1
|
Meyer CM, Vafaeva O, Low H, Speca DJ, Díaz E. Regulation of hippocampal excitatory synapse development by the adhesion G-protein coupled receptor Brain-specific angiogenesis inhibitor 2 (BAI2/ADGRB2). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.02.636169. [PMID: 39975290 PMCID: PMC11838441 DOI: 10.1101/2025.02.02.636169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Glutamatergic synapses and their associated dendritic spines are critical information processing sites within the brain. Proper development of these specialized cellular junctions is important for normal brain functionality. Synaptic adhesion G protein-coupled receptors (aGPCRs) have been identified as regulators of synapse development and function. While two members of the Brain-specific angiogenesis inhibitor (BAI/ADGRB) subfamily of synaptic aGPCRs, BAI1/ADGRB1 and BAI3/ADGRB3, have been found to mediate synapse and spine formation, BAI2/ADGRB2 function remains uncharacterized at the synapse. Here, we show that endogenous BAI2 is expressed throughout the nervous system with prominent expression in synapse dense regions of the hippocampus. In dissociated hippocampal cultures, BAI2 is highly enriched at large postsynaptic sites, defined by the size of the postsynaptic scaffold PSD95. Loss of BAI2 negatively impacts glutamatergic synapses across development in dissociated hippocampal cultures. In contrast, GABAergic synapse density is unchanged. Furthermore, BAI2 deficient neurons have significant alterations in spine morphology with decreased density of mature PSD95-containing mushroom-shaped spines compared with wild-type neurons. Interestingly, no major alterations in dendritic complexity were observed in BAI2 deficient neurons, in contrast to previous results for the other BAIs. The reduction in mature mushroom-shaped spine is commensurate with a reduction in spine volume and head diameter. Altogether, these results demonstrate that the aGPCR BAI2 is an important regulator of glutamatergic synapse and PSD95-associated spine development in cultured hippocampal neurons. These results expand the knowledge of the BAI subfamily of aGPCRs in mediating excitatory synapse and spine development and highlight differences unique to BAI2.
Collapse
Affiliation(s)
- Christina M. Meyer
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Olga Vafaeva
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Henry Low
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - David J. Speca
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| | - Elva Díaz
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, 95616, USA
| |
Collapse
|
2
|
Kordon SP, Cechova K, Bandekar SJ, Leon K, Dutka P, Siffer G, Kossiakoff AA, Vafabakhsh R, Araç D. Conformational coupling between extracellular and transmembrane domains modulates holo-adhesion GPCR function. Nat Commun 2024; 15:10545. [PMID: 39627215 PMCID: PMC11615224 DOI: 10.1038/s41467-024-54836-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 11/20/2024] [Indexed: 12/06/2024] Open
Abstract
Adhesion G Protein-Coupled Receptors (aGPCRs) are key cell-adhesion molecules involved in numerous physiological functions. aGPCRs have large multi-domain extracellular regions (ECRs) containing a conserved GAIN domain that precedes their seven-pass transmembrane domain (7TM). Ligand binding and mechanical force applied on the ECR regulate receptor function. However, how the ECR communicates with the 7TM remains elusive, because the relative orientation and dynamics of the ECR and 7TM within a holoreceptor is unclear. Here, we describe the cryo-EM reconstruction of an aGPCR, Latrophilin3/ADGRL3, and reveal that the GAIN domain adopts a parallel orientation to the transmembrane region and has constrained movement. Single-molecule FRET experiments unveil three slow-exchanging FRET states of the ECR relative to the transmembrane region within the holoreceptor. GAIN-targeted antibodies, and cancer-associated mutations at the GAIN-7TM interface, alter FRET states, cryo-EM conformations, and receptor signaling. Altogether, this data demonstrates conformational and functional coupling between the ECR and 7TM, suggesting an ECR-mediated mechanism for aGPCR activation.
Collapse
Affiliation(s)
- Szymon P Kordon
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL, USA
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
- Center for Mechanical Excitability, University of Chicago, Chicago, IL, USA
| | - Kristina Cechova
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Sumit J Bandekar
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL, USA
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
- Center for Mechanical Excitability, University of Chicago, Chicago, IL, USA
| | - Katherine Leon
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
- Neuroscience Institute, The University of Chicago, Chicago, IL, USA
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA
| | - Przemysław Dutka
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
- Department of Structural Biology, Genentech, South San Francisco, CA, USA
| | - Gracie Siffer
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Anthony A Kossiakoff
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA
| | - Reza Vafabakhsh
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA.
| | - Demet Araç
- Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL, USA.
- Neuroscience Institute, The University of Chicago, Chicago, IL, USA.
- Institute for Biophysical Dynamics, University of Chicago, Chicago, IL, USA.
- Center for Mechanical Excitability, University of Chicago, Chicago, IL, USA.
| |
Collapse
|
3
|
Huang D, Yu Z, Lu H, Jiang P, Qian X, Han Y, Qian P. Adhesion GPCR ADGRE2 Maintains Proteostasis to Promote Progression in Acute Myeloid Leukemia. Cancer Res 2024; 84:2090-2108. [PMID: 39082681 DOI: 10.1158/0008-5472.can-23-2314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 01/19/2024] [Accepted: 03/28/2024] [Indexed: 08/15/2024]
Abstract
Acute myeloid leukemia (AML) is an aggressive and heterogeneous hematologic malignancy. In elderly patients, AML incidence is high and has a poor prognosis due to a lack of effective therapies. G protein-coupled receptors (GPCR) play integral roles in physiologic processes and human diseases. Particularly, one third of adhesion GPCRs, the second largest group of GPCRs, are highly expressed in hematopoietic stem and progenitor cells or lineage cells. Here, we investigate the role of adhesion GPCRs in AML and whether they could be harnessed as antileukemia targets. Systematic screening of the impact of adhesion GPCRs on AML functionality by bioinformatic and functional analyses revealed high expression of ADGRE2 in AML, particularly in leukemic stem cells, which is associated with poor patient outcomes. Silencing ADGRE2 not only exerts antileukemic effects in AML cell lines and cells derived from patients with AML in vitro, but also delays AML progression in xenograft models in vivo. Mechanistically, ADGRE2 activates phospholipase Cβ/protein kinase C/MEK/ERK signaling to enhance the expression of AP1 and transcriptionally drive the expression of DUSP1, a protein phosphatase. DUSP1 dephosphorylates Ser16 in the J-domain of the co-chaperone DNAJB1, which facilitates the DNAJB1-HSP70 interaction and maintenance of proteostasis in AML. Finally, combined inhibition of MEK, AP1, and DUSP1 exhibits robust therapeutic efficacy in AML xenograft mouse models. Collectively, this study deciphers the roles and mechanisms of ADGRE2 in AML and provides a promising therapeutic strategy for treating AML. Significance: Increased expression of the adhesion GPCR member ADGRE2 in AML supports leukemia stem cell self-renewal and leukemogenesis by modulating proteostasis via an MEK/AP1/DUSP1 axis, which can be targeted to suppress AML progression.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Cell Line, Tumor
- Cell Proliferation
- Disease Progression
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/genetics
- Mice, Inbred NOD
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/pathology
- Proteostasis
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/genetics
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Deyu Huang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Zhejiang University and Zhejiang Provincial Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Zebin Yu
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Zhejiang University and Zhejiang Provincial Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Huan Lu
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Zhejiang University and Zhejiang Provincial Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Penglei Jiang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Zhejiang University and Zhejiang Provincial Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Xinyue Qian
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Zhejiang University and Zhejiang Provincial Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yingli Han
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | - Pengxu Qian
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Center for Stem Cell and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
- State Key Laboratory of Experimental Hematology, Institute of Hematology, Zhejiang University and Zhejiang Provincial Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| |
Collapse
|
4
|
Kordon SP, Cechova K, Bandekar SJ, Leon K, Dutka P, Siffer G, Kossiakoff AA, Vafabakhsh R, Araç D. Structural analysis and conformational dynamics of a holo-adhesion GPCR reveal interplay between extracellular and transmembrane domains. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.25.581807. [PMID: 38464178 PMCID: PMC10925191 DOI: 10.1101/2024.02.25.581807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2024]
Abstract
Adhesion G Protein-Coupled Receptors (aGPCRs) are key cell-adhesion molecules involved in numerous physiological functions. aGPCRs have large multi-domain extracellular regions (ECR) containing a conserved GAIN domain that precedes their seven-pass transmembrane domain (7TM). Ligand binding and mechanical force applied on the ECR regulate receptor function. However, how the ECR communicates with the 7TM remains elusive, because the relative orientation and dynamics of the ECR and 7TM within a holoreceptor is unclear. Here, we describe the cryo-EM reconstruction of an aGPCR, Latrophilin3/ADGRL3, and reveal that the GAIN domain adopts a parallel orientation to the membrane and has constrained movement. Single-molecule FRET experiments unveil three slow-exchanging FRET states of the ECR relative to the 7TM within the holoreceptor. GAIN-targeted antibodies, and cancer-associated mutations at the GAIN-7TM interface, alter FRET states, cryo-EM conformations, and receptor signaling. Altogether, this data demonstrates conformational and functional coupling between the ECR and 7TM, suggesting an ECR-mediated mechanism of aGPCR activation.
Collapse
|
5
|
Scharf MM, Humphrys LJ, Berndt S, Di Pizio A, Lehmann J, Liebscher I, Nicoli A, Niv MY, Peri L, Schihada H, Schulte G. The dark sides of the GPCR tree - research progress on understudied GPCRs. Br J Pharmacol 2024. [PMID: 38339984 DOI: 10.1111/bph.16325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/24/2023] [Accepted: 01/08/2024] [Indexed: 02/12/2024] Open
Abstract
A large portion of the human GPCRome is still in the dark and understudied, consisting even of entire subfamilies of GPCRs such as odorant receptors, class A and C orphans, adhesion GPCRs, Frizzleds and taste receptors. However, it is undeniable that these GPCRs bring an untapped therapeutic potential that should be explored further. Open questions on these GPCRs span diverse topics such as deorphanisation, the development of tool compounds and tools for studying these GPCRs, as well as understanding basic signalling mechanisms. This review gives an overview of the current state of knowledge for each of the diverse subfamilies of understudied receptors regarding their physiological relevance, molecular mechanisms, endogenous ligands and pharmacological tools. Furthermore, it identifies some of the largest knowledge gaps that should be addressed in the foreseeable future and lists some general strategies that might be helpful in this process.
Collapse
Affiliation(s)
- Magdalena M Scharf
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Stockholm, Sweden
| | - Laura J Humphrys
- Institute of Pharmacy, University of Regensburg, Regensburg, Germany
| | - Sandra Berndt
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Antonella Di Pizio
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Life Science, Technical University of Munich, Freising, Germany
| | - Juliane Lehmann
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Alessandro Nicoli
- Leibniz Institute for Food Systems Biology at the Technical University of Munich, Freising, Germany
- Chemoinformatics and Protein Modelling, Department of Molecular Life Science, School of Life Science, Technical University of Munich, Freising, Germany
| | - Masha Y Niv
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Lior Peri
- The Institute of Biochemistry, Food Science and Nutrition, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel
| | - Hannes Schihada
- Institute of Pharmaceutical Chemistry, Philipps-University Marburg, Marburg, Germany
| | - Gunnar Schulte
- Karolinska Institutet, Dept. Physiology & Pharmacology, Sec. Receptor Biology & Signaling, Stockholm, Sweden
| |
Collapse
|
6
|
Adediwura VA, Miao Y. Mechanistic Insights into Peptide Binding and Deactivation of an Adhesion G Protein-Coupled Receptor. Molecules 2023; 29:164. [PMID: 38202747 PMCID: PMC10780249 DOI: 10.3390/molecules29010164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/20/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Adhesion G protein-coupled receptors (ADGRGs) play critical roles in the reproductive, neurological, cardiovascular, and endocrine systems. In particular, ADGRG2 plays a significant role in Ewing sarcoma cell proliferation, parathyroid cell function, and male fertility. In 2022, a cryo-EM structure was reported for the active ADGRG2 bound by an optimized peptide agonist IP15 and the Gs protein. The IP15 peptide agonist was also modified to antagonists 4PH-E and 4PH-D with mutations of the 4PH residue to Glu and Asp, respectively. However, experimental structures of inactive antagonist-bound ADGRs remain to be resolved, and the activation mechanism of ADGRs such as ADGRG2 is poorly understood. Here, we applied Gaussian accelerated molecular dynamics (GaMD) simulations to probe conformational dynamics of the agonist- and antagonist-bound ADGRG2. By performing GaMD simulations, we were able to identify important low-energy conformations of ADGRG2 in the active, intermediate, and inactive states, as well as explore the binding conformations of each peptide. Moreover, our simulations revealed critical peptide-receptor residue interactions during the deactivation of ADGRG2. In conclusion, through GaMD simulations, we uncovered mechanistic insights into peptide (agonist and antagonist) binding and deactivation of the ADGRG2. These findings will potentially facilitate rational design of new peptide modulators of ADGRG2 and other ADGRs.
Collapse
Affiliation(s)
| | - Yinglong Miao
- Department of Pharmacology and Computational Medicine Program, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA;
| |
Collapse
|
7
|
Krasnova OA, Kulakova KA, Sopova JV, Smirnov EY, Silonov SA, Lomert EV, Bystrova OA, Martynova MG, Neganova IE. Essential Role of Adhesion GPCR, GPR123, for Human Pluripotent Stem Cells and Reprogramming towards Pluripotency. Cells 2023; 12:cells12020304. [PMID: 36672239 PMCID: PMC9856511 DOI: 10.3390/cells12020304] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 01/10/2023] [Accepted: 01/11/2023] [Indexed: 01/15/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) are the largest family of cell surface receptors. They modulate key physiological functions and are required in diverse developmental processes including embryogenesis, but their role in pluripotency maintenance and acquisition during the reprogramming towards hiPSCs draws little attention. Meanwhile, it is known that more than 106 GPCRs are overexpressed in human pluripotent stem cells (hPSCs). Previously, to identify novel effectors of reprogramming, we performed a high-throughput RNA interference (RNAi) screening assay and identified adhesion GPCR, GPR123, as a potential reprogramming effector. Its role has not been explored before. Herein, by employing GPR123 RNAi we addressed the role of GPR123 for hPSCs. The suppression of GPR123 in hPSCs leads to the loss of pluripotency and differentiation, impacted colony morphology, accumulation of cells at the G2 phase of the cell cycle, and absence of the scratch closure. Application of the GPR123 RNAi at the initiation stage of reprogramming leads to a decrease in the percentage of the "true" hiPSC colonies, a drop in E-cadherin expression, a decrease in the percentage of NANOG+ nuclei, and the absence of actin cytoskeleton remodeling. Together this leads to the absence of the alkaline-phosphatase-positive hiPSCs colonies on the 18th day of the reprogramming process. Overall, these data indicate for the first time the essential role of GPR123 in the maintenance and acquisition of pluripotency.
Collapse
Affiliation(s)
- Olga A. Krasnova
- Laboratory of Molecular Medicine, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St-Petersburg, Russia
| | - Karina A. Kulakova
- Laboratory of Molecular Medicine, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St-Petersburg, Russia
| | - Julia V. Sopova
- Laboratory of Molecular Medicine, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St-Petersburg, Russia
- Center of Transgenesis and Genome Editing, St. Petersburg State University, Universitetskaja Emb., 7/9, 199034 St-Petersburg, Russia
| | - Evgenyi Y. Smirnov
- Laboratory of Regulation of Genes Function, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St-Petersburg, Russia
| | - Sergey A. Silonov
- Laboratory of Structural Dynamics, Stability and Folding of Proteins, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St-Petersburg, Russia
| | - Ekaterina V. Lomert
- Laboratory of Molecular Genetics of Tumor Cells, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St-Petersburg, Russia
| | - Olga A. Bystrova
- Laboratory of Cell Morphology, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St-Petersburg, Russia
| | - Marina G. Martynova
- Laboratory of Cell Morphology, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St-Petersburg, Russia
| | - Irina E. Neganova
- Laboratory of Molecular Medicine, Institute of Cytology of the Russian Academy of Sciences, Tikhoretsky Ave. 4, 194064 St-Petersburg, Russia
- Correspondence:
| |
Collapse
|
8
|
Stephan G, Frenster JD, Liebscher I, Placantonakis DG. Activation of the adhesion G protein-coupled receptor GPR133 by antibodies targeting its N-terminus. J Biol Chem 2022; 298:101949. [PMID: 35447113 PMCID: PMC9133650 DOI: 10.1016/j.jbc.2022.101949] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 04/03/2022] [Accepted: 04/05/2022] [Indexed: 11/28/2022] Open
Abstract
We recently demonstrated that GPR133 (ADGRD1), an adhesion G protein-coupled receptor involved in raising cytosolic cAMP levels, is necessary for growth of glioblastoma (GBM) and is de novo expressed in GBM relative to normal brain tissue. Our previous work suggested that dissociation of autoproteolytically generated N-terminal and C-terminal fragments of GPR133 at the plasma membrane correlates with receptor activation and signaling. To promote the goal of developing biologics that modulate GPR133 function, we investigated the effects of antibodies against the N-terminus of GPR133 on receptor signaling. Here, we show that treatment of HEK293T cells overexpressing GPR133 with these antibodies increased cAMP levels in a concentration-dependent manner. Analysis of culture medium following antibody treatment further indicated the presence of complexes of these antibodies with the autoproteolytically cleaved N-terminal fragments of GPR133. In addition, cells expressing a cleavage-deficient mutant of GPR133 (H543R) did not respond to antibody stimulation, suggesting that the effect is cleavage dependent. Finally, we demonstrate the antibody-mediated stimulation of WT GPR133, but not the cleavage-deficient H543R mutant, was reproducible in patient-derived GBM cells. These findings provide a paradigm for modulation of GPR133 function with biologics and support the hypothesis that the intramolecular cleavage in the N-terminus modulates receptor activation and signaling.
Collapse
Affiliation(s)
- Gabriele Stephan
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, New York, USA.
| | - Joshua D Frenster
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, New York, USA
| | - Ines Liebscher
- Rudolf Schönheimer Institute for Biochemistry, Molecular Biochemistry, University of Leipzig, Leipzig, Germany
| | - Dimitris G Placantonakis
- Department of Neurosurgery, NYU Grossman School of Medicine, New York, New York, USA; Kimmel Center for Stem Cell Biology, NYU Grossman School of Medicine, New York, New York, USA; Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York, USA; Brain and Spine Tumor Center, NYU Grossman School of Medicine, New York, New York, USA; Neuroscience Institute, NYU Grossman School of Medicine, New York, New York, USA.
| |
Collapse
|
9
|
Wilde C, Mitgau J, Suchý T, Schoeneberg T, Liebscher I. Translating the Force - mechano-sensing GPCRs. Am J Physiol Cell Physiol 2022; 322:C1047-C1060. [PMID: 35417266 DOI: 10.1152/ajpcell.00465.2021] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Incorporating mechanical cues into cellular responses allows us to experience our direct environment. Specialized cells can perceive and discriminate between different physical properties such as level of vibration, temperature, or pressure. Mechanical forces are abundant signals that also shape general cellular responses such as cytoskeletal rearrangement, differentiation, or migration and contribute to tissue development and function. The molecular structures that perceive and transduce mechanical forces are specialized cytoskeletal proteins, cell junction molecules, and membrane proteins such as ion channels and metabotropic receptors. G protein-coupled receptors (GPCRs) have attracted attention as metabotropic force receptors as they are among the most important drug targets. This review summarizes the function of mechano-sensitive GPCRs, specifically, the angiotensin II type 1 receptor and adrenergic, apelin, histamine, parathyroid hormone 1, and orphan receptors, focusing particularly on the advanced knowledge gained from adhesion-type GPCRs. We distinguish between shear stress and cell swelling/stretch as the two major types of mechano-activation of these receptors and contemplate the potential contribution of the force-from-lipid and force-from-tether models that have previously been suggested for ion channels.
Collapse
Affiliation(s)
- Caroline Wilde
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Jakob Mitgau
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Tomás Suchý
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Torsten Schoeneberg
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Medical Faculty, Leipzig University, Germany
| |
Collapse
|
10
|
Ng KF, Chen TC, Stacey M, Lin HH. Role of ADGRG1/GPR56 in Tumor Progression. Cells 2021; 10:cells10123352. [PMID: 34943858 PMCID: PMC8699533 DOI: 10.3390/cells10123352] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Accepted: 11/23/2021] [Indexed: 12/13/2022] Open
Abstract
Cellular communication plays a critical role in diverse aspects of tumorigenesis including tumor cell growth/death, adhesion/detachment, migration/invasion, angiogenesis, and metastasis. G protein-coupled receptors (GPCRs) which constitute the largest group of cell surface receptors are known to play fundamental roles in all these processes. When considering the importance of GPCRs in tumorigenesis, the adhesion GPCRs (aGPCRs) are unique due to their hybrid structural organization of a long extracellular cell-adhesive domain and a seven-transmembrane signaling domain. Indeed, aGPCRs have been increasingly shown to be associated with tumor development by participating in tumor cell interaction and signaling. ADGRG1/GPR56, a representative tumor-associated aGPCR, is recognized as a potential biomarker/prognostic factor of specific cancer types with both tumor-suppressive and tumor-promoting functions. We summarize herein the latest findings of the role of ADGRG1/GPR56 in tumor progression.
Collapse
Affiliation(s)
- Kwai-Fong Ng
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; (K.-F.N.); (T.-C.C.)
| | - Tse-Ching Chen
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; (K.-F.N.); (T.-C.C.)
| | - Martin Stacey
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds, Leeds LS2 9JT, UK;
| | - Hsi-Hsien Lin
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan 33305, Taiwan; (K.-F.N.); (T.-C.C.)
- Division of Rheumatology, Allergy, and Immunology, Chang Gung Memorial Hospital-Keelung, Keelung 20401, Taiwan
- Center for Medical and Clinical Immunology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Correspondence:
| |
Collapse
|
11
|
Torregrosa-Carrión R, Piñeiro-Sabarís R, Siguero-Álvarez M, Grego-Bessa J, Luna-Zurita L, Fernandes VS, MacGrogan D, Stainier DYR, de la Pompa JL. Adhesion G protein-coupled receptor Gpr126/Adgrg6 is essential for placental development. SCIENCE ADVANCES 2021; 7:eabj5445. [PMID: 34767447 PMCID: PMC8589310 DOI: 10.1126/sciadv.abj5445] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
Mutations in the G protein–coupled receptor GPR126/ADGRG6 cause human diseases, including defective peripheral nervous system (PNS) myelination. To study GPR126 function, we generated new genetic mice and zebrafish models. Murine Gpr126 is expressed in developing heart endocardium, and global Gpr126 inactivation is embryonically lethal, with mutants having thin-walled ventricles but unaffected heart patterning or maturation. Endocardial-specific Gpr126 deletion does not affect heart development or function, and transgenic endocardial GPR126 expression fails to rescue lethality in Gpr126-null mice. Zebrafish gpr126 mutants display unaffected heart development. Gpr126 is also expressed in placental trophoblast giant cells. Gpr126-null mice with a heterozygous placenta survive but exhibit GPR126-defective PNS phenotype. In contrast, Gpr126-null embryos with homozygous mutant placenta die but are rescued by placental GPR126 expression. Gpr126-deficient placentas display down-regulation of preeclampsia markers Mmp9, Cts7, and Cts8. We propose that the placenta-heart axis accounts for heart abnormalities secondary to placental defects in Gpr126 mutants.
Collapse
Affiliation(s)
- Rebeca Torregrosa-Carrión
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Rebeca Piñeiro-Sabarís
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Marcos Siguero-Álvarez
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Joaquím Grego-Bessa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Luis Luna-Zurita
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Vitor Samuel Fernandes
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Donal MacGrogan
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Didier Y. R. Stainier
- Department of Developmental Genetics, Max Planck Institute for Heart and Lung Research, 61231 Bad Nauheim, Germany
| | - José Luis de la Pompa
- Intercellular Signalling in Cardiovascular Development and Disease Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
- Ciber de Enfermedades Cardiovasculares, 28029 Madrid, Spain
- Corresponding author.
| |
Collapse
|
12
|
Rosa M, Noel T, Harris M, Ladds G. Emerging roles of adhesion G protein-coupled receptors. Biochem Soc Trans 2021; 49:1695-1709. [PMID: 34282836 PMCID: PMC8421042 DOI: 10.1042/bst20201144] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/17/2021] [Accepted: 06/18/2021] [Indexed: 12/24/2022]
Abstract
Adhesion G protein-coupled receptors (aGPCRs) form a sub-group within the GPCR superfamily. Their distinctive structure contains an abnormally large N-terminal, extracellular region with a GPCR autoproteolysis-inducing (GAIN) domain. In most aGPCRs, the GAIN domain constitutively cleaves the receptor into two fragments. This process is often required for aGPCR signalling. Over the last two decades, much research has focussed on aGPCR-ligand interactions, in an attempt to deorphanize the family. Most ligands have been found to bind to regions N-terminal to the GAIN domain. These receptors may bind a variety of ligands, ranging across membrane-bound proteins and extracellular matrix components. Recent advancements have revealed a conserved method of aGPCR activation involving a tethered ligand within the GAIN domain. Evidence for this comes from increased activity in receptor mutants exposing the tethered ligand. As a result, G protein-coupling partners of aGPCRs have been more extensively characterised, making use of their tethered ligand to create constitutively active mutants. This has led to demonstrations of aGPCR function in, for example, neurodevelopment and tumour growth. However, questions remain around the ligands that may bind many aGPCRs, how this binding is translated into changes in the GAIN domain, and the exact mechanism of aGPCR activation following GAIN domain conformational changes. This review aims to examine the current knowledge around aGPCR activation, including ligand binding sites, the mechanism of GAIN domain-mediated receptor activation and how aGPCR transmembrane domains may relate to activation. Other aspects of aGPCR signalling will be touched upon, such as downstream effectors and physiological roles.
Collapse
Affiliation(s)
- Matthew Rosa
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Timothy Noel
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Matthew Harris
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge CB2 1PD, U.K
| |
Collapse
|
13
|
Genome-Wide Association Study Identifies 12 Loci Associated with Body Weight at Age 8 Weeks in Korean Native Chickens. Genes (Basel) 2021; 12:genes12081170. [PMID: 34440344 PMCID: PMC8394794 DOI: 10.3390/genes12081170] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 07/24/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023] Open
Abstract
Meat from Korean native chickens (KNCs) has high consumer demand; however, slow growth performance and high variation in body weight (BW) of KNCs remain an issue. Genome-wide association study (GWAS) is a powerful method to identify quantitative trait-associated genomic loci. A GWAS, based on a large-scale KNC population, is needed to identify underlying genetic mechanisms related to its growth traits. To identify BW-associated genomic regions, we performed a GWAS using the chicken 60K single nucleotide polymorphism (SNP) panel for 1328 KNCs. BW was measured at 8 weeks of age, from 2018 to 2020. Twelve SNPs were associated with BW at the suggestive significance level (p < 2.95 × 10−5) and located near or within 11 candidate genes, including WDR37, KCNIP4, SLIT2, PPARGC1A, MYOCD and ADGRA3. Gene set enrichment analysis based on the GWAS results at p < 0.05 (1680 SNPs) showed that 32 Gene Ontology terms and two Kyoto Encyclopedia of Genes and Genomes pathways, including regulation of transcription, motor activity, the mitogen-activated protein kinase signaling pathway, and tight junction, were significantly enriched (p < 0.05) for BW-associated genes. These pathways are involved in cell growth and development, related to BW gain. The identified SNPs are potential biomarkers in KNC breeding.
Collapse
|
14
|
Conserved residues in the extracellular loop 2 regulate Stachel-mediated activation of ADGRG2. Sci Rep 2021; 11:14060. [PMID: 34234254 PMCID: PMC8263569 DOI: 10.1038/s41598-021-93577-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022] Open
Abstract
Cleavage and dissociation of a large N-terminal fragment and the consequent unmasking of a short sequence (Stachel) remaining on the N-terminus have been proposed as mechanisms of activation of some members of the adhesion G protein-coupled receptor (aGPCR) family. However, the identity of residues that play a role in the activation of aGPCRs by the cognate Stachel remains largely unknown. Protein sequence alignments revealed a conserved stretch of residues in the extracellular loop 2 (ECL2) of all 33 members of the aGPCR family. ADGRG2, an orphan aGPCR, plays a major role in male fertility, Ewing sarcoma cell proliferation, and parathyroid cell function. We used ADGRG2 as a model aGPCR and generated mutants of the conserved residues in the ECL2 via site-directed mutagenesis. We show that tryptophan and isoleucine in the ECL2 are essential for receptor stability and surface expression in the HEK293 cells. By adjusting the receptor surface expression levels, we show that mutation of these residues of ECL2 ablates the Stachel-mediated activation of multiple signaling pathways of ADGRG2. This study provides a novel understanding of the role of the ECL2 in Stachel-mediated signaling and degradation of ADGRG2, which may lay the foundation for the rational design of therapeutics to target aGPCRs.
Collapse
|
15
|
Olaniru OE, Cheng J, Ast J, Arvaniti A, Atanes P, Huang GC, King AJF, Jones PM, Broichhagen J, Hodson DJ, Persaud SJ. SNAP-tag-enabled super-resolution imaging reveals constitutive and agonist-dependent trafficking of GPR56 in pancreatic β-cells. Mol Metab 2021; 53:101285. [PMID: 34224919 PMCID: PMC8326393 DOI: 10.1016/j.molmet.2021.101285] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/19/2021] [Accepted: 06/28/2021] [Indexed: 12/25/2022] Open
Abstract
Objective Members of the adhesion G protein-coupled receptor (aGPCR) subfamily are important actors in metabolic processes, with GPR56 (ADGRG1) emerging as a possible target for type 2 diabetes therapy. GPR56 can be activated by collagen III, its endogenous ligand, and by a synthetic seven amino-acid peptide (TYFAVLM; P7) contained within the GPR56 Stachel sequence. However, the mechanisms regulating GPR56 trafficking dynamics and agonist activities are not yet clear. Methods Here, we introduced SNAPf-tag into the N-terminal segment of GPR56 to monitor GPR56 cellular activity in situ. Confocal and super-resolution microscopy were used to investigate the trafficking pattern of GPR56 in native MIN6 β-cells and in MIN6 β-cells where GPR56 had been deleted by CRISPR-Cas9 gene editing. Insulin secretion, changes in intracellular calcium, and β-cell apoptosis were determined by radioimmunoassay, single-cell calcium microfluorimetry, and measuring caspase 3/7 activities, respectively, in MIN6 β-cells and human islets. Results SNAP-tag labelling indicated that GPR56 predominantly underwent constitutive internalisation in the absence of an exogenous agonist, unlike GLP-1R. Collagen III further stimulated GPR56 internalisation, whereas P7 was without significant effect. The overexpression of GPR56 in MIN6 β-cells did not affect insulin secretion. However, it was associated with reduced β-cell apoptosis, while the deletion of GPR56 made MIN6 β-cells more susceptible to cytokine-induced apoptosis. P7 induced a rapid increase in the intracellular calcium in MIN6 β-cells (in a GPR56-dependent manner) and human islets, and it also caused a sustained and reversible increase in insulin secretion from human islets. Collagen III protected human islets from cytokine-induced apoptosis, while P7 was without significant effect. Conclusions These data indicate that GPR56 exhibits both agonist-dependent and -independent trafficking in β-cells and suggest that while GPR56 undergoes constitutive signalling, it can also respond to its ligands when required. We have also identified that constitutive and agonist-dependent GPR56 activation is coupled to protect β-cells against apoptosis, offering a potential therapeutic target to maintain β-cell mass in type 2 diabetes. GPR56 predominantly underwent constitutive internalisation in β-cells in the absence of exogenous agonist. The GPR56 agonists, collagen III and P7, showed differential effects on GPR56 trafficking and islet functions. Constitutive and agonist-dependent GPR56 activation is coupled to protection of β-cells against apoptosis.
Collapse
Affiliation(s)
- Oladapo E Olaniru
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK.
| | - Jordan Cheng
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering & Imaging Sciences, 4th floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, UK
| | - Julia Ast
- Institute of Metabolism and Systems Research (IMSR), Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Anastasia Arvaniti
- Institute of Metabolism and Systems Research (IMSR), Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Patricio Atanes
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Guo C Huang
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Aileen J F King
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Peter M Jones
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Johannes Broichhagen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP), Robert-Rössle-Str. 10, 13125, Berlin, Germany
| | - David J Hodson
- Institute of Metabolism and Systems Research (IMSR), Centre of Membrane Proteins and Receptors (COMPARE), University of Birmingham, Birmingham, UK; Centre for Endocrinology, Diabetes and Metabolism, Birmingham Health Partners, Birmingham, UK
| | - Shanta J Persaud
- Department of Diabetes, School of Life Course Sciences, King's College London, Guy's Campus, London SE1 1UL, UK.
| |
Collapse
|
16
|
Adhesion receptor ADGRG2/GPR64 is in the GI-tract selectively expressed in mature intestinal tuft cells. Mol Metab 2021; 51:101231. [PMID: 33831593 PMCID: PMC8105302 DOI: 10.1016/j.molmet.2021.101231] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/27/2021] [Accepted: 03/31/2021] [Indexed: 11/23/2022] Open
Abstract
Objective GPR64/ADGRG2 is an orphan Adhesion G protein-coupled receptor (ADGR) known to be mainly expressed in the parathyroid gland and epididymis. This investigation aimed to delineate the cellular expression of GPR64 throughout the body with focus on the gastrointestinal (GI) tract. Methods Transgenic Gpr64mCherry reporter mice were histologically examined throughout the body and reporter protein expression in intestinal tuft cells was confirmed by specific cell ablation. The GPCR repertoire of intestinal Gpr64mCherry-positive tuft cells was analyzed by quantitative RT-PCR analysis and in situ hybridization. The Gpr64mCherry was crossed into the general tuft cell reporter Trpm5GFP to generate small intestinal organoids for time-lapse imaging. Intestinal tuft cells were isolated from small intestine, FACS-purified and transcriptionally compared using RNA-seq analysis. Results Expression of the Gpr64mCherry reporter was identified in multiple organs and specifically in olfactory microvillous cells, enteric nerves, and importantly in respiratory and GI tuft cells. In the small intestine, cell ablation targeting Gpr64-expressing epithelial cells eliminated tuft cells. Transcriptional analysis of small intestinal Gpr64mCherry -positive tuft cells confirmed expression of Gpr64 and the chemo-sensors Sucnr1, Gprc5c, Drd3, and Gpr41/Ffar3. Time-lapse studies of organoids from Trpm5GFP:Gpr64mCherry mice revealed sequential expression of initially Trpm5GFP and subsequently also Gpr64mCherry in maturing intestinal tuft cells. RNA-seq analysis of small intestinal tuft cells based on these two markers demonstrated a dynamic change in expression of transcription factors and GPCRs from young to mature tuft cells. Conclusions GPR64 is expressed in chemosensory epithelial cells across a broad range of tissues; however, in the GI tract, GPR64 is remarkably selectively expressed in mature versus young immunoregulatory tuft cells. GPR64-RFP is expressed mainly in chemosensory epithelial cells, including tuft cells. Maturing intestinal tuft cells initially express Trpm5 and subsequently Gpr64. Mature intestinal Gpr64+ tuft cells express Sucnr1, Gprc5c, Drd3, and Gpr41/Ffar3. RNAseq analysis reveals dynamic transcriptional change of mature vs. young tuft cells.
Collapse
|
17
|
Schöneberg T, Liebscher I. Mutations in G Protein-Coupled Receptors: Mechanisms, Pathophysiology and Potential Therapeutic Approaches. Pharmacol Rev 2021; 73:89-119. [PMID: 33219147 DOI: 10.1124/pharmrev.120.000011] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
There are approximately 800 annotated G protein-coupled receptor (GPCR) genes, making these membrane receptors members of the most abundant gene family in the human genome. Besides being involved in manifold physiologic functions and serving as important pharmacotherapeutic targets, mutations in 55 GPCR genes cause about 66 inherited monogenic diseases in humans. Alterations of nine GPCR genes are causatively involved in inherited digenic diseases. In addition to classic gain- and loss-of-function variants, other aspects, such as biased signaling, trans-signaling, ectopic expression, allele variants of GPCRs, pseudogenes, gene fusion, and gene dosage, contribute to the repertoire of GPCR dysfunctions. However, the spectrum of alterations and GPCR involvement is probably much larger because an additional 91 GPCR genes contain homozygous or hemizygous loss-of-function mutations in human individuals with currently unidentified phenotypes. This review highlights the complexity of genomic alteration of GPCR genes as well as their functional consequences and discusses derived therapeutic approaches. SIGNIFICANCE STATEMENT: With the advent of new transgenic and sequencing technologies, the number of monogenic diseases related to G protein-coupled receptor (GPCR) mutants has significantly increased, and our understanding of the functional impact of certain kinds of mutations has substantially improved. Besides the classical gain- and loss-of-function alterations, additional aspects, such as biased signaling, trans-signaling, ectopic expression, allele variants of GPCRs, uniparental disomy, pseudogenes, gene fusion, and gene dosage, need to be elaborated in light of GPCR dysfunctions and possible therapeutic strategies.
Collapse
Affiliation(s)
- Torsten Schöneberg
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig, Germany
| | - Ines Liebscher
- Rudolf Schönheimer Institute of Biochemistry, Molecular Biochemistry, Medical Faculty, Leipzig, Germany
| |
Collapse
|
18
|
Araújo CAC, Pacheco JPF, Waniek PJ, Geraldo RB, Sibajev A, Dos Santos AL, Evangelho VGO, Dyson PJ, Azambuja P, Ratcliffe NA, Castro HC, Mello CB. A rhamnose-binding lectin from Rhodnius prolixus and the impact of its silencing on gut bacterial microbiota and Trypanosoma cruzi. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 114:103823. [PMID: 32800901 DOI: 10.1016/j.dci.2020.103823] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/31/2020] [Accepted: 08/03/2020] [Indexed: 06/11/2023]
Abstract
Lectins are ubiquitous proteins involved in the immune defenses of different organisms and mainly responsible for non-self-recognition and agglutination reactions. This work describes molecular and biological characterization of a rhamnose-binding lectin (RBL) from Rhodnius prolixus, which possesses a 21 amino acid signal peptide and a mature protein of 34.6 kDa. The in-silico analysis of the primary and secondary structures of RpLec revealed a lectin domain fully conserved among previous insects studied. The three-dimensional homology model of RpLec was similar to other RBL-lectins. Docking predictions with the monosaccharides showed rhamnose and galactose-binding sites comparable to Latrophilin-1 and N-Acetylgalactosamine-binding in a different site. The effects of RpLec gene silencing on levels of infecting Trypanosoma cruzi Dm 28c and intestinal bacterial populations in the R. prolixus midgut were studied by injecting RpLec dsRNA into the R. prolixus hemocoel. Whereas T. cruzi numbers remained unchanged compared with the controls, numbers of bacteria increased significantly. The silencing also induced the up regulation of the R. prolixus defC (defensin) expression gene. These results with RpLec reveal the potential importance of this little studied molecule in the insect vector immune response and homeostasis of the gut bacterial microbiota.
Collapse
Affiliation(s)
- C A C Araújo
- Programa de Pós-Graduação Em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil
| | - J P F Pacheco
- Programa de Pós-Graduação Em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil; Laboratório de Biologia de Insetos, Departamento de Biologia Geral, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil
| | - P J Waniek
- Programa de Pós-Graduação Em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil
| | - R B Geraldo
- Programa de Pós-Graduação Em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil
| | - A Sibajev
- Centro de Ciências da Saúde, Universidade Federal de Roraima, Av. Cap. Enê Garcez 2413, Boa Vista, RR, CEP 69400-000, Brazil
| | - A L Dos Santos
- Programa de Pós-Graduação Em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil
| | - V G O Evangelho
- Programa de Pós-Graduação Em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil
| | - P J Dyson
- Institute of Life Science, School of Medicine, Swansea University, Singleton Park, Swansea, SA2 8PP, UK
| | - P Azambuja
- Programa de Pós-Graduação Em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil; Laboratório de Bioquímica e Fisiologia de Insetos, Instituto Oswaldo Cruz, Fundação, Oswaldo Cruz, Fiocruz, Av. Brasil 4365, Rio de Janeiro, RJ, CEP 21045-900, Brazil; Instituto Nacional de Ciência e Tecnologia Em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil
| | - N A Ratcliffe
- Programa de Pós-Graduação Em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil; Department of Biosciences, Swansea University, Singleton Park, Swansea, SA28PP, UK
| | - H C Castro
- Programa de Pós-Graduação Em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil.
| | - C B Mello
- Programa de Pós-Graduação Em Ciências e Biotecnologia, Instituto de Biologia, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil; Laboratório de Biologia de Insetos, Departamento de Biologia Geral, Universidade Federal Fluminense, Campus Do Gragoatá, Bloco M, São Domingos, Niterói, Rio de Janeiro, RJ, CEP 24201-201, Brazil; Instituto Nacional de Ciência e Tecnologia Em Entomologia Molecular (INCT-EM), Rio de Janeiro, Brazil.
| |
Collapse
|
19
|
Sun Y, Zhang D, Ma ML, Lin H, Song Y, Wang J, Ma C, Yu K, An W, Guo S, He D, Yang Z, Xiao P, Hou G, Yu X, Sun JP. Optimization of a peptide ligand for the adhesion GPCR ADGRG2 provides a potent tool to explore receptor biology. J Biol Chem 2020; 296:100174. [PMID: 33303626 PMCID: PMC7948503 DOI: 10.1074/jbc.ra120.014726] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 12/06/2020] [Accepted: 12/10/2020] [Indexed: 12/11/2022] Open
Abstract
The adhesion GPCR ADGRG2, also known as GPR64, is a critical regulator of male fertility that maintains ion/pH homeostasis and CFTR coupling. The molecular basis of ADGRG2 function is poorly understood, in part because no endogenous ligands for ADGRG2 have been reported, thus limiting the tools available to interrogate ADGRG2 activity. It has been shown that ADGRG2 can be activated by a peptide, termed p15, derived from its own N-terminal region known as the Stachel sequence. However, the low affinity of p15 limits its utility for ADGRG2 characterization. In the current study, we used alanine scanning mutagenesis to examine the critical residues responsible for p15-induced ADGRG2 activity. We next designed systematic strategies to optimize the peptide agonist of ADGRG2, using natural and unnatural amino acid substitutions. We obtained an optimized ADGRG2 Stachel peptide T1V/F3Phe(4-Me) (VPM-p15) that activated ADGRG2 with significantly improved (>2 orders of magnitude) affinity. We then characterized the residues in ADGRG2 that were important for ADGRG2 activation in response to VPM-p15 engagement, finding that the toggle switch W6.53 and residues of the ECL2 region of ADGRG2 are key determinants for VPM-p15 interactions and VPM-p15-induced Gs or arrestin signaling. Our study not only provides a useful tool to investigate the function of ADGRG2 but also offers new insights to guide further optimization of Stachel peptides to activate adhesion GPCR members.
Collapse
Affiliation(s)
- Yujing Sun
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China; Department of Endocrinology, Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Daolai Zhang
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Ming-Liang Ma
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Hui Lin
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Youchen Song
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Junyan Wang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Chuanshun Ma
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Ke Yu
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Wentao An
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Shengchao Guo
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Dongfang He
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Peng Xiao
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China
| | - Guige Hou
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China
| | - Xiao Yu
- Key Laboratory Experimental Teratology of the Ministry of Education and Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, China.
| | - Jin-Peng Sun
- School of Pharmacy, Binzhou Medical University, Yantai, Shandong, China; Key Laboratory Experimental Teratology of the Ministry of Education and Department of Biochemistry and Molecular Biology, Shandong University School of Medicine, Jinan, Shandong, China; Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University, Ministry of Education, Beijing, China.
| |
Collapse
|
20
|
Dannhäuser S, Lux TJ, Hu C, Selcho M, Chen JTC, Ehmann N, Sachidanandan D, Stopp S, Pauls D, Pawlak M, Langenhan T, Soba P, Rittner HL, Kittel RJ. Antinociceptive modulation by the adhesion GPCR CIRL promotes mechanosensory signal discrimination. eLife 2020; 9:e56738. [PMID: 32996461 PMCID: PMC7546736 DOI: 10.7554/elife.56738] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 09/17/2020] [Indexed: 12/17/2022] Open
Abstract
Adhesion-type GPCRs (aGPCRs) participate in a vast range of physiological processes. Their frequent association with mechanosensitive functions suggests that processing of mechanical stimuli may be a common feature of this receptor family. Previously, we reported that the Drosophila aGPCR CIRL sensitizes sensory responses to gentle touch and sound by amplifying signal transduction in low-threshold mechanoreceptors (Scholz et al., 2017). Here, we show that Cirl is also expressed in high-threshold mechanical nociceptors where it adjusts nocifensive behaviour under physiological and pathological conditions. Optogenetic in vivo experiments indicate that CIRL lowers cAMP levels in both mechanosensory submodalities. However, contrasting its role in touch-sensitive neurons, CIRL dampens the response of nociceptors to mechanical stimulation. Consistent with this finding, rat nociceptors display decreased Cirl1 expression during allodynia. Thus, cAMP-downregulation by CIRL exerts opposing effects on low-threshold mechanosensors and high-threshold nociceptors. This intriguing bipolar action facilitates the separation of mechanosensory signals carrying different physiological information.
Collapse
Affiliation(s)
- Sven Dannhäuser
- Department of Animal Physiology, Institute of Biology, Leipzig UniversityLeipzigGermany
- Carl-Ludwig-Institute for Physiology, Leipzig UniversityLeipzigGermany
| | - Thomas J Lux
- Center for Interdisciplinary Pain Medicine, Department of Anaesthesiology, University Hospital WürzburgWürzburgGermany
| | - Chun Hu
- Neuronal Patterning and Connectivity, Center for Molecular Neurobiology, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Mareike Selcho
- Department of Animal Physiology, Institute of Biology, Leipzig UniversityLeipzigGermany
- Carl-Ludwig-Institute for Physiology, Leipzig UniversityLeipzigGermany
| | - Jeremy T-C Chen
- Center for Interdisciplinary Pain Medicine, Department of Anaesthesiology, University Hospital WürzburgWürzburgGermany
| | - Nadine Ehmann
- Department of Animal Physiology, Institute of Biology, Leipzig UniversityLeipzigGermany
- Carl-Ludwig-Institute for Physiology, Leipzig UniversityLeipzigGermany
| | - Divya Sachidanandan
- Department of Animal Physiology, Institute of Biology, Leipzig UniversityLeipzigGermany
- Carl-Ludwig-Institute for Physiology, Leipzig UniversityLeipzigGermany
| | - Sarah Stopp
- Department of Animal Physiology, Institute of Biology, Leipzig UniversityLeipzigGermany
- Carl-Ludwig-Institute for Physiology, Leipzig UniversityLeipzigGermany
| | - Dennis Pauls
- Department of Animal Physiology, Institute of Biology, Leipzig UniversityLeipzigGermany
- Carl-Ludwig-Institute for Physiology, Leipzig UniversityLeipzigGermany
| | - Matthias Pawlak
- Department of Neurophysiology, Institute of Physiology, University of WürzburgWürzburgGermany
| | - Tobias Langenhan
- Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig UniversityLeipzigGermany
| | - Peter Soba
- Neuronal Patterning and Connectivity, Center for Molecular Neurobiology, University Medical Center Hamburg-EppendorfHamburgGermany
| | - Heike L Rittner
- Center for Interdisciplinary Pain Medicine, Department of Anaesthesiology, University Hospital WürzburgWürzburgGermany
| | - Robert J Kittel
- Department of Animal Physiology, Institute of Biology, Leipzig UniversityLeipzigGermany
- Carl-Ludwig-Institute for Physiology, Leipzig UniversityLeipzigGermany
| |
Collapse
|
21
|
G Protein-Coupling of Adhesion GPCRs ADGRE2/EMR2 and ADGRE5/CD97, and Activation of G Protein Signalling by an Anti-EMR2 Antibody. Sci Rep 2020; 10:1004. [PMID: 31969668 PMCID: PMC6976652 DOI: 10.1038/s41598-020-57989-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 01/02/2020] [Indexed: 11/25/2022] Open
Abstract
The experimental evidence that Adhesion G Protein-Coupled Receptors (aGPCRs) functionally couple to heterotrimeric G proteins has been emerging in incremental steps, but attributing biological significance to their G protein signalling function still presents a major challenge. Here, utilising activated truncated forms of the receptors, we show that ADGRE2/EMR2 and ADGRE5/CD97 are G protein-coupled in a variety of recombinant systems. In a yeast-based assay, where heterologous GPCRs are coupled to chimeric G proteins, EMR2 showed broad G protein-coupling, whereas CD97 coupled more specifically to Gα12, Gα13, Gα14 and Gαz chimeras. Both receptors induced pertussis-toxin (PTX) insensitive inhibition of cyclic AMP (cAMP) levels in mammalian cells, suggesting coupling to Gαz. EMR2 was shown to signal via Gα16, and via a Gα16/Gαz chimera, to stimulate IP1 accumulation. Finally, using an NFAT reporter assay, we identified a polyclonal antibody that activates EMR2 G protein signalling in vitro. Our results highlight the potential for the development of soluble agonists to understand further the biological effects and therapeutic opportunities for ADGRE receptor-mediated G protein signalling.
Collapse
|
22
|
Azimzadeh P, Talamantez-Lyburn SC, Chang KT, Inoue A, Balenga N. Spatial regulation of GPR64/ADGRG2 signaling by β-arrestins and GPCR kinases. Ann N Y Acad Sci 2019; 1456:26-43. [PMID: 31502283 DOI: 10.1111/nyas.14227] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 08/05/2019] [Accepted: 08/13/2019] [Indexed: 12/28/2022]
Abstract
Mechanisms of activation, signaling, and trafficking of adhesion G protein-coupled receptors (aGPCRs) have remained largely unknown. Several aGPCRs, including GPR56/ADGRG1 and GPR64/ADGRG2, show increased activity in the absence of their N-terminal fragment (NTF). This constitutive signaling is plausibly caused by the binding of extracellular N-terminal 15-25 amino acid-long tethered agonist to extracellular domains of the cognate aGPCRs. To test the role of NTF and tethered agonist in GPR64 signaling and endocytosis, we generated mutants that lack either NTF alone (ΔNTF) or NTF and tethered agonist (P622). We discover that unlike full-length GPR64, ΔNTF and P622 mutants interact with β-arrestin1 and β-arrestins2 and are constitutively internalized in steady states. However, only ΔNTF shows exaggerated basal activation of the Gαs -cAMP-CRE signaling cascade. Neither ΔNTF nor P622 shows constitutive activation of the Gα13 -SRE pathway, but both mutants respond to exogenously added agonistic peptide via CRE and SRE. GPCR kinases and dynamin mediate the constitutive internalization of ΔNTF and P622 to early endosomes, where ΔNTF constantly induces CRE. These data suggest that NTF not only shields the tethered agonist to prevent G protein signaling but also confers a conformation that inhibits the interaction with β-arrestins and the consequent endocytosis and sustained signaling from endosomes.
Collapse
Affiliation(s)
- Pedram Azimzadeh
- Division of General and Oncologic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | | | - Katarina T Chang
- Graduate Program in Life Sciences, University of Maryland, Baltimore, Maryland
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Nariman Balenga
- Division of General and Oncologic Surgery, Department of Surgery, University of Maryland School of Medicine, Baltimore, Maryland.,Molecular and Structural Biology Program at University of Maryland Marlene and Stewart Greenebaum Comprehensive Cancer Center, Baltimore, Maryland
| |
Collapse
|
23
|
Diamantopoulou E, Baxendale S, de la Vega de León A, Asad A, Holdsworth CJ, Abbas L, Gillet VJ, Wiggin GR, Whitfield TT. Identification of compounds that rescue otic and myelination defects in the zebrafish adgrg6 ( gpr126) mutant. eLife 2019; 8:44889. [PMID: 31180326 PMCID: PMC6598766 DOI: 10.7554/elife.44889] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 06/08/2019] [Indexed: 12/18/2022] Open
Abstract
Adgrg6 (Gpr126) is an adhesion class G protein-coupled receptor with a conserved role in myelination of the peripheral nervous system. In the zebrafish, mutation of adgrg6 also results in defects in the inner ear: otic tissue fails to down-regulate versican gene expression and morphogenesis is disrupted. We have designed a whole-animal screen that tests for rescue of both up- and down-regulated gene expression in mutant embryos, together with analysis of weak and strong alleles. From a screen of 3120 structurally diverse compounds, we have identified 68 that reduce versican b expression in the adgrg6 mutant ear, 41 of which also restore myelin basic protein gene expression in Schwann cells of mutant embryos. Nineteen compounds unable to rescue a strong adgrg6 allele provide candidates for molecules that may interact directly with the Adgrg6 receptor. Our pipeline provides a powerful approach for identifying compounds that modulate GPCR activity, with potential impact for future drug design.
Collapse
Affiliation(s)
- Elvira Diamantopoulou
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Sarah Baxendale
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | | | - Anzar Asad
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Celia J Holdsworth
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Leila Abbas
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| | - Valerie J Gillet
- Information School, University of Sheffield, Sheffield, United Kingdom
| | | | - Tanya T Whitfield
- Bateson Centre and Department of Biomedical Science, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
24
|
Folts CJ, Giera S, Li T, Piao X. Adhesion G Protein-Coupled Receptors as Drug Targets for Neurological Diseases. Trends Pharmacol Sci 2019; 40:278-293. [PMID: 30871735 DOI: 10.1016/j.tips.2019.02.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 02/03/2019] [Accepted: 02/05/2019] [Indexed: 01/06/2023]
Abstract
The family of adhesion G protein-coupled receptors (aGPCRs) consists of 33 members in humans. Although the majority are orphan receptors with unknown functions, many reports have demonstrated critical functions for some members of this family in organogenesis, neurodevelopment, myelination, angiogenesis, and cancer progression. Importantly, mutations in several aGPCRs have been linked to human diseases. The crystal structure of a shared protein domain, the GPCR Autoproteolysis INducing (GAIN) domain, has enabled the discovery of a common signaling mechanism - a tethered agonist - for this class of receptors. A series of recent reports has shed new light on their biological functions and disease relevance. This review focuses on these recent advances in our understanding of aGPCR biology in the nervous system and the untapped potential of aGPCRs as novel therapeutic targets for neurological disease.
Collapse
Affiliation(s)
- Christopher J Folts
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Current address: Vertex Pharmaceuticals, 50 Northern Avenue, Boston, MA 02210, USA
| | - Stefanie Giera
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Current address: Sanofi S.A., 49 New York Avenue, Framingham, MA 01701, USA
| | - Tao Li
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Xianhua Piao
- Division of Newborn Medicine, Department of Medicine, Children's Hospital and Harvard Medical School, Boston, MA 02115, USA; Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143, USA; Newborn Brain Research Institute, University of California at San Francisco, San Francisco, CA 94158, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
25
|
Hosseini M, Fattahi Z, Abedini SS, Hu H, Ropers H, Kalscheuer VM, Najmabadi H, Kahrizi K. GPR126
: A novel candidate gene implicated in autosomal recessive intellectual disability. Am J Med Genet A 2018; 179:13-19. [DOI: 10.1002/ajmg.a.40531] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 07/29/2018] [Accepted: 08/10/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Masoumeh Hosseini
- Genetics Research CenterUniversity of Social Welfare and Rehabilitation Sciences Tehran Iran
| | - Zohreh Fattahi
- Genetics Research CenterUniversity of Social Welfare and Rehabilitation Sciences Tehran Iran
| | | | - Hao Hu
- Department Human Molecular GeneticsMax‐Planck‐Institute for Molecular Genetics Berlin Germany
| | - Hans‐H. Ropers
- Department Human Molecular GeneticsMax‐Planck‐Institute for Molecular Genetics Berlin Germany
| | - Vera M. Kalscheuer
- Department Human Molecular GeneticsMax‐Planck‐Institute for Molecular Genetics Berlin Germany
| | - Hossein Najmabadi
- Genetics Research CenterUniversity of Social Welfare and Rehabilitation Sciences Tehran Iran
| | - Kimia Kahrizi
- Genetics Research CenterUniversity of Social Welfare and Rehabilitation Sciences Tehran Iran
| |
Collapse
|
26
|
Khan MJ, Pollock N, Jiang H, Castro C, Nazli R, Ahmed J, Basit S, Rajkovic A, Yatsenko AN. X-linked ADGRG2 mutation and obstructive azoospermia in a large Pakistani family. Sci Rep 2018; 8:16280. [PMID: 30389958 PMCID: PMC6214919 DOI: 10.1038/s41598-018-34262-5] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Accepted: 10/10/2018] [Indexed: 02/02/2023] Open
Abstract
We performed whole exome sequencing to identify an unknown genetic cause of azoospermia and male infertility in a large Pakistani family. Three infertile males were subjected to semen analysis, hormone testing, testicular histology, ultrasonography, karyotyping, Y-chromosome microdeletion and CFTR testing. The clinical testing suggested a diagnosis of obstructive azoospermia (OA). To identify the cause, we performed whole exome sequencing (WES) for 2 infertile brothers and 2 fertile family members. For segregation analysis and variant confirmation, we performed Sanger sequencing. WES data analysis of the family revealed segregated variants in 3 candidate genes. We considered novel nonsense variant c.2440C > T(p.Arg814*) in X-linked gene ADGRG2 as biologically most plausible. It is predicted to truncate the protein by 204 amino acids (aa) at a key transmembrane domain. Adgrg2-knockout male mice show sperm loss due to obstructive fluid stasis, while ADGRG2 mutations cause OA in the infertile male patients. Our analysis of testicular histology reveals secondary severe reduction of spermatogenesis, consistent with human and knockout mouse phenotypes. The ADGRG2 nonsense mutation is absent in the largest population databases, ExAC and gnomAD. Analysis of the novel nonsense mutation in extended family members confirmed co-segregation of the mutation with OA in all affected males. The likely pathogenic nature of the mutation is supported by its truncation effect on the transmembrane domain and distinctive ultrasound results. The study demonstrates effectiveness of WES in discovering a genetic cause of azoospermia.
Collapse
Affiliation(s)
- Muhammad Jaseem Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Nijole Pollock
- Department of OBGYN and Reproductive Sciences, Magee-Womens Research Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Huaiyang Jiang
- Department of OBGYN and Reproductive Sciences, Magee-Womens Research Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carlos Castro
- Department of OBGYN and Reproductive Sciences, Magee-Womens Research Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rubina Nazli
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Jawad Ahmed
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Sulman Basit
- Center for Genetics and Inherited Diseases, Taibah University, Almadina Almunawarrah, Medina, Saudi Arabia
| | - Aleksandar Rajkovic
- Department of OBGYN and Reproductive Sciences, Magee-Womens Research Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Pathology, University of California San Francisco (current appointment), San Francisco, CA, USA
| | - Alexander N Yatsenko
- Department of OBGYN and Reproductive Sciences, Magee-Womens Research Institute, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA. .,Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, USA. .,Department of Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
27
|
Abstract
The evolutionarily conserved adhesion G protein-coupled receptors (aGPCRs) play critical roles in biological processes as diverse as brain development, cell polarity and innate immune functions. A defining feature of aGPCRs is the GPCR autoproteolysis inducing (GAIN) domain capable of self-catalytic cleavage, resulting in the generation of an extracellular N-terminal fragment (NTF) and a seven-transmembrane C-terminal fragment (CTF) involved in the cellular adhesion and signaling functions, respectively. Interestingly, two different NTF subtypes have previously been identified, namely an NTF that couples non-covalently with the CTF and a membrane-associated NTF that tethers on cell surface independently. The two NTF subtypes are expected to regulate aGPCR signaling via distinct mechanisms however their molecular characteristics are largely unknown. Herein, the membrane-associated NTF of EMR2/ADGRE2 is investigated and found to be modified by differential N-glycosylation. The membrane association of EMR2-NTF occurs in post-ER compartments and site-specific N-glycosylation in the GAIN domain is involved in modulating its membrane-association ability. Finally, a unique amphipathic α-helix in the GAIN domain is identified as a putative membrane anchor of EMR2-NTF. These results provide novel insights into the complex interaction and activation mechanisms of aGPCRs.
Collapse
|
28
|
Fine-Tuning Limited Proteolysis: A Major Role for Regulated Site-Specific O-Glycosylation. Trends Biochem Sci 2018; 43:269-284. [PMID: 29506880 DOI: 10.1016/j.tibs.2018.02.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 01/26/2018] [Accepted: 02/02/2018] [Indexed: 11/23/2022]
Abstract
Limited proteolytic processing is an essential and ubiquitous post-translational modification (PTM) affecting secreted proteins; failure to regulate the process is often associated with disease. Glycosylation is also a ubiquitous protein PTM and site-specific O-glycosylation in close proximity to sites of proteolysis can regulate and direct the activity of proprotein convertases, a disintegrin and metalloproteinases (ADAMs), and metalloproteinases affecting the activation or inactivation of many classes of proteins, including G-protein-coupled receptors (GPCRs). Here, we summarize the emerging data that suggest O-glycosylation to be a key regulator of limited proteolysis, and highlight the potential for crosstalk between multiple PTMs.
Collapse
|
29
|
Zhang DL, Sun YJ, Ma ML, Wang YJ, Lin H, Li RR, Liang ZL, Gao Y, Yang Z, He DF, Lin A, Mo H, Lu YJ, Li MJ, Kong W, Chung KY, Yi F, Li JY, Qin YY, Li J, Thomsen ARB, Kahsai AW, Chen ZJ, Xu ZG, Liu M, Li D, Yu X, Sun JP. Gq activity- and β-arrestin-1 scaffolding-mediated ADGRG2/CFTR coupling are required for male fertility. eLife 2018; 7:e33432. [PMID: 29393851 PMCID: PMC5839696 DOI: 10.7554/elife.33432] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/30/2018] [Indexed: 12/23/2022] Open
Abstract
Luminal fluid reabsorption plays a fundamental role in male fertility. We demonstrated that the ubiquitous GPCR signaling proteins Gq and β-arrestin-1 are essential for fluid reabsorption because they mediate coupling between an orphan receptor ADGRG2 (GPR64) and the ion channel CFTR. A reduction in protein level or deficiency of ADGRG2, Gq or β-arrestin-1 in a mouse model led to an imbalance in pH homeostasis in the efferent ductules due to decreased constitutive CFTR currents. Efferent ductule dysfunction was rescued by the specific activation of another GPCR, AGTR2. Further mechanistic analysis revealed that β-arrestin-1 acts as a scaffold for ADGRG2/CFTR complex formation in apical membranes, whereas specific residues of ADGRG2 confer coupling specificity for different G protein subtypes, this specificity is critical for male fertility. Therefore, manipulation of the signaling components of the ADGRG2-Gq/β-arrestin-1/CFTR complex by small molecules may be an effective therapeutic strategy for male infertility.
Collapse
Affiliation(s)
- Dao-Lai Zhang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
- School of PharmacyBinzhou Medical UniversityYantaiChina
| | - Yu-Jing Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Ming-Liang Ma
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Yi-jing Wang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Hui Lin
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Rui-Rui Li
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Zong-Lai Liang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Yuan Gao
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Zhao Yang
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Dong-Fang He
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Amy Lin
- Department of BiochemistrySchool of Medicine, Duke UniversityDurhamUnited States
| | - Hui Mo
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Yu-Jing Lu
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Meng-Jing Li
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Wei Kong
- Key Laboratory of Molecular Cardiovascular Science, Department of Physiology and PathophysiologySchool of Basic Medical Sciences, Peking UniversityBeijingChina
| | | | - Fan Yi
- Department of PharmacologyShandong University School of MedicineJinanChina
| | - Jian-Yuan Li
- Key Laboratory of Male Reproductive Health, National Research Institute for Family PlanningNational Health and Family Planning CommissionBeijingChina
| | - Ying-Ying Qin
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsShandong UniversityJinanChina
| | - Jingxin Li
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Alex R B Thomsen
- Department of BiochemistrySchool of Medicine, Duke UniversityDurhamUnited States
| | - Alem W Kahsai
- Department of BiochemistrySchool of Medicine, Duke UniversityDurhamUnited States
| | - Zi-Jiang Chen
- National Research Center for Assisted Reproductive Technology and Reproductive GeneticsShandong UniversityJinanChina
| | - Zhi-Gang Xu
- Shandong Provincial Key Laboratory of Animal Cells and Developmental BiologyShandong University School of Life SciencesJinanChina
| | - Mingyao Liu
- Shanghai Key Laboratory of Regulatory Biology, School of Life SciencesInstitute of Biomedical Sciences, East China Normal UniversityShanghaiChina
- Department of Molecular and Cellular Medicine, Institute of Biosciences and TechnologyTexas A&M University Health Science CenterHoustonUnited States
| | - Dali Li
- Shanghai Key Laboratory of Regulatory Biology, School of Life SciencesInstitute of Biomedical Sciences, East China Normal UniversityShanghaiChina
| | - Xiao Yu
- Department of PhysiologyShandong University School of MedicineJinanChina
| | - Jin-Peng Sun
- Key Laboratory Experimental Teratology of the Ministry of Education, Department of Biochemistry and Molecular BiologyShandong University School of MedicineJinanChina
- Department of BiochemistrySchool of Medicine, Duke UniversityDurhamUnited States
| |
Collapse
|
30
|
Frenster JD, Inocencio JF, Xu Z, Dhaliwal J, Alghamdi A, Zagzag D, Bayin NS, Placantonakis DG. GPR133 Promotes Glioblastoma Growth in Hypoxia. Neurosurgery 2017; 64:177-181. [PMID: 28899043 PMCID: PMC6433437 DOI: 10.1093/neuros/nyx227] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Accepted: 04/04/2017] [Indexed: 02/07/2023] Open
Affiliation(s)
- Joshua D. Frenster
- Department of Neurosurgery, NYU School of Medicine, New York, New York
- Kimmel Center for Stem Cell Biology, NYU School of Medicine, New York, New
York
| | | | - Zhongye Xu
- Department of Neurosurgery, NYU School of Medicine, New York, New York
| | - Joravar Dhaliwal
- Department of Neurosurgery, NYU School of Medicine, New York, New York
| | | | - David Zagzag
- Department of Neurosurgery, NYU School of Medicine, New York, New York
- Department of Pathology, NYU School of Medicine, New York, New York
- Brain Tumor Center, NYU School of Medicine, New York, New York
- Perlmutter Cancer Center, NYU School of Medicine, New York, New York
| | - N. Sumru Bayin
- Developmental Biology Program, Sloan Kettering Institute, NYU School of
Medicine, New York, New York
| | - Dimitris G. Placantonakis
- Department of Neurosurgery, NYU School of Medicine, New York, New York
- Kimmel Center for Stem Cell Biology, NYU School of Medicine, New York, New
York
- Brain Tumor Center, NYU School of Medicine, New York, New York
- Perlmutter Cancer Center, NYU School of Medicine, New York, New York
- Neuroscience Institute, NYU School of Medicine, New York, New York
| |
Collapse
|
31
|
Scholz N, Guan C, Nieberler M, Grotemeyer A, Maiellaro I, Gao S, Beck S, Pawlak M, Sauer M, Asan E, Rothemund S, Winkler J, Prömel S, Nagel G, Langenhan T, Kittel RJ. Mechano-dependent signaling by Latrophilin/CIRL quenches cAMP in proprioceptive neurons. eLife 2017; 6. [PMID: 28784204 PMCID: PMC5548486 DOI: 10.7554/elife.28360] [Citation(s) in RCA: 113] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 06/29/2017] [Indexed: 01/02/2023] Open
Abstract
Adhesion-type G protein-coupled receptors (aGPCRs), a large molecule family with over 30 members in humans, operate in organ development, brain function and govern immunological responses. Correspondingly, this receptor family is linked to a multitude of diverse human diseases. aGPCRs have been suggested to possess mechanosensory properties, though their mechanism of action is fully unknown. Here we show that the Drosophila aGPCR Latrophilin/dCIRL acts in mechanosensory neurons by modulating ionotropic receptor currents, the initiating step of cellular mechanosensation. This process depends on the length of the extended ectodomain and the tethered agonist of the receptor, but not on its autoproteolysis, a characteristic biochemical feature of the aGPCR family. Intracellularly, dCIRL quenches cAMP levels upon mechanical activation thereby specifically increasing the mechanosensitivity of neurons. These results provide direct evidence that the aGPCR dCIRL acts as a molecular sensor and signal transducer that detects and converts mechanical stimuli into a metabotropic response.
Collapse
Affiliation(s)
- Nicole Scholz
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany.,Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Chonglin Guan
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Matthias Nieberler
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Alexander Grotemeyer
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Isabella Maiellaro
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany.,Rudolf Virchow Center, DFG-Research Center for Experimental Biomedicine, University of Würzburg, Würzburg, Germany
| | - Shiqiang Gao
- Department of Biology, Institute for Molecular Plant Physiology and Biophysics, University of Würzburg Biocenter, Würzburg, Germany
| | - Sebastian Beck
- Department of Biology, Institute for Molecular Plant Physiology and Biophysics, University of Würzburg Biocenter, Würzburg, Germany
| | - Matthias Pawlak
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| | - Markus Sauer
- Department of Biotechnology and Biophysics, University of Würzburg Biocenter, Würzburg, Germany
| | - Esther Asan
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg, Germany
| | - Sven Rothemund
- Core Unit Peptide Technologies, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Jana Winkler
- Rudolf Schönheimer Institute of Biochemistry, Division of Molecular Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Simone Prömel
- Rudolf Schönheimer Institute of Biochemistry, Division of Molecular Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Georg Nagel
- Department of Biology, Institute for Molecular Plant Physiology and Biophysics, University of Würzburg Biocenter, Würzburg, Germany
| | - Tobias Langenhan
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany.,Rudolf Schönheimer Institute of Biochemistry, Division of General Biochemistry, Medical Faculty, Leipzig University, Leipzig, Germany
| | - Robert J Kittel
- Department of Neurophysiology, Institute of Physiology, University of Würzburg, Würzburg, Germany
| |
Collapse
|
32
|
Opportunities for therapeutic antibodies directed at G-protein-coupled receptors. Nat Rev Drug Discov 2017; 16:787-810. [PMID: 28706220 DOI: 10.1038/nrd.2017.91] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
G-protein-coupled receptors (GPCRs) are activated by a diverse range of ligands, from large proteins and proteases to small peptides, metabolites, neurotransmitters and ions. They are expressed on all cells in the body and have key roles in physiology and homeostasis. As such, GPCRs are one of the most important target classes for therapeutic drug discovery. The development of drugs targeting GPCRs has therapeutic value across a wide range of diseases, including cancer, immune and inflammatory disorders as well as neurological and metabolic diseases. The progress made by targeting GPCRs with antibody-based therapeutics, as well as technical hurdles to overcome, are presented and discussed in this Review. Antibody therapeutics targeting C-C chemokine receptor type 4 (CCR4), CCR5 and calcitonin gene-related peptide (CGRP) are used as illustrative clinical case studies.
Collapse
|
33
|
Hernández-Vásquez MN, Adame-García SR, Hamoud N, Chidiac R, Reyes-Cruz G, Gratton JP, Côté JF, Vázquez-Prado J. Cell adhesion controlled by adhesion G protein-coupled receptor GPR124/ADGRA2 is mediated by a protein complex comprising intersectins and Elmo-Dock. J Biol Chem 2017; 292:12178-12191. [PMID: 28600358 DOI: 10.1074/jbc.m117.780304] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 05/30/2017] [Indexed: 11/06/2022] Open
Abstract
Developmental angiogenesis and the maintenance of the blood-brain barrier involve endothelial cell adhesion, which is linked to cytoskeletal dynamics. GPR124 (also known as TEM5/ADGRA2) is an adhesion G protein-coupled receptor family member that plays a pivotal role in brain angiogenesis and in ensuring a tight blood-brain barrier. However, the signaling properties of GPR124 remain poorly defined. Here, we show that ectopic expression of GPR124 promotes cell adhesion, additive to extracellular matrix-dependent effect, coupled with filopodia and lamellipodia formation and an enrichment of a pool of the G protein-coupled receptor at actin-rich cellular protrusions containing VASP, a filopodial marker. Accordingly, GPR124-expressing cells also displayed increased activation of both Rac and Cdc42 GTPases. Mechanistically, we uncover novel direct interactions between endogenous GPR124 and the Rho guanine nucleotide exchange factors Elmo/Dock and intersectin (ITSN). Small fragments of either Elmo or ITSN1 that bind GPR124 blocked GPR124-induced cell adhesion. In addition, Gβγ interacts with the C-terminal tail of GPR124 and promotes the formation of a GPR124-Elmo complex. Furthermore, GPR124 also promotes the activation of the Elmo-Dock complex, as measured by Elmo phosphorylation on a conserved C-terminal tyrosine residue. Interestingly, Elmo and ITSN1 also interact with each other independently of their GPR124-recognition regions. Moreover, endogenous phospho-Elmo and ITSN1 co-localize with GPR124 at lamellipodia of adhering endothelial cells, where GPR124 expression contributes to polarity acquisition during wound healing. Collectively, our results indicate that GPR124 promotes cell adhesion via Elmo-Dock and ITSN. This constitutes a previously unrecognized complex formed of atypical and conventional Rho guanine nucleotide exchange factors for Rac and Cdc42 that is putatively involved in GPR124-dependent angiogenic responses.
Collapse
Affiliation(s)
- Magda Nohemí Hernández-Vásquez
- Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City 14740, Mexico
| | - Sendi Rafael Adame-García
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City 14740, Mexico
| | - Noumeira Hamoud
- Institut de Recherches Cliniques de Montréal, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Rony Chidiac
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Guadalupe Reyes-Cruz
- Department of Cell Biology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City 14740, Mexico
| | - Jean Philippe Gratton
- Department of Pharmacology, Faculty of Medicine, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Jean-François Côté
- Institut de Recherches Cliniques de Montréal, Université de Montréal, Montreal, Quebec H3T 1J4, Canada
| | - José Vázquez-Prado
- Department of Pharmacology, Center for Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV-IPN), Mexico City 14740, Mexico.
| |
Collapse
|
34
|
I KY, Huang YS, Hu CH, Tseng WY, Cheng CH, Stacey M, Gordon S, Chang GW, Lin HH. Activation of Adhesion GPCR EMR2/ADGRE2 Induces Macrophage Differentiation and Inflammatory Responses via Gα 16/Akt/MAPK/NF-κB Signaling Pathways. Front Immunol 2017; 8:373. [PMID: 28421075 PMCID: PMC5376562 DOI: 10.3389/fimmu.2017.00373] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Accepted: 03/15/2017] [Indexed: 11/13/2022] Open
Abstract
EMR2/ADGRE2 is a human myeloid-restricted adhesion G protein-coupled receptor critically implicated in vibratory urticaria, a rare type of allergy caused by vibration-induced mast cell activation. In addition, EMR2 is also highly expressed by monocyte/macrophages and has been linked to neutrophil migration and activation. Despite these findings, little is known of EMR2-mediated signaling and its role in myeloid biology. In this report, we show that activation of EMR2 via a receptor-specific monoclonal antibody promotes the differentiation of human THP-1 monocytic cell line and induces the expression of pro-inflammatory mediators, including IL-8, TNF-α, and MMP-9. Using specific signaling inhibitors and siRNA knockdowns, biochemical and functional analyses reveal that the EMR2-mediated signaling is initiated by Gα16, followed by the subsequent activation of Akt, extracellular signal-regulated kinase, c-Jun N-terminal kinase, and nuclear factor kappa-light-chain-enhancer of activated B cells. Our results demonstrate a functional role for EMR2 in the differentiation and inflammatory activation of human monocytic cells and provide potential targets for myeloid cell-mediated inflammatory disorders.
Collapse
Affiliation(s)
- Kuan-Yu I
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Shu Huang
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ching-Hsun Hu
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Yi Tseng
- Division of Rheumatology, Allergy and Immunology, Chang Gung Memorial Hospital-Keelung, Keelung, Taiwan
| | - Chia-Hsin Cheng
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Martin Stacey
- Faculty of Biological Sciences, School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Siamon Gordon
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Sir William Dunn School of Pathology, University of Oxford, Oxford, UK
| | - Gin-Wen Chang
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hsi-Hsien Lin
- Department of Microbiology and Immunology, College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan.,Chang Gung Immunology Consortium, Chang Gung Memorial Hospital, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
35
|
Goth CK, Tuhkanen HE, Khan H, Lackman JJ, Wang S, Narimatsu Y, Hansen LH, Overall CM, Clausen H, Schjoldager KT, Petäjä-Repo UE. Site-specific O-Glycosylation by Polypeptide N-Acetylgalactosaminyltransferase 2 (GalNAc-transferase T2) Co-regulates β 1-Adrenergic Receptor N-terminal Cleavage. J Biol Chem 2017; 292:4714-4726. [PMID: 28167537 PMCID: PMC5377785 DOI: 10.1074/jbc.m116.730614] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 01/29/2017] [Indexed: 01/08/2023] Open
Abstract
The β1-adrenergic receptor (β1AR) is a G protein-coupled receptor (GPCR) and the predominant adrenergic receptor subtype in the heart, where it mediates cardiac contractility and the force of contraction. Although it is the most important target for β-adrenergic antagonists, such as β-blockers, relatively little is yet known about its regulation. We have shown previously that β1AR undergoes constitutive and regulated N-terminal cleavage participating in receptor down-regulation and, moreover, that the receptor is modified by O-glycosylation. Here we demonstrate that the polypeptide GalNAc-transferase 2 (GalNAc-T2) specifically O-glycosylates β1AR at five residues in the extracellular N terminus, including the Ser-49 residue at the location of the common S49G single-nucleotide polymorphism. Using in vitro O-glycosylation and proteolytic cleavage assays, a cell line deficient in O-glycosylation, GalNAc-T-edited cell line model systems, and a GalNAc-T2 knock-out rat model, we show that GalNAc-T2 co-regulates the metalloproteinase-mediated limited proteolysis of β1AR. Furthermore, we demonstrate that impaired O-glycosylation and enhanced proteolysis lead to attenuated receptor signaling, because the maximal response elicited by the βAR agonist isoproterenol and its potency in a cAMP accumulation assay were decreased in HEK293 cells lacking GalNAc-T2. Our findings reveal, for the first time, a GPCR as a target for co-regulatory functions of site-specific O-glycosylation mediated by a unique GalNAc-T isoform. The results provide a new level of β1AR regulation that may open up possibilities for new therapeutic strategies for cardiovascular diseases.
Collapse
Affiliation(s)
- Christoffer K Goth
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Hanna E Tuhkanen
- the Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland
| | - Hamayun Khan
- the Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland
| | - Jarkko J Lackman
- the Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland
| | - Shengjun Wang
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Yoshiki Narimatsu
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Lasse H Hansen
- the Department of Clinical Biochemistry, Rigshospitalet, Copenhagen University Hospital, DK-2100 Copenhagen Ø, Denmark and
| | - Christopher M Overall
- the Centre for Blood Research, Department of Oral Biological and Medical Sciences, and Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| | - Henrik Clausen
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark
| | - Katrine T Schjoldager
- From the Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Blegdamsvej 3, DK-2200 Copenhagen N, Denmark,
| | - Ulla E Petäjä-Repo
- the Medical Research Center Oulu, Research Unit of Biomedicine, University of Oulu, P.O. Box 5000, FI-90014 Oulu, Finland,
| |
Collapse
|
36
|
|
37
|
Chiang NY, Peng YM, Juang HH, Chen TC, Pan HL, Chang GW, Lin HH. GPR56/ADGRG1 Activation Promotes Melanoma Cell Migration via NTF Dissociation and CTF-Mediated Gα 12/13/RhoA Signaling. J Invest Dermatol 2016; 137:727-736. [PMID: 27818281 DOI: 10.1016/j.jid.2016.10.031] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2016] [Revised: 10/06/2016] [Accepted: 10/17/2016] [Indexed: 02/07/2023]
Abstract
GPR56/ADGRG1 is a versatile adhesion G protein-coupled receptor with diverse biological functions. GPR56 expression is variably detected in human melanoma cell lines and correlates inversely with the metastatic potential of melanoma lesions. GPR56 associates with the tetraspanins CD9 and CD81 on the melanoma cell surface. GPR56 activation by immobilized CG4 monoclonal antibody facilitates N-terminal fragment dissociation in a CD9/CD81-dependent manner specifically inducing IL-6 production, which promotes cell migration and invasion. Interestingly, expression of GPR56-C-terminal fragment alone recapitulates the antibody-induced receptor function, implicating a major role for the C-terminal fragment in GPR56 activation and signaling. Analysis of site-directed mutant receptors attests the importance of the conserved N-terminal residues of the C-terminal fragment for its self-activation. Finally, we show that the GPR56-induced signaling in melanoma cells is mediated by the Gα12/13/RhoA pathway. In summary, the expression and activation of GPR56 may modulate melanoma progression in part by inducing IL-6 production after N-terminal fragment dissociation and C-terminal fragment self-activation.
Collapse
Affiliation(s)
- Nien-Yi Chiang
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Yen-Ming Peng
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Horng-Heng Juang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan; Department of Anatomy, College of Medicine, Chang Gung University, Taoyuan, Taiwan; Department of Urology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Tse-Ching Chen
- Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan
| | - Hsiao-Lin Pan
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
| | - Gin-Wen Chang
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan
| | - Hsi-Hsien Lin
- Department of Microbiology and Immunology, Chang Gung University, Taoyuan, Taiwan; Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan; Department of Anatomic Pathology, Chang Gung Memorial Hospital-Linkou, Taoyuan, Taiwan; Chang Gung Immunology Consortium, Chang Gung Memorial Hospital and Chang Gung University, Taoyuan, Taiwan.
| |
Collapse
|
38
|
Bayin NS, Frenster JD, Kane JR, Rubenstein J, Modrek AS, Baitalmal R, Dolgalev I, Rudzenski K, Scarabottolo L, Crespi D, Redaelli L, Snuderl M, Golfinos JG, Doyle W, Pacione D, Parker EC, Chi AS, Heguy A, MacNeil DJ, Shohdy N, Zagzag D, Placantonakis DG. GPR133 (ADGRD1), an adhesion G-protein-coupled receptor, is necessary for glioblastoma growth. Oncogenesis 2016; 5:e263. [PMID: 27775701 PMCID: PMC5117849 DOI: 10.1038/oncsis.2016.63] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 08/09/2016] [Accepted: 08/24/2016] [Indexed: 02/06/2023] Open
Abstract
Glioblastoma (GBM) is a deadly primary brain malignancy with extensive intratumoral hypoxia. Hypoxic regions of GBM contain stem-like cells and are associated with tumor growth and angiogenesis. The molecular mechanisms that regulate tumor growth in hypoxic conditions are incompletely understood. Here, we use primary human tumor biospecimens and cultures to identify GPR133 (ADGRD1), an orphan member of the adhesion family of G-protein-coupled receptors, as a critical regulator of the response to hypoxia and tumor growth in GBM. GPR133 is selectively expressed in CD133+ GBM stem cells (GSCs) and within the hypoxic areas of PPN in human biospecimens. GPR133 mRNA is transcriptionally upregulated by hypoxia in hypoxia-inducible factor 1α (Hif1α)-dependent manner. Genetic inhibition of GPR133 with short hairpin RNA reduces the prevalence of CD133+ GSCs, tumor cell proliferation and tumorsphere formation in vitro. Forskolin rescues the GPR133 knockdown phenotype, suggesting that GPR133 signaling is mediated by cAMP. Implantation of GBM cells with short hairpin RNA-mediated knockdown of GPR133 in the mouse brain markedly reduces tumor xenograft formation and increases host survival. Analysis of the TCGA data shows that GPR133 expression levels are inversely correlated with patient survival. These findings indicate that GPR133 is an important mediator of the hypoxic response in GBM and has significant protumorigenic functions. We propose that GPR133 represents a novel molecular target in GBM and possibly other malignancies where hypoxia is fundamental to pathogenesis.
Collapse
Affiliation(s)
- N S Bayin
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
- Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY, USA
| | - J D Frenster
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
- Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY, USA
| | - J R Kane
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - J Rubenstein
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - A S Modrek
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - R Baitalmal
- Department of Pathology, New York University School of Medicine, New York, NY, USA
| | - I Dolgalev
- Genome Technology Center, New York University School of Medicine, New York, NY, USA
| | - K Rudzenski
- Office for Therapeutic Alliances, New York University School of Medicine, New York, NY, USA
| | | | | | | | - M Snuderl
- Department of Pathology, New York University School of Medicine, New York, NY, USA
- Brain Tumor Center, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - J G Golfinos
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
- Brain Tumor Center, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - W Doyle
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - D Pacione
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - E C Parker
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
| | - A S Chi
- Brain Tumor Center, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
- Department of Neurology, New York University School of Medicine, New York, NY, USA
| | - A Heguy
- Genome Technology Center, New York University School of Medicine, New York, NY, USA
| | - D J MacNeil
- Office for Therapeutic Alliances, New York University School of Medicine, New York, NY, USA
| | - N Shohdy
- Office for Therapeutic Alliances, New York University School of Medicine, New York, NY, USA
| | - D Zagzag
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
- Department of Pathology, New York University School of Medicine, New York, NY, USA
- Brain Tumor Center, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - D G Placantonakis
- Department of Neurosurgery, New York University School of Medicine, New York, NY, USA
- Kimmel Center for Stem Cell Biology, New York University School of Medicine, New York, NY, USA
- Brain Tumor Center, New York University School of Medicine, New York, NY, USA
- Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| |
Collapse
|
39
|
Patat O, Pagin A, Siegfried A, Mitchell V, Chassaing N, Faguer S, Monteil L, Gaston V, Bujan L, Courtade-Saïdi M, Marcelli F, Lalau G, Rigot JM, Mieusset R, Bieth E. Truncating Mutations in the Adhesion G Protein-Coupled Receptor G2 Gene ADGRG2 Cause an X-Linked Congenital Bilateral Absence of Vas Deferens. Am J Hum Genet 2016; 99:437-42. [PMID: 27476656 DOI: 10.1016/j.ajhg.2016.06.012] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/10/2016] [Indexed: 12/26/2022] Open
Abstract
In 80% of infertile men with obstructive azoospermia caused by a congenital bilateral absence of the vas deferens (CBAVD), mutations are identified in the cystic fibrosis transmembrane conductance regulator gene (CFTR). For the remaining 20%, the origin of the CBAVD is unknown. A large cohort of azoospermic men with CBAVD was retrospectively reassessed with more stringent selection criteria based on consistent clinical data, complete description of semen and reproductive excurrent ducts, extensive CFTR testing, and kidney ultrasound examination. To maximize the phenotypic prioritization, men with CBAVD and with unilateral renal agenesis were considered ineligible for the present study. We performed whole-exome sequencing on 12 CFTR-negative men with CBAVD and targeted sequencing on 14 additional individuals. We identified three protein-truncating hemizygous mutations, c.1545dupT (p.Glu516Ter), c.2845delT (p.Cys949AlafsTer81), and c.2002_2006delinsAGA (p.Leu668ArgfsTer21), in ADGRG2, encoding the epididymal- and efferent-ducts-specific adhesion G protein-coupled receptor G2, in four subjects, including two related individuals with X-linked transmission of their infertility. Previous studies have demonstrated that Adgrg2-knockout male mice develop obstructive infertility. Our study confirms the crucial role of ADGRG2 in human male fertility and brings new insight into congenital obstructive azoospermia pathogenesis. In men with CBAVD who are CFTR-negative, ADGRG2 testing could allow for appropriate genetic counseling with regard to the X-linked transmission of the molecular defect.
Collapse
|
40
|
Chang GW, Hsiao CC, Peng YM, Vieira Braga F, Kragten N, Remmerswaal E, van de Garde M, Straussberg R, König G, Kostenis E, Knäuper V, Meyaard L, van Lier R, van Gisbergen K, Lin HH, Hamann J. The Adhesion G Protein-Coupled Receptor GPR56/ADGRG1 Is an Inhibitory Receptor on Human NK Cells. Cell Rep 2016; 15:1757-70. [DOI: 10.1016/j.celrep.2016.04.053] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 01/22/2016] [Accepted: 04/13/2016] [Indexed: 10/21/2022] Open
|
41
|
Buckley SJ, Fitzgibbon QP, Smith GG, Ventura T. In silico prediction of the G-protein coupled receptors expressed during the metamorphic molt of Sagmariasus verreauxi (Crustacea: Decapoda) by mining transcriptomic data: RNA-seq to repertoire. Gen Comp Endocrinol 2016; 228:111-127. [PMID: 26850661 DOI: 10.1016/j.ygcen.2016.02.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 01/29/2016] [Accepted: 02/01/2016] [Indexed: 10/22/2022]
Abstract
Against a backdrop of food insecurity, the farming of decapod crustaceans is a rapidly expanding and globally significant source of food protein. Sagmariasus verreauxi spiny lobster, the subject of this study, are decapods of underdeveloped aquaculture potential. Crustacean neuropeptide G-protein coupled receptors (GPCRs) mediate endocrine pathways that are integral to animal fecundity, growth and survival. The potential use of novel biotechnologies to enhance GPCR-mediated physiology may assist in improving the health and productivity of farmed decapod populations. This study catalogues the GPCRs expressed in the early developmental stages, as well as adult tissues, with a view to illuminating key neuropeptide receptors. De novo assembled contiguous sequences generated from transcriptomic reads of metamorphic and post metamorphic S. verreauxi were filtered for seven transmembrane domains, and used as a reference for iterative re-mapping. Subsequent putative GPCR open reading frames (ORFs) were BLAST annotated, categorised, and compared to published orthologues based on phylogenetic analysis. A total of 85 GPCRs were digitally predicted, that represented each of the four arthropod subfamilies. They generally displayed low-level and non-differential metamorphic expression with few exceptions that we examined using RT-PCR and qPCR. Two putative CHH-like neuropeptide receptors were annotated. Three dimensional structural modelling suggests that these receptors exhibit a conserved extracellular ligand binding pocket, providing support to the notion that these receptors co-evolved with their ligands across Decapoda. This perhaps narrows the search for means to increase productivity of farmed decapod populations.
Collapse
Affiliation(s)
- Sean J Buckley
- GeneCology Research Centre, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, 4 Locked Bag, Maroochydore, Queensland 4558, Australia
| | - Quinn P Fitzgibbon
- Fisheries and Aquaculture, Institute for Marine and Antarctic Studies, University of Tasmania, Hobart, Tasmania, Australia
| | - Gregory G Smith
- Fisheries and Aquaculture, Institute for Marine and Antarctic Studies, University of Tasmania, Hobart, Tasmania, Australia
| | - Tomer Ventura
- GeneCology Research Centre, Faculty of Science, Health, Education and Engineering, University of the Sunshine Coast, 4 Locked Bag, Maroochydore, Queensland 4558, Australia.
| |
Collapse
|
42
|
Abstract
Development of the aGPCR scientific field based on PubMed-listed research articles and selected key findings Since the discovery of adhesion G-protein-coupled receptors (aGPCRs) 20 years ago, reverse genetics approaches have dominated the elucidation of their function and work mechanisms. Seminal findings in this field comprise the description of aGPCRs as seven-transmembrane (7TM) molecules with an extended extracellular region, the identification of matricellular ligands that bind to distinct protein folds at the N-terminus, the clarification of an autoproteolytic cleavage event at a juxtamembranous GPCR proteolysis site (GPS), the elucidation of the crystal structure of the GPCR autoproteolysis-inducing (GAIN) domain that embeds the GPS and connects the receptor fragments, the demonstration that a short N-terminal sequence of the seven-transmembrane (7TM) region can serve as a tethered agonist, and, recently, the notification that aGPCRs can serve as mechanosensors. We here discuss how these discoveries have moved forward aGPCR research and, finally, linked the field to the GPCR field. We argue that crucial questions remain to be addressed before we can fully appreciate the biological nature of these fascinating receptors.
Collapse
Affiliation(s)
- Jörg Hamann
- Department of Experimental Immunology, K0-144, Academic Medical Center, University of Amsterdam, Meibergdreef 9, 1105AZ, Amsterdam, The Netherlands.
| | - Alexander G Petrenko
- Laboratory of Receptor Cell Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, ul. Miklukho-Maklaya 16/10, 117997, Moscow, Russia.
| |
Collapse
|
43
|
Abstract
Proteolytic processing events in adhesion GPCRs. aGPCRs can undergo multiple autoproteolytic (red asterisks) and proteolytic processing events by exogenous proteases (yellow asterisks) that may be involved in signaling events of the receptors. Proteolytic processing is an unusual property of adhesion family G protein-coupled receptors (aGPCRs) that was observed upon their cloning and biochemical characterization.Ever since, much effort has been dedicated to delineate the mechanisms and requirements for cleavage events in the control of aGPCR function. Most notably, all aGPCRs possess a juxtamembrane protein fold, the GPCR autoproteolysis-inducing (GAIN) domain, which operates as an autoprotease for many aGPCR homologs investigated thus far. Analysis of its autoproteolytic reaction, the consequences for receptor fate and function, and the allocation of physiological effects to this peculiar feature of aGPCRs has occupied the experimental agenda of the aGPCR field and shaped our current understanding of the signaling properties and cell biological effects of aGPCRs. Interestingly, individual aGPCRs may undergo additional proteolytic steps, one of them resulting in shedding of the entire ectodomain that is secreted and can function independently. Here, we summarize the current state of knowledge on GAIN domain-mediated and GAIN domain-independent aGPCR cleavage events and their significance for the pharmacological and cellular actions of aGPCRs. Further, we compare and contrast the proteolytic profile of aGPCRs with known signaling routes that are governed through proteolysis of surface molecules such as the Notch and ephrin pathways.
Collapse
|
44
|
Abstract
Adhesion GPCRs harbor a tethered agonist sequence (reproduced from [24]) As the past years have seen a magnificent increase in knowledge on adhesion GPCR (aGPCR) signal transduction, the time had come to fill the gap on how these receptors can be activated. Based on experimental observations that deletion of the ectodomain can induce signaling, the idea arose that aGPCRs, just like other atypical GPCRs, may harbor a tethered agonist sequence. In this chapter, we describe the recent findings and characteristics of this agonist, called the Stachel sequence, and discuss potential mechanisms that cause liberation of this encrypted sequence. Further, we provide perspectives for application of Stachel-derived synthetic peptides in future studies of aGPCR function.
Collapse
Affiliation(s)
- Ines Liebscher
- Medical Faculty, Institute of Biochemistry, University Leipzig, Johannisallee 30, Leipzig, 04103, Germany.
| | - Torsten Schöneberg
- Medical Faculty, Institute of Biochemistry, University Leipzig, Johannisallee 30, Leipzig, 04103, Germany
| |
Collapse
|
45
|
Abstract
G protein-coupled receptors (GPCRs) remain a major domain of pharmaceutical discovery. The identification of GPCR lead compounds and their optimization are now structure-based, thanks to advances in X-ray crystallography, molecular modeling, protein engineering and biophysical techniques. In silico screening provides useful hit molecules. New pharmacological approaches to tuning the pleotropic action of GPCRs include: allosteric modulators, biased ligands, GPCR heterodimer-targeted compounds, manipulation of polypharmacology, receptor antibodies and tailoring of drug molecules to fit GPCR pharmacogenomics. Measurements of kinetics and drug efficacy are factors influencing clinical success. With the exception of inhibitors of GPCR kinases, targeting of intracellular GPCR signaling or receptor cycling for therapeutic purposes remains a futuristic concept. New assay approaches are more efficient and multidimensional: cell-based, label-free, fluorescence-based assays, and biosensors. Tailoring GPCR drugs to a patient's genetic background is now being considered. Chemoinformatic tools can predict ADME-tox properties. New imaging technology visualizes drug action in vivo. Thus, there is reason to be optimistic that new technology for GPCR ligand discovery will help reverse the current narrowing of the pharmaceutical pipeline.
Collapse
Affiliation(s)
- Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bldg. 8A, Rm. B1A-19, Bethesda, Maryland 20892, USA.
| |
Collapse
|
46
|
Bohn LM, Lohse MJ, Nitabach MN, Taghert PH, Smit MJ. Exploring the Biology of G Protein-Coupled Receptors from In Vitro to In Vivo. Mol Pharmacol 2015; 88:534-5. [PMID: 26162863 DOI: 10.1124/mol.115.100750] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 07/10/2015] [Indexed: 12/29/2022] Open
Abstract
In August 2014, an international group of researchers gathered for 5 days at the Lorentz Center in Leiden, The Netherlands, to explore the technical and conceptual issues associated with the analysis of G protein-coupled receptor functions utilizing information from crystal structure models to the use of model organisms. This collection of review articles evolved from the 5-day meeting, with brief presentations and structured discussion periods that were designed to identify key questions remaining in understanding G protein-coupled receptor function and to propose novel strategies by integrating scientific disciplines to guide future research.
Collapse
Affiliation(s)
- Laura M Bohn
- The Scripps Research Institute, Jupiter, Florida (L.M.B.), University of Würzburg, Würzburg, Germany (M.J.L.); Yale University, New Haven, Connecticut (M.N.N.); Washington University Medical School, St. Louis, Missouri (P.H.T.); and VU University Amsterdam, Amsterdam, The Netherlands (M.J.S.)
| | - Martin J Lohse
- The Scripps Research Institute, Jupiter, Florida (L.M.B.), University of Würzburg, Würzburg, Germany (M.J.L.); Yale University, New Haven, Connecticut (M.N.N.); Washington University Medical School, St. Louis, Missouri (P.H.T.); and VU University Amsterdam, Amsterdam, The Netherlands (M.J.S.)
| | - Michael N Nitabach
- The Scripps Research Institute, Jupiter, Florida (L.M.B.), University of Würzburg, Würzburg, Germany (M.J.L.); Yale University, New Haven, Connecticut (M.N.N.); Washington University Medical School, St. Louis, Missouri (P.H.T.); and VU University Amsterdam, Amsterdam, The Netherlands (M.J.S.)
| | - Paul H Taghert
- The Scripps Research Institute, Jupiter, Florida (L.M.B.), University of Würzburg, Würzburg, Germany (M.J.L.); Yale University, New Haven, Connecticut (M.N.N.); Washington University Medical School, St. Louis, Missouri (P.H.T.); and VU University Amsterdam, Amsterdam, The Netherlands (M.J.S.)
| | - Martine J Smit
- The Scripps Research Institute, Jupiter, Florida (L.M.B.), University of Würzburg, Würzburg, Germany (M.J.L.); Yale University, New Haven, Connecticut (M.N.N.); Washington University Medical School, St. Louis, Missouri (P.H.T.); and VU University Amsterdam, Amsterdam, The Netherlands (M.J.S.)
| |
Collapse
|
47
|
Langenhan T, Barr MM, Bruchas MR, Ewer J, Griffith LC, Maiellaro I, Taghert PH, White BH, Monk KR. Model Organisms in G Protein-Coupled Receptor Research. Mol Pharmacol 2015; 88:596-603. [PMID: 25979002 PMCID: PMC4551050 DOI: 10.1124/mol.115.098764] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2015] [Accepted: 05/14/2015] [Indexed: 12/19/2022] Open
Abstract
The study of G protein-coupled receptors (GPCRs) has benefited greatly from experimental approaches that interrogate their functions in controlled, artificial environments. Working in vitro, GPCR receptorologists discovered the basic biologic mechanisms by which GPCRs operate, including their eponymous capacity to couple to G proteins; their molecular makeup, including the famed serpentine transmembrane unit; and ultimately, their three-dimensional structure. Although the insights gained from working outside the native environments of GPCRs have allowed for the collection of low-noise data, such approaches cannot directly address a receptor's native (in vivo) functions. An in vivo approach can complement the rigor of in vitro approaches: as studied in model organisms, it imposes physiologic constraints on receptor action and thus allows investigators to deduce the most salient features of receptor function. Here, we briefly discuss specific examples in which model organisms have successfully contributed to the elucidation of signals controlled through GPCRs and other surface receptor systems. We list recent examples that have served either in the initial discovery of GPCR signaling concepts or in their fuller definition. Furthermore, we selectively highlight experimental advantages, shortcomings, and tools of each model organism.
Collapse
Affiliation(s)
- Tobias Langenhan
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Maureen M Barr
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Michael R Bruchas
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - John Ewer
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Leslie C Griffith
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Isabella Maiellaro
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Paul H Taghert
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Benjamin H White
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| | - Kelly R Monk
- Institute of Physiology, Department of Neurophysiology (T.L.), and Institute of Pharmacology and Toxicology, Rudolf Virchow Center (I.M.), University of Würzburg, Germany, Würzburg, Germany; Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, New Jersey (M.M.B.); Division of Basic Research, Department of Anesthesiology, Washington University Pain Center (M.R.B.), Division of Biological and Biomedical Sciences, Department of Anatomy and Neurobiology (M.R.B., P.H.T.), and Department of Developmental Biology, Hope Center for Neurologic Disorders, (K.R.M.), Washington University School of Medicine, St. Louis, Missouri; Centro Interdisciplinario de Neurociencia, Universidad de Valparaiso, Valparaiso, Chile (J.E.); National Center of Behavioral Genomics, Volen Center for Complex Systems, and Department of Biology, Brandeis University, Waltham, Massachusetts (L.C.G.); and Laboratory of Molecular Biology, National Institutes of Health National Institute of Mental Health, Bethesda, Maryland (B.H.W.)
| |
Collapse
|
48
|
van Unen J, Woolard J, Rinken A, Hoffmann C, Hill SJ, Goedhart J, Bruchas MR, Bouvier M, Adjobo-Hermans MJW. A Perspective on Studying G-Protein-Coupled Receptor Signaling with Resonance Energy Transfer Biosensors in Living Organisms. Mol Pharmacol 2015; 88:589-95. [PMID: 25972446 PMCID: PMC4551049 DOI: 10.1124/mol.115.098897] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 05/13/2015] [Indexed: 01/09/2023] Open
Abstract
The last frontier for a complete understanding of G-protein-coupled receptor (GPCR) biology is to be able to assess GPCR activity, interactions, and signaling in vivo, in real time within biologically intact systems. This includes the ability to detect GPCR activity, trafficking, dimerization, protein-protein interactions, second messenger production, and downstream signaling events with high spatial resolution and fast kinetic readouts. Resonance energy transfer (RET)-based biosensors allow for all of these possibilities in vitro and in cell-based assays, but moving RET into intact animals has proven difficult. Here, we provide perspectives on the optimization of biosensor design, of signal detection in living organisms, and the multidisciplinary development of in vitro and cell-based assays that more appropriately reflect the physiologic situation. In short, further development of RET-based probes, optical microscopy techniques, and mouse genome editing hold great potential over the next decade to bring real-time in vivo GPCR imaging to the forefront of pharmacology.
Collapse
Affiliation(s)
- Jakobus van Unen
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
| | - Jeanette Woolard
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
| | - Ago Rinken
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
| | - Carsten Hoffmann
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
| | - Stephen J Hill
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
| | - Joachim Goedhart
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
| | - Michael R Bruchas
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
| | - Michel Bouvier
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
| | - Merel J W Adjobo-Hermans
- Department of Biochemistry, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands (M.J.W.A.-H.); Department of Biochemistry, Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec, Canada (M.B.); Department of Anesthesiology, Washington University, St. Louis, Missouri (M.R.B.); Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Amsterdam, The Netherlands (J.U., J.G.); Cell Signalling Research Group, School of Biomedical Sciences, Medical School, Queen's Medical Centre, University of Nottingham, Nottingham, United Kingdom (J.W., S.J.H.); Bio-Imaging-Center/Rudolf-Virchow-Zentrum and Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany (C.H.); and Institute of Chemistry, University of Tartu, Tartu, Estonia (A.R.)
| |
Collapse
|