institution: King's College London The copyright of this thesis rests with the author and no quotation from it or information derived from it may be published without proper acknowledgement. END USER LICENCE AGREEMENT Unless another licence is stated on the immediately following page this work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International licence. https://creativecommons.org/licenses/by-nc-nd/4.0/ You are free to copy, distribute and transmit the work Under the following conditions: Attribution: You must attribute the work in the manner specified by the author (but not in any way that suggests that they endorse you or your use of the work). Non Commercial: You may not use this work for commercial purposes. No Derivative Works - You may not alter, transform, or build upon this work. Any of these conditions can be waived if you receive permission from the author. Your fair dealings and other rights are in no way affected by the above. Take down policy If you believe that this document 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 Strategies to increase β-cell mass expansion A thesis submitted by Robert Drynda For the degree of Doctor of Philosophy from King’s College London Diabetes Research Group Division of Diabetes & Nutritional Sciences Faculty of Life Sciences & Medicine King’s College London 2017 Table of contents Table of contents .................................................................................................
    [Show full text]
  • Investigation of the Underlying Hub Genes and Molexular Pathogensis in Gastric Cancer by Integrated Bioinformatic Analyses
    bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Investigation of the underlying hub genes and molexular pathogensis in gastric cancer by integrated bioinformatic analyses Basavaraj Vastrad1, Chanabasayya Vastrad*2 1. Department of Biochemistry, Basaveshwar College of Pharmacy, Gadag, Karnataka 582103, India. 2. Biostatistics and Bioinformatics, Chanabasava Nilaya, Bharthinagar, Dharwad 580001, Karanataka, India. * Chanabasayya Vastrad [email protected] Ph: +919480073398 Chanabasava Nilaya, Bharthinagar, Dharwad 580001 , Karanataka, India bioRxiv preprint doi: https://doi.org/10.1101/2020.12.20.423656; this version posted December 22, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder. All rights reserved. No reuse allowed without permission. Abstract The high mortality rate of gastric cancer (GC) is in part due to the absence of initial disclosure of its biomarkers. The recognition of important genes associated in GC is therefore recommended to advance clinical prognosis, diagnosis and and treatment outcomes. The current investigation used the microarray dataset GSE113255 RNA seq data from the Gene Expression Omnibus database to diagnose differentially expressed genes (DEGs). Pathway and gene ontology enrichment analyses were performed, and a proteinprotein interaction network, modules, target genes - miRNA regulatory network and target genes - TF regulatory network were constructed and analyzed. Finally, validation of hub genes was performed. The 1008 DEGs identified consisted of 505 up regulated genes and 503 down regulated genes.
    [Show full text]
  • Supplementary Table 2
    Supplementary Table 2. Differentially Expressed Genes following Sham treatment relative to Untreated Controls Fold Change Accession Name Symbol 3 h 12 h NM_013121 CD28 antigen Cd28 12.82 BG665360 FMS-like tyrosine kinase 1 Flt1 9.63 NM_012701 Adrenergic receptor, beta 1 Adrb1 8.24 0.46 U20796 Nuclear receptor subfamily 1, group D, member 2 Nr1d2 7.22 NM_017116 Calpain 2 Capn2 6.41 BE097282 Guanine nucleotide binding protein, alpha 12 Gna12 6.21 NM_053328 Basic helix-loop-helix domain containing, class B2 Bhlhb2 5.79 NM_053831 Guanylate cyclase 2f Gucy2f 5.71 AW251703 Tumor necrosis factor receptor superfamily, member 12a Tnfrsf12a 5.57 NM_021691 Twist homolog 2 (Drosophila) Twist2 5.42 NM_133550 Fc receptor, IgE, low affinity II, alpha polypeptide Fcer2a 4.93 NM_031120 Signal sequence receptor, gamma Ssr3 4.84 NM_053544 Secreted frizzled-related protein 4 Sfrp4 4.73 NM_053910 Pleckstrin homology, Sec7 and coiled/coil domains 1 Pscd1 4.69 BE113233 Suppressor of cytokine signaling 2 Socs2 4.68 NM_053949 Potassium voltage-gated channel, subfamily H (eag- Kcnh2 4.60 related), member 2 NM_017305 Glutamate cysteine ligase, modifier subunit Gclm 4.59 NM_017309 Protein phospatase 3, regulatory subunit B, alpha Ppp3r1 4.54 isoform,type 1 NM_012765 5-hydroxytryptamine (serotonin) receptor 2C Htr2c 4.46 NM_017218 V-erb-b2 erythroblastic leukemia viral oncogene homolog Erbb3 4.42 3 (avian) AW918369 Zinc finger protein 191 Zfp191 4.38 NM_031034 Guanine nucleotide binding protein, alpha 12 Gna12 4.38 NM_017020 Interleukin 6 receptor Il6r 4.37 AJ002942
    [Show full text]
  • Somatostatin Receptor Types 1 and 2 in the Developing Mammalian Cochlea
    Original Paper Dev Neurosci 2012;34:342–353 Received: December 20, 2011 DOI: 10.1159/000341291 Accepted after revision: June 20, 2012 Published online: September 13, 2012 Somatostatin Receptor Types 1 and 2 in the Developing Mammalian Cochlea Daniel Bodmer Yves Brand Vesna Radojevic Department of Biomedicine and Clinic for Otorhinolaryngology, University Hospital Basel, Basel , Switzerland Key Words (P0) to P10; the majority of immunostained cells were inner Central nervous system ؒ Cochlea ؒ Double-knockout hair cells, outer hair cells, and supporting cells. Finally, a peak mouse ؒ Inner ear ؒ Somatostatin ؒ Somatostatin receptors in the mRNA and protein expression of both receptors is present near the time when they respond to physiological hearing (i.e., hearing of airborne sound) at P14. At P21, SSTR1 Abstract and SSTR2 levels decrease dramatically. A similar develop- The neuropeptide somatostatin (SST) exerts several impor- mental pattern was observed for SSTR1 and SSTR2 mRNA, tant physiological actions in the adult central nervous sys- suggesting that the expression of the SSTR1 and SSTR2 tem through interactions with membrane-bound receptors. genes is controlled at the transcriptional level throughout Transient expression of SST and its receptors has been de- development. In addition, we observed reduced levels of scribed in several brain areas during early ontogeny. It is phospho-Akt and total Akt in SSTR1 knockout and SSTR1/ therefore believed that SST may play a role in neural matura- SSTR2 double-knockout mice compared with wild-type tion. The present study provides the first evidence for the mice. We know from previous studies that Akt is involved developmental expression of SST receptors in the mamma- in hair cell survival.
    [Show full text]
  • Internalization of Sst2, Sst3, and Sst5 Receptors: Effects of Somatostatin Agonists and Antagonists
    Internalization of sst2, sst3, and sst5 Receptors: Effects of Somatostatin Agonists and Antagonists Renzo Cescato, PhD1; Stefan Schulz, PhD2; Beatrice Waser1;Ve´ronique Eltschinger1; Jean E. Rivier, PhD3; Hans-Ju¨rgen Wester, PhD4; Michael Culler, PhD5; Mihaela Ginj, PhD6; Qisheng Liu, MD, PhD7; Agnes Schonbrunn, PhD7; and Jean Claude Reubi, MD1 1Division of Cell Biology and Experimental Cancer Research, Institute of Pathology, University of Berne, Berne, Switzerland; 2Institute of Pharmacology and Toxicology, Otto-von-Guericke University, Magdeburg, Germany; 3Clayton Foundation Laboratories for Peptide Biology, Salk Institute, La Jolla, California; 4Nuklearmedizinische Klinik und Poliklinik, Technical University of Munich, Munich, Germany; 5Endocrine Research, Beaufour-Ipsen Group, Milford, Massachusetts; 6Division of Radiological Chemistry, University Hospital, Basel, Switzerland; and 7Department of Integrative Biology and Pharmacology, University of Texas Health Science Center–Houston, Houston, Texas sensitive and reproducible immunocytochemical methods, the The uptake of radiolabeled somatostatin analogs by tumor cells ability of various somatostatin analogs to induce sst2, sst3, and through receptor-mediated internalization is a critical process sst5 internalization has been qualitatively and quantitatively de- for the in vivo targeting of tumoral somatostatin receptors. In termined. Whereas all agonists triggered sst2 and sst3 internali- the present study, the somatostatin receptor internalization in- zation, sst5 internalization
    [Show full text]
  • Adenylyl Cyclase 2 Selectively Regulates IL-6 Expression in Human Bronchial Smooth Muscle Cells Amy Sue Bogard University of Tennessee Health Science Center
    University of Tennessee Health Science Center UTHSC Digital Commons Theses and Dissertations (ETD) College of Graduate Health Sciences 12-2013 Adenylyl Cyclase 2 Selectively Regulates IL-6 Expression in Human Bronchial Smooth Muscle Cells Amy Sue Bogard University of Tennessee Health Science Center Follow this and additional works at: https://dc.uthsc.edu/dissertations Part of the Medical Cell Biology Commons, and the Medical Molecular Biology Commons Recommended Citation Bogard, Amy Sue , "Adenylyl Cyclase 2 Selectively Regulates IL-6 Expression in Human Bronchial Smooth Muscle Cells" (2013). Theses and Dissertations (ETD). Paper 330. http://dx.doi.org/10.21007/etd.cghs.2013.0029. This Dissertation is brought to you for free and open access by the College of Graduate Health Sciences at UTHSC Digital Commons. It has been accepted for inclusion in Theses and Dissertations (ETD) by an authorized administrator of UTHSC Digital Commons. For more information, please contact [email protected]. Adenylyl Cyclase 2 Selectively Regulates IL-6 Expression in Human Bronchial Smooth Muscle Cells Document Type Dissertation Degree Name Doctor of Philosophy (PhD) Program Biomedical Sciences Track Molecular Therapeutics and Cell Signaling Research Advisor Rennolds Ostrom, Ph.D. Committee Elizabeth Fitzpatrick, Ph.D. Edwards Park, Ph.D. Steven Tavalin, Ph.D. Christopher Waters, Ph.D. DOI 10.21007/etd.cghs.2013.0029 Comments Six month embargo expired June 2014 This dissertation is available at UTHSC Digital Commons: https://dc.uthsc.edu/dissertations/330 Adenylyl Cyclase 2 Selectively Regulates IL-6 Expression in Human Bronchial Smooth Muscle Cells A Dissertation Presented for The Graduate Studies Council The University of Tennessee Health Science Center In Partial Fulfillment Of the Requirements for the Degree Doctor of Philosophy From The University of Tennessee By Amy Sue Bogard December 2013 Copyright © 2013 by Amy Sue Bogard.
    [Show full text]
  • Genetic Deletion of Somatostatin Receptor 1 Alters Somatostatinergic
    Neuropharmacology 45 (2003) 1080–1092 www.elsevier.com/locate/neuropharm Genetic deletion of somatostatin receptor 1 alters somatostatinergic transmission in the mouse retina Massimo Dal Monte a, Cristina Petrucci a, Anna Vasilaki b, Davide Cervia a, Dominique Grouselle c, Jacques Epelbaum c, Hans-Jurgen Kreienkamp d, Dietmar Richter d, Daniel Hoyer b, Paola Bagnoli a,∗ a Dipartimento di Fisiologia e Biochimica, Universita` di Pisa, via San Zeno 31, 56127 Pisa, Italy b Nervous System Research, Novartis Pharma AG, WSJ-386/745, 4002 Basel, Switzerland c INSERUM, U-549, IFR Broca-Sainte Anne, Centre Paul Broca, 2ter rue d’Alesia, 75014 Paris, France d Institut fu¨r Zellbiochemie und Klinische Neurobiologie, Universita¨ts Kronkenhaus Eppendorf, 20246, Hamburg, Germany Received 30 April 2003; received in revised form 8 July 2003; accepted 16 July 2003 Abstract In the mammalian retina, sparse amacrine cells contain somatostatin-14 (SRIF) which acts at multiple levels of neuronal circuitry through distinct SRIF receptors (sst1–5). Among them, the sst1 receptor has been localised to SRIF-containing amacrine cells in the rat and rabbit retina. Little is known about sst1 receptor localisation and function in the mouse retina. We have addressed this question in the retina of mice with deletion of sst1 receptors (sst1 KO mice). In the retina of wild type (WT) mice, sst1 receptors are localised to SRIF-containing amacrine cells, whereas in the retina of sst1 KO mice, sst1 receptors are absent. sst1 receptor loss causes a significant increase in retinal levels of SRIF, whereas it does not affect SRIF messenger RNA indicating that sst1 receptors play a role in limiting retinal SRIF at the post-transcriptional level.
    [Show full text]
  • Dynamic Mass Redistribution Reveals Diverging Importance of PDZ
    Pharmacological Research 105 (2016) 13–21 Contents lists available at ScienceDirect Pharmacological Research j ournal homepage: www.elsevier.com/locate/yphrs Dynamic mass redistribution reveals diverging importance of PDZ-ligands for G protein-coupled receptor pharmacodynamics a b b b Nathan D. Camp , Kyung-Soon Lee , Allison Cherry , Jennifer L. Wacker-Mhyre , b b b b Timothy S. Kountz , Ji-Min Park , Dorathy-Ann Harris , Marianne Estrada , b b a b,∗ Aaron Stewart , Nephi Stella , Alejandro Wolf-Yadlin , Chris Hague a Department of Genome Sciences, University of Washington School of Medicine, Seattle, WA 98195, USA b Department of Pharmacology, University of Washington School of Medicine, Seattle, WA 98195, USA a r t i c l e i n f o a b s t r a c t Article history: G protein-coupled receptors (GPCRs) are essential membrane proteins that facilitate cell-to-cell Received 19 October 2015 communication and co-ordinate physiological processes. At least 30 human GPCRs contain a Type I PSD- Received in revised form 95/DLG/Zo-1 (PDZ) ligand in their distal C-terminal domain; this four amino acid motif of X-[S/T]-X-[␸] 28 December 2015 sequence facilitates interactions with PDZ domain-containing proteins. Because PDZ protein interactions Accepted 1 January 2016 have profound effects on GPCR ligand pharmacology, cellular localization, signal-transduction effector Available online 7 January 2016 coupling and duration of activity, we analyzed the importance of Type I PDZ ligands for the function of 23 full-length and PDZ-ligand truncated (PDZ) human GPCRs in cultured human cells. SNAP-epitope tag Keywords: polyacrylamide gel electrophoresis revealed most Type I PDZ GPCRs exist as both monomers and mul- G protein-coupled receptor timers; removal of the PDZ ligand played minimal role in multimer formation.
    [Show full text]
  • Hippo Signaling: Emerging Pathway in Stress-Related Psychiatric Disorders?
    MINI REVIEW published: 21 December 2018 doi: 10.3389/fpsyt.2018.00715 Hippo Signaling: Emerging Pathway in Stress-Related Psychiatric Disorders? Jens Stepan 1, Elmira Anderzhanova 2 and Nils C. Gassen 2* 1 Department Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, Germany, 2 Clinic and Polyclinic of Psychiatry and Psychotherapy, Bonn University Clinic, Bonn, Germany Discovery of the Hippo pathway and its core components has made a significant impact on our progress in the understanding of organ development, tissue homeostasis, and regeneration. Upon diverse extracellular and intracellular stimuli, Hippo signaling regulates stemness, cell proliferation and apoptosis by a well-conserved signaling cascade, and disruption of these systems has been implicated in cancer as well as metabolic and neurodegenerative diseases. The central role of Hippo signaling in cell biology also results in prominent links to stress-regulated pathways. Genetic variations, epigenetically provoked upregulation of Hippo pathway members and dysregulation of cellular processes implicated in learning and memory, are linked to an increased risk of stress-related psychiatric disorders (SRPDs). In this review, we summarize recent Edited by: Naguib Mechawar, findings, supporting the role of Hippo signaling in SRPDs by canonical and non-canonical McGill University, Canada Hippo pathway interactions. Reviewed by: Keywords: hippo pathway, KIBRA, psychophysiological stress, synaptic plasticity, glucocorticoids, GPCRs Emmanuel Moyse, Université de Tours, France Antoine Besnard, Harvard Medical School, INTRODUCTION United States *Correspondence: When the Hippo pathway was first discovered in Drosophila, it appeared as a linear kinase Nils C. Gassen cascade highly relevant for proliferation and homeostasis, because deletion of core component [email protected] genes resulted in an uncontrolled growth of multiple tissues (1, 2).
    [Show full text]
  • 763.Full.Pdf
    1521-0081/70/4/763–835$35.00 https://doi.org/10.1124/pr.117.015388 PHARMACOLOGICAL REVIEWS Pharmacol Rev 70:763–835, October 2018 Copyright © 2018 The Author(s). This is an open access article distributed under the CC BY Attribution 4.0 International license. ASSOCIATE EDITOR: ELIOT H. OHLSTEIN International Union of Basic and Clinical Pharmacology. CV. Somatostatin Receptors: Structure, Function, Ligands, and New Nomenclature Thomas Günther, Giovanni Tulipano, Pascal Dournaud, Corinne Bousquet, Zsolt Csaba, Hans-Jürgen Kreienkamp, Amelie Lupp, Márta Korbonits, Justo P. Castaño, Hans-Jürgen Wester, Michael Culler,1 Shlomo Melmed, and Stefan Schulz Institute of Pharmacology and Toxicology, Jena University Hospital, Friedrich-Schiller-University, Jena, Germany (T.G., A.L., S.S.); Unit of Pharmacology, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy (G.T.); PROTECT, INSERM, Université Paris Diderot, Sorbonne Paris Cité, Paris, France (P.D., Z.C.); Cancer Research Center of Toulouse, INSERM UMR 1037-University Toulouse III Paul Sabatier, Toulouse, France (C.B.); Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany (H.-J.K.); Centre for Endocrinology, William Harvey Research Institute, Barts and London School of Medicine, Queen Mary University of London, London, United Kingdom (M.K.); Maimonides Institute for Biomedical Research of Cordoba, Córdoba, Spain (J.P.C.); Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Córdoba, Spain (J.P. C.); Reina Sofia University Hospital, Córdoba, Spain (J.P.C.); CIBER Fisiopatología de la Obesidad y Nutrición, Córdoba, Spain (J.P.C.); Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany (H.-J.W.); Culler Consulting LLC, Hopkinton, Massachusetts (M.C.); and Pituitary Center, Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California (S.M.) Downloaded from Abstract ...................................................................................765 I.
    [Show full text]
  • Characterisation of L-Cell Secretory Mechanisms and Colonic
    Characterisation of L -cell secretory mechanisms and colonic enteroendocrine cell subpopulations Lawrence Billing Downing College, Cambridge This dissertation is submitted for the degree of Doctor of Philosophy September 2018 Abstract Enteroendocrine cells (EECs) are chemosensitive cells of the gastrointestinal epithelium that exert a wide range of physiological effects via production and secretion of hormones in response to ingested nutrients, bacterial metabolites and systemic signals. Glucagon-like peptide-1 (GLP-1) is one such hormone secreted from so-called L-cells found in both the small and large intestines. GLP-1 exerts an anorexigenic effect and together with glucose- dependent insulinotropic polypeptide (GIP), restores postprandial normoglycaemia through the incretin effect. These effects are exploited by GLP-1 analogues in the treatment of type 2 diabetes. GLP-1 may also contribute to weight-loss and remission of type 2 diabetes following bariatric surgery which increases postprandial GLP-1 excursions. Here we investigated stimulus secretion coupling in L-cells. A novel 2D culture system from murine small intestinal organoids was established as an in vitro model. This was used to characterise synergistic stimulation of GLP-1 secretion in response to concomitant stimulation by bile acids through the Gs-protein coupled receptor GPBAR1 and free fatty acids through the Gq-coupled receptor FFAR1. Roughly half of colonic, but not small intestinal, L-cells co-produce the orexigenic peptide insulin-like peptide 5 (INSL5). This hitherto poorly examined subpopulation of L-cells was characterised through transcriptomic analysis, intracellular calcium imaging (using a novel GCaMP6F-based transgenic mouse model), LC/MS peptide quantification and 3D super resolution microscopy (3D-SIM).
    [Show full text]
  • RT² Profiler PCR Array (384-Well Format) Mouse G Protein Coupled Receptors 384HT
    RT² Profiler PCR Array (384-Well Format) Mouse G Protein Coupled Receptors 384HT Cat. no. 330231 PAMM-3009ZE For pathway expression analysis Format For use with the following real-time cyclers RT² Profiler PCR Array, Applied Biosystems® models 7900HT (384-well block), Format E ViiA™ 7 (384-well block); Bio-Rad CFX384™ RT² Profiler PCR Array, Roche® LightCycler® 480 (384-well block) Format G Description The Mouse G Protein Coupled Receptors 384HT RT² Profiler™ PCR Array profiles the expression of a comprehensive panel of 370 genes encoding the most important G Protein Coupled Receptors (GPCR). GPCR regulate a number of normal biological processes and play roles in the pathophysiology of many diseases upon dysregulation of their downstream signal transduction activities. As a result, they represent 30 percent of the targets for all current drug development. Developing drug screening assays requires a survey of which GPCR the chosen cell-based model system expresses, to determine not only the expression of the target GPCR, but also related GPCR to assess off-target side effects. Expression of other unrelated GPCR (even orphan receptors whose ligand are unknown) may also correlate with off-target side effects. The ligands that bind and activate the receptors on this array include neurotransmitters and neuropeptides, hormones, chemokines and cytokines, lipid signaling molecules, light-sensitive compounds, and odorants and pheromones. The normal biological processes regulated by GPCR include, but are not limited to, behavioral and mood regulation (serotonin, dopamine, GABA, glutamate, and other neurotransmitter receptors), autonomic (sympathetic and parasympathetic) nervous system transmission (blood pressure, heart rate, and digestive processes via hormone receptors), inflammation and immune system regulation (chemokine receptors, histamine receptors), vision (opsins like rhodopsin), and smell (olfactory receptors for odorants and vomeronasal receptors for pheromones).
    [Show full text]