1
|
Lindahl TL, Kumar AP, Hallström T, Al-Hashimi A, du Rietz A, Arlaman E, Uvdal K, Macwan AS. Dabigatran Attenuates the Binding of Thrombin to Platelets-A Novel Mechanism of Action. Thromb Haemost 2024. [PMID: 39586831 DOI: 10.1055/a-2483-0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
BACKGROUND Thrombin is a multifunctional regulatory enzyme of the haemostasis and has both pro- and anticoagulant roles. It has, therefore, been a main target for drug discovery over many decades. Thrombin is a serine protease and possesses two positively charged regions called exosites, through which it is known to bind to many substrates. Dabigatran is a thrombin inhibitor and is widely used as an oral anticoagulant for the antithrombotic treatment of atrial fibrillation and venous thromboembolism. The mechanism by which dabigatran inhibits thrombin is the blockage of the active site, however, its effect on thrombin binding to its substrates has not been studied thoroughly and is thus poorly understood. MATERIAL AND METHODS The effect of dabigatran on thrombin binding to platelets was evaluated by flow cytometry using fluorescently labelled thrombin and washed platelets. Further, to confirm the results we utilized modern techniques for biomolecular binding studies, microscale thermophoresis (MST) and surface plasmon resonance (SPR), which validated the results. RESULTS Dabigatran inhibited thrombin binding to platelets as analysed by flow cytometry. The inhibition was dose dependent with IC50 of 118 nM which was slightly lower than for inhibition of platelet activation and is close to the clinically relevant plasma concentration of dabigatran. MST and SPR also confirmed inhibitory effect of dabigatran on thrombin binding to platelets. CONCLUSION Apart from blocking the active site, dabigatran also inhibits thrombin binding to platelets. Since thrombin has numerous functions beyond the cardiovascular system, this finding may have important implications.
Collapse
Affiliation(s)
- Tomas L Lindahl
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Aishwarya Prasanna Kumar
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | | | - Ahmed Al-Hashimi
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Anna du Rietz
- Division of Molecular Surface Physics and Nanoscience, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Elena Arlaman
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Kajsa Uvdal
- Division of Molecular Surface Physics and Nanoscience, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Ankit S Macwan
- Division of Clinical Chemistry and Pharmacology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
2
|
Anthofer L, Gmach P, Uretmen Kagiali ZC, Kleinau G, Rotter J, Opitz R, Scheerer P, Beck-Sickinger AG, Wolf P, Biebermann H, Bechmann I, Kühnen P, Krude H, Paisdzior S. Melanocortin-4 Receptor PLC Activation Is Modulated by an Interaction with the Monocarboxylate Transporter 8. Int J Mol Sci 2024; 25:7565. [PMID: 39062808 PMCID: PMC11277258 DOI: 10.3390/ijms25147565] [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: 06/14/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
The melanocortin-4 receptor (MC4R) is a key player in the hypothalamic leptin-melanocortin pathway that regulates satiety and hunger. MC4R belongs to the G protein-coupled receptors (GPCRs), which are known to form heterodimers with other membrane proteins, potentially modulating receptor function or characteristics. Like MC4R, thyroid hormones (TH) are also essential for energy homeostasis control. TH transport across membranes is facilitated by the monocarboxylate transporter 8 (MCT8), which is also known to form heterodimers with GPCRs. Based on the finding in single-cell RNA-sequencing data that both proteins are simultaneously expressed in hypothalamic neurons, we investigated a putative interplay between MC4R and MCT8. We developed a novel staining protocol utilizing a fluorophore-labeled MC4R ligand and demonstrated a co-localization of MC4R and MCT8 in human brain tissue. Using in vitro assays such as BRET, IP1, and cAMP determination, we found that MCT8 modulates MC4R-mediated phospholipase C activation but not cAMP formation via a direct interaction, an effect that does not require a functional MCT8 as it was not altered by a specific MCT8 inhibitor. This suggests an extended functional spectrum of MCT8 as a GPCR signaling modulator and argues for the investigation of further GPCR-protein interactions with hitherto underrepresented physiological functions.
Collapse
Affiliation(s)
- Larissa Anthofer
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
- Institute of Anatomy, Leipzig University, D-04103 Leipzig, Germany
| | - Philipp Gmach
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Zeynep Cansu Uretmen Kagiali
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Gunnar Kleinau
- Group Structural Biology of Cellular Signaling, Institute of Medical Physics and Biophysics, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Jonas Rotter
- Institute of Anatomy, Leipzig University, D-04103 Leipzig, Germany
| | - Robert Opitz
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Patrick Scheerer
- Group Structural Biology of Cellular Signaling, Institute of Medical Physics and Biophysics, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | | | - Philipp Wolf
- Faculty of Life Sciences, Institute of Biochemistry, Leipzig University, D-04103 Leipzig, Germany
| | - Heike Biebermann
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Ingo Bechmann
- Institute of Anatomy, Leipzig University, D-04103 Leipzig, Germany
| | - Peter Kühnen
- Department for Pediatric Endocrinology and Diabetology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Heiko Krude
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| | - Sarah Paisdzior
- Institute of Experimental Pediatric Endocrinology, Charité—Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, D-10117 Berlin, Germany
| |
Collapse
|
3
|
Kuo CY, Tay RJ, Lin HC, Juan SC, Vidal-Diez de Ulzurrun G, Chang YC, Hoki J, Schroeder FC, Hsueh YP. The nematode-trapping fungus Arthrobotrys oligospora detects prey pheromones via G protein-coupled receptors. Nat Microbiol 2024; 9:1738-1751. [PMID: 38649409 PMCID: PMC11724650 DOI: 10.1038/s41564-024-01679-w] [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: 08/11/2023] [Accepted: 03/20/2024] [Indexed: 04/25/2024]
Abstract
The ability to sense prey-derived cues is essential for predatory lifestyles. Under low-nutrient conditions, Arthrobotrys oligospora and other nematode-trapping fungi develop dedicated structures for nematode capture when exposed to nematode-derived cues, including a conserved family of pheromones, the ascarosides. A. oligospora senses ascarosides via conserved MAPK and cAMP-PKA pathways; however, the upstream receptors remain unknown. Here, using genomic, transcriptomic and functional analyses, we identified two families of G protein-coupled receptors (GPCRs) involved in sensing distinct nematode-derived cues. GPCRs homologous to yeast glucose receptors are required for ascaroside sensing, whereas Pth11-like GPCRs contribute to ascaroside-independent nematode sensing. Both GPCR classes activate conserved cAMP-PKA signalling to trigger trap development. This work demonstrates that predatory fungi use multiple GPCRs to sense several distinct nematode-derived cues for prey recognition and to enable a switch to a predatory lifestyle. Identification of these receptors reveals the molecular mechanisms of cross-kingdom communication via conserved pheromones also sensed by plants and animals.
Collapse
Affiliation(s)
- Chih-Yen Kuo
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Rebecca J Tay
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Hung-Che Lin
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Sheng-Chian Juan
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | | | - Yu-Chu Chang
- Department of Biochemistry and Molecular Cell Biology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Jason Hoki
- Boyce Thompson Institute, Cornell University, Ithaca, NY, USA
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Frank C Schroeder
- Boyce Thompson Institute, Cornell University, Ithaca, NY, USA
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Yen-Ping Hsueh
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and Graduate Institute of Life Science, National Defense Medical Center, Taipei, Taiwan.
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
| |
Collapse
|
4
|
Gates CA, Backos DS, Reigan P, Natale NR. The Lateral Metalation of Isoxazolo[3,4- d]pyridazinones towards Hit-to-Lead Development of Selective Positive Modulators of Metabotropic Glutamate Receptors. Molecules 2023; 28:6800. [PMID: 37836643 PMCID: PMC10574779 DOI: 10.3390/molecules28196800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/22/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Isoxazolo[3,4-d] pyridazinones ([3,4-d]s) were previously shown to have selective positive modulation at the metabotropic glutamate receptor (mGluR) Subtypes 2 and 4, with no functional cross-reactivity at mGluR1a, mGluR5, or mGluR8. Additional analogs were prepared to access more of the allosteric pocket and achieve higher binding affinity, as suggested by homology modeling. Two different sets of analogs were generated. One uses the fully formed [3,4-d] with an N6-aryl with and without halogens. These underwent successful selective lateral metalation and electrophilic quenching (LM&EQ) at the C3 of the isoxazole. In a second set of analogs, a phenyl group was introduced at the C4 position of the [3,4-d] ring via a condensation of 4-phenylacetyl-3-ethoxcarbonyl-5-methyl isoxazole with the corresponding hydrazine to generate the 3,4-ds 2b and 2j to 2n.
Collapse
Affiliation(s)
- Christina A Gates
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
| | - Donald S Backos
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, USA
| | - Philip Reigan
- Skaggs School of Pharmacy and Pharmaceutical Sciences, Anschutz Medical Campus, University of Colorado Denver, Aurora, CO 80045, USA
| | - Nicholas R Natale
- Department of Biomedical and Pharmaceutical Sciences, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
| |
Collapse
|
5
|
Lee-Rivera I, López E, López-Colomé AM. Diversification of PAR signaling through receptor crosstalk. Cell Mol Biol Lett 2022; 27:77. [PMID: 36088291 PMCID: PMC9463773 DOI: 10.1186/s11658-022-00382-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 09/02/2022] [Indexed: 11/17/2022] Open
Abstract
Protease activated receptors (PARs) are among the first receptors shown to transactivate other receptors: noticeably, these interactions are not limited to members of the same family, but involve receptors as diverse as receptor kinases, prostanoid receptors, purinergic receptors and ionic channels among others. In this review, we will focus on the evidence for PAR interactions with members of their own family, as well as with other types of receptors. We will discuss recent evidence as well as what we consider as emerging areas to explore; from the signalling pathways triggered, to the physiological and pathological relevance of these interactions, since this additional level of molecular cross-talk between receptors and signaling pathways is only beginning to be explored and represents a novel mechanism providing diversity to receptor function and play important roles in physiology and disease.
Collapse
|
6
|
Portales AE, Mustafá ER, McCarthy CI, Cornejo MP, Couto PM, Gironacci MM, Caramelo JJ, Perelló M, Raingo J. ACE2 internalization induced by a SARS-CoV-2 recombinant protein is modulated by angiotensin II type 1 and bradykinin 2 receptors. Life Sci 2022; 293:120284. [PMID: 35038454 PMCID: PMC8758573 DOI: 10.1016/j.lfs.2021.120284] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 12/20/2022]
Abstract
AIMS Angiotensin-converting enzyme 2 (ACE2) is a key regulator of the renin-angiotensin system (RAS) recently identified as the membrane receptor for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here we aim to study whether two receptors from RAS, the angiotensin receptor type 1 (AT1R) and the bradykinin 2 receptor (B2R) modulate ACE2 internalization induced by a recombinant receptor binding domain (RBD) of SARS-CoV-2 spike protein. Also, we investigated the impact of ACE2 coexpression on AT1R and B2R functionality. MATERIALS AND METHODS To study ACE2 internalization, we assessed the distribution of green fluorescent protein (GFP) signal in HEK293T cells coexpressing GFP-tagged ACE2 and AT1R, or B2R, or AT1R plus B2R in presence of RBD alone or in combination with AT1R or B2R ligands. To estimate ACE2 internalization, we classified GFP signal distribution as plasma membrane uniform GFP (PMU-GFP), plasma membrane clustered GFP (PMC-GFP) or internalized GFP and calculated its relative frequency. Additionally, we investigated the effect of ACE2 coexpression on AT1R and B2R inhibitory action on voltage-gated calcium channels (CaV2.2) currents by patch-clamp technique. KEY FINDINGS RBD induced ACE2-GFP internalization in a time-dependent manner. RBD-induced ACE2-GFP internalization was increased by angiotensin II and reduced by telmisartan in cells coexpressing AT1R. RBD-induced ACE2-GFP internalization was strongly inhibited by B2R co-expression. This effect was mildly modified by bradykinin and rescued by angiotensin II in presence of AT1R. ACE2 coexpression impacted on B2R- and AT1R-mediated inhibition of CaV2.2 currents. SIGNIFICANCE Our work contributes to understand the role of RAS modulators in the susceptibility to SARS-CoV-2 infection and severity of COVID-19.
Collapse
Affiliation(s)
- Andrea Estefanía Portales
- Laboratorio de Electrofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional de La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC), Calle 526 1499-1579, B1906APM Tolosa, Buenos Aires, Argentina
| | - Emilio Román Mustafá
- Laboratorio de Electrofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional de La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC), Calle 526 1499-1579, B1906APM Tolosa, Buenos Aires, Argentina
| | - Clara Inés McCarthy
- Laboratorio de Electrofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional de La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC), Calle 526 1499-1579, B1906APM Tolosa, Buenos Aires, Argentina
| | - María Paula Cornejo
- Laboratorio de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional de La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC), Calle 526 1499-1579, B1906APM Tolosa, Buenos Aires, Argentina
| | - Paula Monserrat Couto
- Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Mariela Mercedes Gironacci
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB, UBA-CONICET), Argentina
| | - Julio Javier Caramelo
- Fundación Instituto Leloir and Instituto de Investigaciones Bioquímicas de Buenos Aires, Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Argentina
| | - Mario Perelló
- Laboratorio de Neurofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional de La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC), Calle 526 1499-1579, B1906APM Tolosa, Buenos Aires, Argentina
| | - Jesica Raingo
- Laboratorio de Electrofisiología, Instituto Multidisciplinario de Biología Celular (IMBICE), Universidad Nacional de La Plata (UNLP), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) and Comisión de Investigaciones Científicas de la Provincia de Buenos Aires (CIC), Calle 526 1499-1579, B1906APM Tolosa, Buenos Aires, Argentina..
| |
Collapse
|
7
|
El Khamlichi C, Cobret L, Arrang JM, Morisset-Lopez S. BRET Analysis of GPCR Dimers in Neurons and Non-Neuronal Cells: Evidence for Inactive, Agonist, and Constitutive Conformations. Int J Mol Sci 2021; 22:ijms221910638. [PMID: 34638980 PMCID: PMC8508734 DOI: 10.3390/ijms221910638] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 09/24/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) are dimeric proteins, but the functional consequences of the process are still debated. Active GPCR conformations are promoted either by agonists or constitutive activity. Inverse agonists decrease constitutive activity by promoting inactive conformations. The histamine H3 receptor (H3R) is the target of choice for the study of GPCRs because it displays high constitutive activity. Here, we study the dimerization of recombinant and brain H3R and explore the effects of H3R ligands of different intrinsic efficacy on dimerization. Co-immunoprecipitations and Western blots showed that H3R dimers co-exist with monomers in transfected HEK 293 cells and in rodent brains. Bioluminescence energy transfer (BRET) analysis confirmed the existence of spontaneous H3R dimers, not only in living HEK 293 cells but also in transfected cortical neurons. In both cells, agonists and constitutive activity of the H3R decreased BRET signals, whereas inverse agonists and GTPγS, which promote inactive conformations, increased BRET signals. These findings show the existence of spontaneous H3R dimers not only in heterologous systems but also in native tissues, which are able to adopt a number of allosteric conformations, from more inactive to more active states.
Collapse
Affiliation(s)
- Chayma El Khamlichi
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d’Orléans, Rue Charles Sadron, CEDEX 2, 45071 Orléans, France; (C.E.K.); (L.C.)
| | - Laetitia Cobret
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d’Orléans, Rue Charles Sadron, CEDEX 2, 45071 Orléans, France; (C.E.K.); (L.C.)
| | - Jean-Michel Arrang
- Centre de Psychiatrie et Neurosciences, 2 ter Rue d’Alésia, 75014 Paris, France;
- Institut de Psychiatrie et Neurosciences de Paris, UMR_S1266 INSERM, Université Paris Descartes, 102 Rue de la Santé, 75014 Paris, France
| | - Séverine Morisset-Lopez
- Centre de Biophysique Moléculaire, CNRS UPR 4301, Université d’Orléans, Rue Charles Sadron, CEDEX 2, 45071 Orléans, France; (C.E.K.); (L.C.)
- Institut de Psychiatrie et Neurosciences de Paris, UMR_S1266 INSERM, Université Paris Descartes, 102 Rue de la Santé, 75014 Paris, France
- Correspondence: ; Tel.: +33-238257858
| |
Collapse
|
8
|
Shweta K, Basargekar A, Ratnaparkhi A. FGFR/Heartless and Smog interact synergistically to negatively regulate Fog mediated G-protein coupled receptor signaling in the Drosophila nervous system. G3-GENES GENOMES GENETICS 2021; 11:6174503. [PMID: 33729500 PMCID: PMC8022937 DOI: 10.1093/g3journal/jkaa029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Accepted: 10/27/2020] [Indexed: 11/13/2022]
Abstract
Folded gastrulation (Fog) is a secreted ligand that signals through the G-protein-coupled receptors Mist and Smog and the G-protein Concertina to activate downstream effectors to elicit cell-shape change during gastrulation. In the embryonic central nervous system (CNS), Fog has roles in axon guidance and glial morphogenesis. However, the elements of the pathway as well as mechanisms required for transducing the signal in this context have not been determined. We find that while Concertina is essential for Fog signaling, Mist is dispensable and Smog, surprisingly, functions as a negative regulator of the pathway in the CNS. Interestingly Heartless, a fibroblast growth factor receptor, also functions as a negative regulator. Furthermore, both Heartless and Smog interact in a synergistic manner to regulate Fog signaling. Our results thus identify Heartless and Smog as part of a common regulatory pathway that functions to restrict Fog signaling in the embryonic CNS and highlights the context-specific role for Fog receptors during development.
Collapse
Affiliation(s)
- Kumari Shweta
- MACS-Agharkar Research Institute (affiliated to SPPU, Pune), Developmental Biology Group, G.G. Agarkar Road, Pune 411 004, India
| | - Anagha Basargekar
- MACS-Agharkar Research Institute (affiliated to SPPU, Pune), Developmental Biology Group, G.G. Agarkar Road, Pune 411 004, India
| | - Anuradha Ratnaparkhi
- MACS-Agharkar Research Institute (affiliated to SPPU, Pune), Developmental Biology Group, G.G. Agarkar Road, Pune 411 004, India
| |
Collapse
|
9
|
Watabe M, Yoshimura H, Arjunan SNV, Kaizu K, Takahashi K. Signaling activations through G-protein-coupled-receptor aggregations. Phys Rev E 2020; 102:032413. [PMID: 33076007 DOI: 10.1103/physreve.102.032413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 08/31/2020] [Indexed: 06/11/2023]
Abstract
Eukaryotic cells transmit extracellular signal information to cellular interiors through the formation of a ternary complex made up of a ligand (or agonist), G-protein, and G-protein-coupled receptor (GPCR). Previously formalized theories of ternary complex formation have mainly assumed that observable states of receptors can only take the form of monomers. Here, we propose a multiary complex model of GPCR signaling activations via the vector representation of various unobserved aggregated receptor states. Our results from model simulations imply that receptor aggregation processes can govern cooperative effects in a regime inaccessible by previous theories. In particular, we show how the affinity of ligand-receptor binding can be largely varied by various oligomer formations in the low concentration range of G-protein stimulus.
Collapse
Affiliation(s)
- Masaki Watabe
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka 565-0874, Japan
| | - Hideaki Yoshimura
- School of Science, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-0033, Japan
| | - Satya N V Arjunan
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka 565-0874, Japan
- Lowy Cancer Research Centre, The University of New South Wales, Sydney 2052, Australia
| | - Kazunari Kaizu
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka 565-0874, Japan
| | - Koichi Takahashi
- Laboratory for Biologically Inspired Computing, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka 565-0874, Japan
- Institute for Advanced Biosciences, Keio University, Fujisawa, Kanagawa 252-8520, Japan
| |
Collapse
|
10
|
Zyma M, Pawliczak R. Characteristics and the role of purinergic receptors in pathophysiology with focus on immune response. Int Rev Immunol 2020; 39:97-117. [PMID: 32037918 DOI: 10.1080/08830185.2020.1723582] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The nucleotide adenosine-5'-triphosphate (ATP) is mostly thought to be energy carrier, but evidence presented in multiple studies proves ATP involvement into variety of processes, due to its neuromodulatory capabilities. ATP and its metabolite-adenosine, bind to the purinergic receptors, which are divided into two types: adenosine binding P1 receptor and ADP/ATP binding P2 receptor. These receptors are expressed in different tissues and organs. Recent studies report their immunomodulatory characteristics, connected with varying immunological processes, such as immunological response or antigen presentation. Besides, they seem to play an important role in medical conditions such as bronchial asthma or variety of cancers. In this article, we would like to review recent discoveries on the field of purinergic receptors research focusing on their role in immunological system, and shed a new light upon the importance of these receptors in modern medicine development.
Collapse
Affiliation(s)
- Marharyta Zyma
- Department of Immunopathology, Division of Biomedical Science, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| | - Rafał Pawliczak
- Department of Immunopathology, Division of Biomedical Science, Faculty of Medicine, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
11
|
Guidolin D, Marcoli M, Tortorella C, Maura G, Agnati LF. Adenosine A 2A-dopamine D 2 receptor-receptor interaction in neurons and astrocytes: Evidence and perspectives. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2019; 169:247-277. [PMID: 31952688 DOI: 10.1016/bs.pmbts.2019.11.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
The discovery of receptor-receptor interactions in the early 1980s, together with a more accurate focusing of allosteric mechanisms in proteins, expanded the knowledge on the G protein-coupled receptor (GPCR)-mediated signaling processes. GPCRs were seen to operate not only as monomers, but also as quaternary structures shaped by allosteric interactions. These integrative mechanisms can change the function of the GPCRs involved, leading to a sophisticated dynamic of the receptor assembly in terms of modulation of recognition and signaling. In this context, the heterodimeric complex formed by the adenosine A2A and the dopamine D2 receptors likely represents a prototypical example. The pharmacological evidence obtained, together with the tissue distribution of the A2A-D2 heteromeric complexes, suggested they could represent a target for new therapeutic strategies addressing significant disorders of the central nervous system. The research findings and the perspectives they offer from the therapeutic standpoint are the focus of the here presented discussion.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience, Section of Anatomy, University of Padova, Padova, Italy.
| | - Manuela Marcoli
- Department of Pharmacy and Center of Excellence for Biomedical Research, University of Genova, Genova, Italy
| | - Cinzia Tortorella
- Department of Neuroscience, Section of Anatomy, University of Padova, Padova, Italy
| | - Guido Maura
- Department of Pharmacy and Center of Excellence for Biomedical Research, University of Genova, Genova, Italy
| | - Luigi F Agnati
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy; Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
12
|
Edfawy M, Guedes JR, Pereira MI, Laranjo M, Carvalho MJ, Gao X, Ferreira PA, Caldeira G, Franco LO, Wang D, Cardoso AL, Feng G, Carvalho AL, Peça J. Abnormal mGluR-mediated synaptic plasticity and autism-like behaviours in Gprasp2 mutant mice. Nat Commun 2019; 10:1431. [PMID: 30926797 PMCID: PMC6440958 DOI: 10.1038/s41467-019-09382-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 03/07/2019] [Indexed: 12/13/2022] Open
Abstract
Autism spectrum disorder (ASD) is characterized by dysfunction in social interactions, stereotypical behaviours and high co-morbidity with intellectual disability. A variety of syndromic and non-syndromic neurodevelopmental disorders have been connected to alterations in metabotropic glutamate receptor (mGluR) signalling. These receptors contribute to synaptic plasticity, spine maturation and circuit development. Here, we investigate the physiological role of Gprasp2, a gene linked to neurodevelopmental disabilities and involved in the postendocytic sorting of G-protein-coupled receptors. We show that Gprasp2 deletion leads to ASD-like behaviour in mice and alterations in synaptic communication. Manipulating the levels of Gprasp2 bidirectionally modulates the surface availability of mGluR5 and produces alterations in dendritic complexity, spine density and synaptic maturation. Loss of Gprasp2 leads to enhanced hippocampal long-term depression, consistent with facilitated mGluR-dependent activation. These findings demonstrate a role for Gprasp2 in glutamatergic synapses and suggest a possible mechanism by which this gene is linked to neurodevelopmental diseases.
Collapse
Affiliation(s)
- Mohamed Edfawy
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal.,PhD Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, 3030-789, Coimbra, Portugal
| | - Joana R Guedes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal
| | - Marta I Pereira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Mariana Laranjo
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Mário J Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Xian Gao
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Key Laboratory of Brain Functional Genomics, Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai, 200062, China.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Pedro A Ferreira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Gladys Caldeira
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal
| | - Lara O Franco
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal.,PhD Program in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, 3030-789, Coimbra, Portugal
| | - Dongqing Wang
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Ana Luisa Cardoso
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.,Key Laboratory of Brain Functional Genomics, Institute of Cognitive Neuroscience, School of Psychology and Cognitive Science, East China Normal University, Shanghai, 200062, China.,Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Ana Luisa Carvalho
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal.,Department of Life Sciences, University of Coimbra, 3004-517, Coimbra, Portugal
| | - João Peça
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal. .,Institute for Interdisciplinary Research (IIIUC), University of Coimbra, 3030-789, Coimbra, Portugal.
| |
Collapse
|
13
|
Elevated pre-activation basal level of nuclear NF-κB in native macrophages accelerates LPS-induced translocation of cytosolic NF-κB into the cell nucleus. Sci Rep 2019; 9:4563. [PMID: 30872589 PMCID: PMC6418260 DOI: 10.1038/s41598-018-36052-5] [Citation(s) in RCA: 69] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/08/2018] [Indexed: 02/01/2023] Open
Abstract
Signaling via Toll-like receptor 4 (TLR4) in macrophages constitutes an essential part of the innate immune response to bacterial infections. Detailed and quantified descriptions of TLR4 signal transduction would help to understand and exploit the first-line response of innate immune defense. To date, most mathematical modelling studies were performed on transformed cell lines. However, properties of primary macrophages differ significantly. We therefore studied TLR4-dependent activation of NF-κB transcription factor in bone marrow-derived and peritoneal primary macrophages. We demonstrate that the kinetics of NF-κB phosphorylation and nuclear translocation induced by a wide range of bacterial lipopolysaccharide (LPS) concentrations in primary macrophages is much faster than previously reported for macrophage cell lines. We used a comprehensive combination of experiments and mathematical modeling to understand the mechanisms of this rapid response. We found that elevated basal NF-κB in the nuclei of primary macrophages is a mechanism increasing native macrophage sensitivity and response speed to the infection. Such pre-activated state of macrophages accelerates the NF-κB translocation kinetics in response to low agonist concentrations. These findings enabled us to refine and construct a new model combining both NF-κB phosphorylation and translocation processes and predict the existence of a negative feedback loop inactivating phosphorylated NF-κB.
Collapse
|
14
|
Jiang WY, Li Y, Li ZY, Cui ZJ. Permanent Photodynamic Cholecystokinin 1 Receptor Activation: Dimer-to-Monomer Conversion. Cell Mol Neurobiol 2018; 38:1283-1292. [PMID: 29869099 PMCID: PMC11482018 DOI: 10.1007/s10571-018-0596-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 05/30/2018] [Indexed: 02/07/2023]
Abstract
The G protein-coupled cholecystokinin 1 receptor (CCK1R) is activated permanently by type II photodynamic action (i.e., by singlet oxygen) in the freshly isolated rat pancreatic acini, in contrast to reversible activation by CCK. But how CCK1R is photodynamically activated is not known. Therefore, in the present work, we subjected membrane proteins extracted from isolated rat pancreatic acini to photodynamic action with photosensitiser sulphonated aluminium phthalocyanine (SALPC), and used reducing gel electrophoresis and Western blot to detect possible changes in CCK1R oligomerization status. Photodynamic action (SALPC 1 µM, light 36.7 mW cm- 2 × 10 min) was found to convert dimeric CCK1R nearly quantitatively to monomers. Such conversion was dependent on both irradiance (8.51-36.7 mW cm- 2) and irradiation time (1-20 min). Minimum effective irradiance was found to be 11.1 mW cm- 2 (× 10 min, with SALPC 1 µM), and brief photodynamic action (SALPC 1 µM, 36.7 mW cm- 2 × 1 min) was effective. Whilst CCK stimulation of purified membrane proteins alone had no effect on CCK1R dimer/monomer balance, sub-threshold photodynamic action (SALPC 100 nM, 36.7 mW cm- 2 × 10 min) plus CCK revealed a bell-shaped CCK dose response curve for CCK1R monomerization, which was remarkably similar to the dose response curve for CCK-stimulated amylase secretion in isolated rat pancreatic acini. These two lines of evidence together suggest that during photodynamic CCK1R activation, CCK1R is permanently monomerized, thus providing a unique approach for permanent G protein-coupled receptor (GPCR) activation which has not been achieved before.
Collapse
Affiliation(s)
- Wen Yi Jiang
- Institute of Cell Biology, Beijing Normal University, Beijing, 100875, China
| | - Yuan Li
- Institute of Cell Biology, Beijing Normal University, Beijing, 100875, China
| | - Zhi Ying Li
- Institute of Cell Biology, Beijing Normal University, Beijing, 100875, China
| | - Zong Jie Cui
- Institute of Cell Biology, Beijing Normal University, Beijing, 100875, China.
| |
Collapse
|
15
|
Effect of Site-Specific Intermolecular Lysine-Tryptophan Interactions on the Aggregation of Gramicidin-Based Peptides Leading to Pore Formation in Lipid Membranes. J Membr Biol 2018; 251:633-640. [PMID: 29995247 DOI: 10.1007/s00232-018-0040-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 07/09/2018] [Indexed: 10/28/2022]
Abstract
In contrast to the parent pentadecapeptide gramicidin A (gA), some of its cationic analogs have been shown previously to form large-diameter pores in lipid membranes. These pores are permeable to fluorescent dyes, which allows one to monitor pore formation by using the fluorescence de-quenching assay. According to the previously proposed model, the gA analog with lysine substituted for alanine at position 3, [Lys3]gA, forms pores by a homopentameric assembly of gramicidin double-stranded β-helical dimers. Here, we studied the newly synthesized analogs of [Lys3]gA with single, double and triple substitutions of isoleucines for tryptophans at positions 9, 11, 13, and 15. Replacement of any of the tryptophans of [Lys3]gA with isoleucine resulted in suppression of the pore-forming activity of the peptide, the effect being significantly dependent on the position of tryptophans. In particular, the peptide with a single substitution of tryptophan 13 showed much lower activity than the analogs with single substitutions at positions 9, 11, or 15. Of the peptides with double substitutions, the strongest suppression of the leakage was observed with tryptophans 13 and 15. In the case of triple substitutions, only the peptide retaining tryptophan 11 exhibited noticeable activity. It is concluded that tryptophans 11 and 13 contribute most to pore stabilization in the membrane, whereas tryptophan 9 is not so important for pore formation. Cation-π interactions between the lysine and tryptophan residues of the peptide are suggested to be crucial for the formation of the [Lys3]gA pore.
Collapse
|
16
|
Fievez V, Szpakowska M, Mosbah A, Arumugam K, Mathu J, Counson M, Beaupain N, Seguin-Devaux C, Deroo S, Baudy-Floc'h M, Chevigné A. Development of Mimokines, chemokine N terminus-based CXCR4 inhibitors optimized by phage display and rational design. J Leukoc Biol 2018; 104:343-357. [PMID: 29570832 DOI: 10.1002/jlb.3ma0118-007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 01/25/2018] [Indexed: 12/13/2022] Open
Abstract
The chemokine receptor CXCR4 (C-X-C chemokine receptor type 4 also known as fusin or CD184 (cluster of differentiation 184)) is implicated in various biological and pathological processes of the hematopoietic and immune systems. CXCR4 is also one of the major coreceptors for HIV-1 entry into target cells and is overexpressed in many cancers, supporting cell survival, proliferation, and migration. CXCR4 is thus an extremely relevant drug target. Among the different strategies to block CXCR4, chemokine-derived peptide inhibitors hold great therapeutic potential. In this study, we used the N-terminus of vCCL2/vMIPII, a viral CXCR4 antagonist chemokine, as a scaffold motif to engineer and select CXCR4 peptide inhibitors, called Mimokines, which imitate the chemokine-binding mode but display an enhanced receptor affinity, antiviral properties, and receptor selectivity. We first engineered a Mimokine phage displayed library based on the first 21 residues of vCCL2, in which cysteine 11 and 12 were fully randomized and screened it against purified CXCR4 stabilized in liposomes. We identified Mimokines displaying up to 4-fold higher affinity for CXCR4 when compared to the reference peptide and fully protected MT-4 cells against HIV-1 infection. These selected Mimokines were then subjected to dimerization, D-amino acid, and aza-β3-amino acid substitution to further enhance their potency and selectivity. Optimized Mimokines exhibited up to 120-fold enhanced CXCR4 binding (range of 20 nM) and more than 200-fold improved antiviral properties (≤ 1 μM) compared to the parental Mimokines. Interestingly, these optimized Mimokines also showed up to 25-fold weaker affinity for ACKR3/CXCR7 and may therefore serve as lead compounds for further development of more selective CXCR4 peptide inhibitors and probes.
Collapse
Affiliation(s)
- Virginie Fievez
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), House of BioHealth, 4354 Esch-sur-Alzette, Luxembourg
| | - Martyna Szpakowska
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), House of BioHealth, 4354 Esch-sur-Alzette, Luxembourg
| | - Amor Mosbah
- Université de Rennes 1, UMR CNRS 6226, 35042 Rennes, France
| | - Karthik Arumugam
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), House of BioHealth, 4354 Esch-sur-Alzette, Luxembourg
| | - Julie Mathu
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), House of BioHealth, 4354 Esch-sur-Alzette, Luxembourg
| | - Manuel Counson
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), House of BioHealth, 4354 Esch-sur-Alzette, Luxembourg
| | - Nadia Beaupain
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), House of BioHealth, 4354 Esch-sur-Alzette, Luxembourg
| | - Carole Seguin-Devaux
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), House of BioHealth, 4354 Esch-sur-Alzette, Luxembourg
| | - Sabrina Deroo
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), House of BioHealth, 4354 Esch-sur-Alzette, Luxembourg
| | | | - Andy Chevigné
- Department of Infection and Immunity, Immuno-Pharmacology and Interactomics, Luxembourg Institute of Health (LIH), House of BioHealth, 4354 Esch-sur-Alzette, Luxembourg
| |
Collapse
|
17
|
Mangini M, Iaccino E, Mosca MG, Mimmi S, D'Angelo R, Quinto I, Scala G, Mariggiò S. Peptide-guided targeting of GPR55 for anti-cancer therapy. Oncotarget 2018; 8:5179-5195. [PMID: 28029647 PMCID: PMC5354900 DOI: 10.18632/oncotarget.14121] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 11/21/2016] [Indexed: 12/21/2022] Open
Abstract
Expression of the lysophosphatidylinositol receptor GPR55 correlates with invasive potential of metastatic cells and bone metastasis formation of different types of tumors. These findings suggest a role for GPR55 signaling in cancer progression, including in lymphoproliferative diseases. Here, we screened a M13-phage-displayed random library using the bait of HEK293 cells that heterologously expressed full-length HA-GPR55. We selected a set of phagotopes that carried 7-mer insert peptides flanked by a pair of cysteine residues, which resulted in cyclized peptides. Sequencing of selected phagotopes dictated the primary structure for the synthetic FITC-labeled peptide P1, which was analyzed for binding specificity to immunoprecipitated HA-GPR55, and to endogenously expressed GPR55, using cells interfered or not for GPR55, as well as for co-localization imaging with HA-GPR55 at the membrane level. The peptide P1 stimulated GPR55 endocytosis and inhibited GPR55-dependent proliferation of EHEB and DeFew cells, two human B-lymphoblastoid cell lines. Our data support the potential therapeutic application of peptide ligands of GPR55 for targeting and inhibiting growth of neoplastic cells, which overexpress GPR55 and are dependent on GPR55 signaling for their proliferation.
Collapse
Affiliation(s)
- Maria Mangini
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Enrico Iaccino
- Department of Experimental and Clinical Medicine, University 'Magna Graecia' of Catanzaro, Catanzaro, Italy
| | | | - Selena Mimmi
- Department of Experimental and Clinical Medicine, University 'Magna Graecia' of Catanzaro, Catanzaro, Italy
| | - Rosa D'Angelo
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| | - Ileana Quinto
- Department of Experimental and Clinical Medicine, University 'Magna Graecia' of Catanzaro, Catanzaro, Italy
| | - Giuseppe Scala
- Department of Experimental and Clinical Medicine, University 'Magna Graecia' of Catanzaro, Catanzaro, Italy
| | - Stefania Mariggiò
- Institute of Protein Biochemistry, National Research Council, Naples, Italy
| |
Collapse
|
18
|
Guidolin D, Marcoli M, Tortorella C, Maura G, Agnati LF. G protein-coupled receptor-receptor interactions give integrative dynamics to intercellular communication. Rev Neurosci 2018; 29:703-726. [DOI: 10.1515/revneuro-2017-0087] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 01/01/2018] [Indexed: 01/14/2023]
Abstract
Abstract
The proposal of receptor-receptor interactions (RRIs) in the early 1980s broadened the view on the role of G protein-coupled receptors (GPCR) in the dynamics of the intercellular communication. RRIs, indeed, allow GPCR to operate not only as monomers but also as receptor complexes, in which the integration of the incoming signals depends on the number, spatial arrangement, and order of activation of the protomers forming the complex. The main biochemical mechanisms controlling the functional interplay of GPCR in the receptor complexes are direct allosteric interactions between protomer domains. The formation of these macromolecular assemblies has several physiologic implications in terms of the modulation of the signaling pathways and interaction with other membrane proteins. It also impacts on the emerging field of connectomics, as it contributes to set and tune the synaptic strength. Furthermore, recent evidence suggests that the transfer of GPCR and GPCR complexes between cells via the exosome pathway could enable the target cells to recognize/decode transmitters and/or modulators for which they did not express the pertinent receptors. Thus, this process may also open the possibility of a new type of redeployment of neural circuits. The fundamental aspects of GPCR complex formation and function are the focus of the present review article.
Collapse
Affiliation(s)
- Diego Guidolin
- Department of Neuroscience , University of Padova, via Gabelli 65 , I-35121 Padova , Italy
| | - Manuela Marcoli
- Department of Pharmacy and Center of Excellence for Biomedical Research , University of Genova , I-16126 Genova , Italy
| | - Cinzia Tortorella
- Department of Neuroscience , University of Padova, via Gabelli 65 , I-35121 Padova , Italy
| | - Guido Maura
- Department of Pharmacy and Center of Excellence for Biomedical Research , University of Genova , I-16126 Genova , Italy
| | - Luigi F. Agnati
- Department of Biomedical Sciences , University of Modena and Reggio Emilia , I-41121 Modena , Italy
- Department of Neuroscience , Karolinska Institutet , S-17177 Stockholm , Sweden
| |
Collapse
|
19
|
Ligand Binding Dynamics for Pre-dimerised G Protein-Coupled Receptor Homodimers: Linear Models and Analytical Solutions. Bull Math Biol 2018; 81:3542-3574. [PMID: 29349610 PMCID: PMC6722261 DOI: 10.1007/s11538-017-0387-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 12/21/2017] [Indexed: 11/25/2022]
Abstract
Evidence suggests that many G protein-coupled receptors (GPCRs) are bound together forming dimers. The implications of dimerisation for cellular signalling outcomes, and ultimately drug discovery and therapeutics, remain unclear. Consideration of ligand binding and signalling via receptor dimers is therefore required as an addition to classical receptor theory, which is largely built on assumptions of monomeric receptors. A key factor in developing theoretical models of dimer signalling is cooperativity across the dimer, whereby binding of a ligand to one protomer affects the binding of a ligand to the other protomer. Here, we present and analyse linear models for one-ligand and two-ligand binding dynamics at homodimerised receptors, as an essential building block in the development of dimerised receptor theory. For systems at equilibrium, we compute analytical solutions for total bound labelled ligand and derive conditions on the cooperativity factors under which multiphasic log dose–response curves are expected. This could help explain data extracted from pharmacological experiments that do not fit to the standard Hill curves that are often used in this type of analysis. For the time-dependent problems, we also obtain analytical solutions. For the single-ligand case, the construction of the analytical solution is straightforward; it is bi-exponential in time, sharing a similar structure to the well-known monomeric competition dynamics of Motulsky–Mahan. We suggest that this model is therefore practically usable by the pharmacologist towards developing insights into the potential dynamics and consequences of dimerised receptors.
Collapse
|
20
|
Strader ME, Aglyamova GV, Matz MV. Molecular characterization of larval development from fertilization to metamorphosis in a reef-building coral. BMC Genomics 2018; 19:17. [PMID: 29301490 PMCID: PMC5755313 DOI: 10.1186/s12864-017-4392-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Accepted: 12/15/2017] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Molecular mechanisms underlying coral larval competence, the ability of larvae to respond to settlement cues, determine their dispersal potential and are potential targets of natural selection. Here, we profiled competence, fluorescence and genome-wide gene expression in embryos and larvae of the reef-building coral Acropora millepora daily throughout 12 days post-fertilization. RESULTS Gene expression associated with competence was positively correlated with transcriptomic response to the natural settlement cue, confirming that mature coral larvae are "primed" for settlement. Rise of competence through development was accompanied by up-regulation of sensory and signal transduction genes such as ion channels, genes involved in neuropeptide signaling, and G-protein coupled receptor (GPCRs). A drug screen targeting components of GPCR signaling pathways confirmed a role in larval settlement behavior and metamorphosis. CONCLUSIONS These results gives insight into the molecular complexity underlying these transitions and reveals receptors and pathways that, if altered by changing environments, could affect dispersal capabilities of reef-building corals. In addition, this dataset provides a toolkit for asking broad questions about sensory capacity in multicellular animals and the evolution of development.
Collapse
Affiliation(s)
- Marie E Strader
- Department of Integrative Biology, The University of Texas at Austin, 1 University Station C0990, Austin, TX, 78712, USA.
| | - Galina V Aglyamova
- Department of Integrative Biology, The University of Texas at Austin, 1 University Station C0990, Austin, TX, 78712, USA
| | - Mikhail V Matz
- Department of Integrative Biology, The University of Texas at Austin, 1 University Station C0990, Austin, TX, 78712, USA
| |
Collapse
|
21
|
Rasmussen AQ, Jørgensen NR, Schwarz P. Identification and Functional Characterization of a Novel Mutation in the Human Calcium-Sensing Receptor That Co-Segregates With Autosomal-Dominant Hypocalcemia. Front Endocrinol (Lausanne) 2018; 9:200. [PMID: 29743878 PMCID: PMC5930847 DOI: 10.3389/fendo.2018.00200] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 04/10/2018] [Indexed: 12/31/2022] Open
Abstract
The human calcium-sensing receptor (CASR) is the key controller of extracellular Cao2+ homeostasis, and different mutations in the CASR gene have been linked to different calcium diseases, such as familial hypocalciuric hypercalcemia, severe hyperparathyroidism, autosomal-dominant hypocalcemia (ADH), and Bartter's syndrome type V. In this study, two generations of a family with biochemically and clinically confirmed ADH who suffered severe muscle pain, arthralgia, tetany, abdominal pain, and fatigue were evaluated for mutations in the CASR gene. The study comprises genotyping of all family members, functional characterization of a potential mutant receptor by in vitro analysis related to the wild-type receptor to reveal an association between the genotype and phenotype in the affected family members. The in vitro analysis of functional characteristics includes measurements of inositol trisphosphate accumulation, Ca2+ mobilization in response to [Ca2+]o-stimulation and receptor expression. The results reveal a significant leftward shift of inositol trisphosphate accumulation as a result of the "gain-of-function" mutant receptor and surprisingly a normalization of the response in (Ca2+)i release in the downstream pathway and additionally the maximal response of (Ca2+)i release was significantly decreased compared to the wild type. However, no gross differences were seen in D126V and the D126V/WT CASR dimeric >250 kDa band expression compared to the WT receptor, however, the D126V and D126V/WT CASR immature ~140 kDa species appear to have reduced expression compared to the WT receptor. In conclusion, in this study, a family with a clinical diagnosis of ADH in two generations was evaluated to identify a mutation in the CASR gene and reveal an association between genotype and phenotype in the affected family members. The clinical condition was caused by a novel, activating, missense mutation (D126V) in the CASR gene and the in vitro functional characteristics of the mutation co-segregated with their individual phenotype.
Collapse
Affiliation(s)
- Anne Qvist Rasmussen
- Research Centre of Ageing and Osteoporosis, Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
- *Correspondence: Anne Qvist Rasmussen,
| | - Niklas Rye Jørgensen
- Department of Clinical Biochemistry, Rigshospitalet, Copenhagen, Denmark
- Institute of Clinical Research, University of Southern, Odense, Denmark
| | - Peter Schwarz
- Research Centre of Ageing and Osteoporosis, Department of Endocrinology, Rigshospitalet, Copenhagen, Denmark
- Faculty of Health Sciences, Copenhagen University, Copenhagen, Denmark
| |
Collapse
|
22
|
Altwaijry NA, Baron M, Wright DW, Coveney PV, Townsend-Nicholson A. An Ensemble-Based Protocol for the Computational Prediction of Helix-Helix Interactions in G Protein-Coupled Receptors using Coarse-Grained Molecular Dynamics. J Chem Theory Comput 2017; 13:2254-2270. [PMID: 28383913 PMCID: PMC5557214 DOI: 10.1021/acs.jctc.6b01246] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
![]()
The
accurate identification of the specific points of interaction
between G protein-coupled receptor (GPCR) oligomers is essential for
the design of receptor ligands targeting oligomeric receptor targets.
A coarse-grained molecular dynamics computer simulation approach would
provide a compelling means of identifying these specific protein–protein
interactions and could be applied both for known oligomers of interest
and as a high-throughput screen to identify novel oligomeric targets.
However, to be effective, this in silico modeling must provide accurate,
precise, and reproducible information. This has been achieved recently
in numerous biological systems using an ensemble-based all-atom molecular
dynamics approach. In this study, we describe an equivalent methodology
for ensemble-based coarse-grained simulations. We report the performance
of this method when applied to four different GPCRs known to oligomerize
using error analysis to determine the ensemble size and individual
replica simulation time required. Our measurements of distance between
residues shown to be involved in oligomerization of the fifth transmembrane
domain from the adenosine A2A receptor are in very good
agreement with the existing biophysical data and provide information
about the nature of the contact interface that cannot be determined
experimentally. Calculations of distance between rhodopsin, CXCR4,
and β1AR transmembrane domains reported to form contact
points in homodimers correlate well with the corresponding measurements
obtained from experimental structural data, providing an ability to predict
contact interfaces computationally. Interestingly, error analysis
enables identification of noninteracting regions. Our results confirm
that GPCR interactions can be reliably predicted using this novel
methodology.
Collapse
Affiliation(s)
- Nojood A Altwaijry
- Institute of Structural and Molecular Biology, Research Department of Structural and Molecular Biology, Division of Biosciences, University College London , London, WC1E 6BT, United Kingdom.,King Saud University , Riyadh, Kingdom of Saudi Arabia
| | - Michael Baron
- Institute of Structural and Molecular Biology, Research Department of Structural and Molecular Biology, Division of Biosciences, University College London , London, WC1E 6BT, United Kingdom
| | - David W Wright
- Centre for Computational Science, Department of Chemistry, University College London , London WC1H 0AJ, United Kingdom
| | - Peter V Coveney
- Centre for Computational Science, Department of Chemistry, University College London , London WC1H 0AJ, United Kingdom
| | - Andrea Townsend-Nicholson
- Institute of Structural and Molecular Biology, Research Department of Structural and Molecular Biology, Division of Biosciences, University College London , London, WC1E 6BT, United Kingdom
| |
Collapse
|
23
|
Hasegawa K, Kudoh S, Ito T. Somatostatin receptor staining in FFPE sections using a ligand derivative dye as an alternative to immunostaining. PLoS One 2017; 12:e0172030. [PMID: 28182792 PMCID: PMC5300255 DOI: 10.1371/journal.pone.0172030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2016] [Accepted: 01/30/2017] [Indexed: 11/18/2022] Open
Abstract
The confirmation of target expression in tissues is a prerequisite for molecular-targeted therapy. However, difficulties are sometimes associated with the production of appropriate antibodies against receptors. We herein developed a ligand derivative dye for the staining of receptors. The somatostatin receptor (sstr) was selected as the target and FITC-octreotate as the detective agent. We performed a blot analysis to detect sstr in the transfer membrane. The sstr2 recombinant protein or cell lysate from a small cell lung carcinoma cell line (H69) was boiled and loaded onto SDS-PAGE, and the proteins were transferred to a membrane. Even after denaturing processes, FITC-octreotate still bound sstr on the membrane. Furthermore, FITC-octreotate depicted the expression of sstr in formalin-fixed and paraffin-embedded (FFPE) sections, a method that we named ligand derivative staining (LDS). The accuracies of immunostaining and LDS were compared at the points of the detection of sstr using FFPE sections of 30 neuroendocrine tumor specimens. The sensitivity of LDS was 81.8%, while those of immunostaining using anti-sstr2 and sstr5 antibodies were 72.7% and 63.6%, respectively. Thus, LDS appears to be superior to immunostaining. A ligand derivative may be used as a substitute for antibodies, and has the potential to support economical, simple, and accurate detection methods.
Collapse
Affiliation(s)
- Koki Hasegawa
- Department of Pathology and Experimental Medicine, Kumamoto University, Graduate School of Medical Sciences, Kumamoto, Japan
- Center for Instrumental Analysis, Kyoto Pharmaceutical University, Kyoto, Japan
| | - Shinji Kudoh
- Department of Pathology and Experimental Medicine, Kumamoto University, Graduate School of Medical Sciences, Kumamoto, Japan
| | - Takaaki Ito
- Department of Pathology and Experimental Medicine, Kumamoto University, Graduate School of Medical Sciences, Kumamoto, Japan
| |
Collapse
|
24
|
Chang K, Roche KW. Structural and molecular determinants regulating mGluR5 surface expression. Neuropharmacology 2016; 115:10-19. [PMID: 27211252 DOI: 10.1016/j.neuropharm.2016.04.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 02/29/2016] [Accepted: 04/26/2016] [Indexed: 01/29/2023]
Abstract
Trafficking of G protein-coupled receptors (GPCRs) to the plasma membrane is a pivotal process to fulfill their biological functions. Metabotropic glutamate receptors (mGluRs; mGluR1-8) are expressed throughout the CNS and are important for modulating synaptic transmission and plasticity. Group I mGluRs, including mGluR1 and mGluR5, have long intracellular C-terminal tails containing multiple protein binding domains and sites for phosphorylation and ER retention. We have now investigated some of the structural determinants for mGluR5 trafficking to the plasma membrane by studying a series of truncations and ligand binding mutants. We also take advantage of dimer formation between the extracellular domain (ECD) of mGluR5 and design an ECD based surface-binding assay to evaluate dimerization and surface expression of mGluR5 containing various truncations or point mutations. We found that the C terminus is not essential for mGluR5 surface expression. In contrast, the 7th transmembrane domain (TM7) plays a critical role in its surface expression in both heterologous cells and neurons. Furthermore, a ligand binding mutation within the ECD of mGluR5 (Y64A/T174A) that blocks ligand binding impairs both surface expression and dimerization of mGluR5 in neurons. The integrity of both the whole 7TM domain and the C- terminal tail of mGluR5 are also important for stabilizing dimerization with the ECD. Thus multiple domains regulate dimerization and trafficking of mGluR5. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.
Collapse
Affiliation(s)
- Kai Chang
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 35, Room 2C903, Bethesda, MD 20892, USA
| | - Katherine W Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Building 35, Room 2C903, Bethesda, MD 20892, USA.
| |
Collapse
|
25
|
Arnatt CK, Zhang Y. Bivalent ligands targeting chemokine receptor dimerization: molecular design and functional studies. Curr Top Med Chem 2016; 14:1606-18. [PMID: 25159160 DOI: 10.2174/1568026614666140827144752] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 05/14/2014] [Accepted: 05/14/2014] [Indexed: 12/27/2022]
Abstract
Increasing evidence has shown that chemokine receptors may form functional dimers with unique pharmacological profiles. A common practice to characterize such G protein-coupled receptor dimerization processes is to apply bivalent ligands as chemical probes which can interact with both receptors simultaneously. Currently, two chemokine receptor dimers have been studied by applying bivalent compounds: the CXCR4-CXCR4 homodimer and the CCR5-MOR heterodimer. These bivalent compounds have revealed how dimerization influences receptor function and may lead to novel therapeutics. Future design of bivalent ligands for chemokine receptor dimers may be aided with the recently available CXCR4 homodimer, and CCR5 monomer crystal structures by more accurately simulating chemokine receptors and their dimers.
Collapse
Affiliation(s)
| | - Yan Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, Richmond, VA 23298, USA.
| |
Collapse
|
26
|
In Search of Concomitant Alterations of Dopaminergic and Neurotensinergic Systems in Stress Conditions. Neurochem Res 2016; 41:423-30. [DOI: 10.1007/s11064-016-1849-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2015] [Revised: 01/21/2016] [Accepted: 01/25/2016] [Indexed: 01/03/2023]
|
27
|
Martinucci I, Blandizzi C, de Bortoli N, Bellini M, Antonioli L, Tuccori M, Fornai M, Marchi S, Colucci R. Genetics and pharmacogenetics of aminergic transmitter pathways in functional gastrointestinal disorders. Pharmacogenomics 2016; 16:523-39. [PMID: 25916523 DOI: 10.2217/pgs.15.12] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Functional gastrointestinal disorders (FGIDs) are highly prevalent syndromes, without evident underlying organic causes. Their pathogenesis is multifactorial in nature, with a combination of environmental and genetic factors contributing to their clinical manifestations, for which most of current treatments are not satisfactory. It is acknowledged that amine mediators (noradrenaline, dopamine and serotonin) play pivotal regulatory actions on gut functions and visceral sensation. In addition, drugs of therapeutic interest for FGIDs act on these transmitter pathways. The present article reviews current knowledge on the impact of genetics and pharmacogenetics of aminergic pathways on FGID pathophysiology, clinical presentations, symptom severity and medical management, in an attempt of highlighting the most relevant evidence and point out issues that should be addressed in future investigations.
Collapse
Affiliation(s)
- Irene Martinucci
- Gastroenterology Unit, Department of Translational Research & New Technologies in Medicine, University of Pisa, Via Paradisa 2, I-56124 Pisa, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Santa Maria C, Cheng Z, Li A, Wang J, Shoback D, Tu CL, Chang W. Interplay between CaSR and PTH1R signaling in skeletal development and osteoanabolism. Semin Cell Dev Biol 2016; 49:11-23. [PMID: 26688334 PMCID: PMC4761456 DOI: 10.1016/j.semcdb.2015.12.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 12/05/2015] [Indexed: 12/01/2022]
Abstract
Parathyroid hormone (PTH)-related peptide (PTHrP) controls the pace of pre- and post-natal growth plate development by activating the PTH1R in chondrocytes, while PTH maintains mineral and skeletal homeostasis by modulating calciotropic activities in kidneys, gut, and bone. The extracellular calcium-sensing receptor (CaSR) is a member of family C, G protein-coupled receptor, which regulates mineral and skeletal homeostasis by controlling PTH secretion in parathyroid glands and Ca(2+) excretion in kidneys. Recent studies showed the expression of CaSR in chondrocytes, osteoblasts, and osteoclasts and confirmed its non-redundant roles in modulating the recruitment, proliferation, survival, and differentiation of the cells. This review emphasizes the actions of CaSR and PTH1R signaling responses in cartilage and bone and discusses how these two signaling cascades interact to control growth plate development and maintain skeletal metabolism in physiological and pathological conditions. Lastly, novel therapeutic regimens that exploit interrelationship between the CaSR and PTH1R are proposed to produce more robust osteoanabolism.
Collapse
Affiliation(s)
- Christian Santa Maria
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Zhiqiang Cheng
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Alfred Li
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Jiali Wang
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Dolores Shoback
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Chia-Ling Tu
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA
| | - Wenhan Chang
- Endocrine Research Unit, University of California, San Francisco, Veterans Affairs Medical Center, San Francisco, CA, USA.
| |
Collapse
|
29
|
Abstract
Since their discovery, G protein-coupled receptors (GPCRs) constitute one of the most studied proteins leading to important discoveries and perspectives in terms of their biology and implication in physiology and pathophysiology. This is mostly linked to the remarkable advances in the development and application of the biophysical resonance energy transfer (RET)-based approaches, including bioluminescence and fluorescence resonance energy transfer (BRET and FRET, respectively). Indeed, BRET and FRET have been extensively applied to study different aspects of GPCR functioning such as their activation and regulation either statically or dynamically, in real-time and intact cells. Consequently, our view on GPCRs has considerably changed opening new challenges for the study of GPCRs in their native tissues in the aim to get more knowledge on how these receptors control the biological responses. Moreover, the technological aspect of this field of research promises further developments for robust and reliable new RET-based assays that may be compatible with high-throughput screening as well as drug discovery programs.
Collapse
Affiliation(s)
- Mohammed Akli Ayoub
- Biologie et Bioinformatique des Systèmes de Signalisation, Institut National de la Recherche Agronomique, UMR85, Unité Physiologie de la Reproduction et des Comportements; CNRS, UMR7247, Nouzilly, France; LE STUDIUM(®) Loire Valley Institute for Advanced Studies, Orléans, France.
| |
Collapse
|
30
|
Abstract
OBJECTIVE Monocyte inflammatory processes are fundamental events in AIDS pathogenesis. HIV-1 matrix protein p17, released from infected cells, was found to exert an interleukin (IL)-8 chemokine-like activity on human monocytes, promoting their trafficking and sustaining inflammatory processes, after binding to CXCR1. A haplotype of the CXCR1 gene (CXCR1_300_142) has been associated with slow HIV disease progression. Here, we determine how CXCR1 genetic variations impact on p17 biological activity. DESIGN/METHODS/RESULTS Our results show that Jurkat cells overexpressing CXCR1 or the receptor carrying single polymorphism CXCR1_300 or CXCR1_142 are able to adhere and migrate in response to both IL-8 and p17. On the contrary, Jurkat cells overexpressing CXCR1_300_142 and monocytes of individuals with such CXCR1 polymorphisms lose the capacity to adhere and migrate in response to p17, but not to their physiological ligand IL-8. Surface plasmon resonance (SPR) and multispectral imaging flow cytometry showed that p17 bound with similar affinity to CXCR1 and CXCR1_300_142. Moreover, whereas p17 was able to activate CXCR1, it was incapable of functionally interacting with CXCR1_300_142 by phosphorylating extracellular signal-regulated kinase 1/2, which regulates chemokine-induced cellular responses. Finally, mutagenesis studies showed that, unlike IL-8, p17 does not use Glu-Leu-Arg-like motifs to activate CXCR1. CONCLUSIONS Our results, showing the inability of p17 to activate CXCR1_300_142, a receptor found to be expressed on immune cells of patients with a low progression of HIV disease, point to a crucial role of p17 in AIDS pathogenesis. Our findings herein call for an exploration of the therapeutic potential of blocking the p17/CXCR1 axis in HIV infection.
Collapse
|
31
|
Siddiquee K, Hampton J, McAnally D, May L, Smith L. The apelin receptor inhibits the angiotensin II type 1 receptor via allosteric trans-inhibition. Br J Pharmacol 2014; 168:1104-17. [PMID: 22935142 DOI: 10.1111/j.1476-5381.2012.02192.x] [Citation(s) in RCA: 112] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Revised: 08/21/2012] [Accepted: 08/27/2012] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND AND PURPOSE The apelin receptor (APJ) is often co-expressed with the angiotensin II type-1 receptor (AT1) and acts as an endogenous counter-regulator. Apelin antagonizes Ang II signalling, but the precise molecular mechanism has not been elucidated. Understanding this interaction may lead to new therapies for the treatment of cardiovascular disease. EXPERIMENTAL APPROACH The physical interaction of APJ and AT1 receptors was detected by co-immunoprecipitation and bioluminescence resonance energy transfer (BRET). Functional and pharmacological interactions were measured by G-protein-dependent signalling and recruitment of β-arrestin. Allosterism and cooperativity between APJ and AT1 were measured by radioligand binding assays. KEY RESULTS Apelin, but not Ang II, induced APJ : AT1 heterodimerization forced AT1 into a low-affinity state, reducing Ang II binding. Likewise, apelin mediated a concentration-dependent depression in the maximal production of inositol phosphate (IP(1) ) and β-arrestin recruitment to AT1 in response to Ang II. The signal depression approached a limit, the magnitude of which was governed by the cooperativity indicative of a negative allosteric interaction. Fitting the data to an operational model of allosterism revealed that apelin-mediated heterodimerization significantly reduces Ang II signalling efficacy. These effects were not observed in the absence of apelin. CONCLUSIONS AND IMPLICATIONS Apelin-dependent heterodimerization between APJ and AT1 causes negative allosteric regulation of AT1 function. As AT1 is significant in the pathogenesis of cardiovascular disease, these findings suggest that impaired apelin and APJ function may be a common underlying aetiology. LINKED ARTICLE This article is commented on by Goupil et al., pp. 1101-1103 of this issue. To view this commentary visit http://dx.doi.org/10.1111/bph.12040.
Collapse
Affiliation(s)
- K Siddiquee
- Cardiovascular Pathobiology Program, Sanford Burnham Medical Research Institute at Lake Nona, Orlando, FL, USA
| | | | | | | | | |
Collapse
|
32
|
Distinct human and mouse membrane trafficking systems for sweet taste receptors T1r2 and T1r3. PLoS One 2014; 9:e100425. [PMID: 25029362 PMCID: PMC4100762 DOI: 10.1371/journal.pone.0100425] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 05/27/2014] [Indexed: 11/23/2022] Open
Abstract
The sweet taste receptors T1r2 and T1r3 are included in the T1r taste receptor family that belongs to class C of the G protein-coupled receptors. Heterodimerization of T1r2 and T1r3 is required for the perception of sweet substances, but little is known about the mechanisms underlying this heterodimerization, including membrane trafficking. We developed tagged mouse T1r2 and T1r3, and human T1R2 and T1R3 and evaluated membrane trafficking in human embryonic kidney 293 (HEK293) cells. We found that human T1R3 surface expression was only observed when human T1R3 was coexpressed with human T1R2, whereas mouse T1r3 was expressed without mouse T1r2 expression. A domain-swapped chimera and truncated human T1R3 mutant showed that the Venus flytrap module and cysteine-rich domain (CRD) of human T1R3 contain a region related to the inhibition of human T1R3 membrane trafficking and coordinated regulation of human T1R3 membrane trafficking. We also found that the Venus flytrap module of both human T1R2 and T1R3 are needed for membrane trafficking, suggesting that the coexpression of human T1R2 and T1R3 is required for this event. These results suggest that the Venus flytrap module and CRD receive taste substances and play roles in membrane trafficking of human T1R2 and T1R3. These features are different from those of mouse receptors, indicating that human T1R2 and T1R3 are likely to have a novel membrane trafficking system.
Collapse
|
33
|
C5L2 is required for C5a-triggered receptor internalization and ERK signaling. Cell Signal 2014; 26:1409-19. [PMID: 24631530 DOI: 10.1016/j.cellsig.2014.02.021] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2014] [Accepted: 02/24/2014] [Indexed: 12/26/2022]
Abstract
C5L2 is a receptor that binds to C5a and belongs to the family of G protein-coupled receptors, but its role in physiological C5a-mediated responses remains under debate. Here we show that, like the canonical C5a receptor C5aR, C5L2 plays a pro-inflammatory role in a murine model of acute experimental colitis. We demonstrate that C5L2 physically interacts with C5aR and is required for optimal C5a-mediated C5aR internalization and associated ERK activation. Abrogation of C5a-induced receptor internalization by treatment with the dynamin inhibitor dynasore(TM) impaired C5a-induced MEK and ERK signaling. Although the presence of C5aR alone was sufficient to recruit the scaffold protein β-arrestin1 to the cell membrane in response to C5a stimulation, it was inadequate to mediate AP2 recruitment and subsequent C5aR internalization. Expression of C5L2 allowed normal internalization of C5aR in response to C5a stimulation, followed by normal ERK signaling. Thus, our work reveals an essential role for C5L2 in C5a-triggered, AP2-dependent C5aR internalization and downstream ERK signaling.
Collapse
|
34
|
Hazari PP, Schulz J, Vimont D, Chadha N, Allard M, Szlosek-Pinaud M, Fouquet E, Mishra AK. A new SiF-Dipropargyl glycerol scaffold as a versatile prosthetic group to design dimeric radioligands: synthesis of the [(18) F]BMPPSiF tracer to image serotonin receptors. ChemMedChem 2013; 9:337-49. [PMID: 24376058 DOI: 10.1002/cmdc.201300458] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Indexed: 11/12/2022]
Abstract
A novel SiX-dipropargyl glycerol scaffold (X: H, F, or (18) F) was developed as a versatile prosthetic group that provides technical advantages for the preparation of dimeric radioligands based on silicon fluoride acceptor pre- or post-labeling with fluorine-18. Rapid conjugation with the prosthetic group takes place in microwave-assisted click conjugation under mild conditions. Thus, a bivalent homodimeric SiX-dipropargyl glycerol derivatized radioligand, [(18) F]BMPPSiF, with enhanced affinity was developed by using click conjugation. High uptake of the radioligand was demonstrated in 5-HT1A receptor-rich regions in the brain with positron emission tomography. Molecular docking studies (rigid protein-flexible ligand) of BMPPSiF and known antagonists (WAY-100635, MPPF, and MefWAY) with monomeric, dimeric, and multimeric 5-HT1A receptor models were performed, with the highest G score obtained for docked BMPPSiF: -6.766 as compared with all three antagonists on the monomeric model. Multimeric induced-fit docking was also performed to visualize the comparable mode of binding under in vivo conditions, and a notably improved G score of -8.455 was observed for BMPPSiF. These data directly correlate the high binding potential of BMPPSiF with the bivalent binding mode obtained in the biological studies. The present study warrants wide application of the SiX-dipropargyl glycerol prosthetic group in the development of ligands for imaging with enhanced affinity markers for specific targeting based on peptides, nucleosides, and lipids.
Collapse
Affiliation(s)
- Puja Panwar Hazari
- Division of Cyclotron and Radiopharamceutical Sciences, Brig. SK Mazumdar Road, Delhi 110054 (India)
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Bagher AM, Laprairie RB, Kelly MEM, Denovan-Wright EM. Co-expression of the human cannabinoid receptor coding region splice variants (hCB₁) affects the function of hCB₁ receptor complexes. Eur J Pharmacol 2013; 721:341-54. [PMID: 24091169 DOI: 10.1016/j.ejphar.2013.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 08/26/2013] [Accepted: 09/04/2013] [Indexed: 10/26/2022]
Abstract
The human type 1 cannabinoid (hCB1) receptor is expressed at high levels in the central nervous system. mRNA variants of the coding region of this receptor, human cannabinoid hCB1a and hCB1b receptors, have been identified, their biological function remains unclear. The present study demonstrated that the three human cannabinoid hCB1 coding region variants are expressed in the human and monkey (Macaca fascicularis) brain. Western blot analyses of homogenates from different regions of the monkey brain demonstrated that proteins with the expected molecular weights of the cannabinoid CB1, CB1a and CB1b receptors were co-expressed throughout the brain. Given the co-localization of these receptors, we hypothesized that physical interactions between the three splice variants may affect cannabinoid pharmacology. The human cannabinoid hCB1, hCB1a, and hCB1b receptors formed homodimers and heterodimers, as determined by BRET in transiently transfected HEK 293A cells. We found that the co-expression of the human cannabinoid hCB1 and each of the splice variants increased cell surface expression of the human cannabinoid hCB1 receptor and increased Gi/o-dependent ERK phosphorylation in response to cannabinoid agonists. Therefore, the human cannabinoid hCB1 coding region splice variants play an important physiological role in the activity of the endocannabinoid system.
Collapse
Affiliation(s)
- Amina M Bagher
- Department of Pharmacology, Dalhousie University, 6E Sir Charles Tupper Medical Bldg., 5850 College St., Halifax, NS, Canada B3H 4R2
| | | | | | | |
Collapse
|
36
|
Abstract
Hypertensive disorders are life-threatening diseases with high morbidity and mortality, affecting billions of individuals worldwide. A multitude of underlying conditions may contribute to hypertension, thus the need for a plethora of treatment options to identify the approach that best meets the needs of individual patients. A growing body of evidence indicates that (1) autoantibodies that bind to and activate the major angiotensin II type I (AT₁) receptor exist in the circulation of patients with hypertensive disorders, (2) these autoantibodies contribute to disease pathophysiology, (3) antibody titers correlate to the severity of the disease, and (4) efforts to block or remove these pathogenic autoantibodies have therapeutic potential. These autoantibodies, termed AT₁ agonistic autoantibodies have been extensively characterized in preeclampsia, a life-threatening hypertensive condition of pregnancy. As reviewed here, these autoantibodies cause symptoms of preeclampsia when injected into pregnant mice. Somewhat surprisingly, these auto antibodies also appear in 3 animal models of preeclampsia. However, the occurrence of AT₁ agonistic autoantibodies is not restricted to pregnancy. These autoantibodies are prevalent among kidney transplant recipients who develop severe transplant rejection and malignant hypertension during the first week after transplantation. AT₁ agonistic autoantibodies are also highly abundant among a group of patients with essential hypertension that are refractory to standard therapy. More recently these autoantibodies have been seen in patients with the autoimmune disease, systemic sclerosis. These 3 examples extend the clinical impact of AT₁ agonistic autoantibodies beyond pregnancy. Research reviewed here raises the intriguing possibility that preeclampsia and other hypertensive conditions are autoimmune diseases characterized by the presence of pathogenic autoantibodies that activate the major angiotensin receptor, AT₁. These pathogenic autoantibodies could serve as presymptomatic biomarkers and therapeutic targets, thereby providing improved medical management for these conditions.
Collapse
Affiliation(s)
- Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas Medical School at Houston, Houston, TX 77030, USA.
| | | |
Collapse
|
37
|
Shim J, Coop A, MacKerell AD. Molecular details of the activation of the μ opioid receptor. J Phys Chem B 2013; 117:7907-17. [PMID: 23758404 DOI: 10.1021/jp404238n] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Molecular details of μ opioid receptor activations were obtained using molecular dynamics simulations of the receptor in the presence of three agonists, three antagonists, and a partial agonist and on the constitutively active T279K mutant. Agonists have a higher probability of direct interactions of their basic nitrogen (N) with Asp147 as compared with antagonists, indicating that direct ligand-Asp147 interactions modulate activation. Medium-size substituents on the basic N of antagonists lead to steric interactions that perturb N-Asp147 interactions, while additional favorable interactions occur with larger basic N substituents, such as in N-phenethylnormorphine, restoring N-Asp147 interactions, leading to agonism. With the orvinols, the increased size of the C19 substituent in buprenorphine over diprenorphine leads to increased interactions with residues adjacent to Asp147, partially overcoming the presence of the cyclopropyl N substituent, such that buprenorphine is a partial agonist. Results also indicate different conformational properties of the intracellular regions of the transmembrane helices in agonists versus antagonists.
Collapse
Affiliation(s)
- Jihyun Shim
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, USA
| | | | | |
Collapse
|
38
|
Hurevich M, Ratner-Hurevich M, Tal-Gan Y, Shalev DE, Ben-Sasson SZ, Gilon C. Backbone cyclic helix mimetic of chemokine (C-C motif) receptor 2: a rational approach for inhibiting dimerization of G protein-coupled receptors. Bioorg Med Chem 2013; 21:3958-66. [PMID: 23706536 DOI: 10.1016/j.bmc.2013.03.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Revised: 02/28/2013] [Accepted: 03/01/2013] [Indexed: 10/27/2022]
Abstract
The transmembrane helical bundle of G protein-coupled receptors (GPCRs) dimerize through helix-helix interactions in response to inflammatory stimulation. A strategy was developed to target the helical dimerization site of GPCRs by peptidomimetics with drug like properties. The concept was demonstrated by selecting a potent backbone cyclic helix mimetic from a library that derived from the dimerization region of chemokine (C-C motif) receptor 2 (CCR2) that is a key player in Multiple Sclerosis. We showed that CCR2 based backbone cyclic peptide having a stable helix structure inhibits specific CCR2-mediated chemotactic migration.
Collapse
Affiliation(s)
- Mattan Hurevich
- Institute of Chemistry, The Hebrew University of Jerusalem, Safra Campus, Givat Ram, Jerusalem 91904, Israel
| | | | | | | | | | | |
Collapse
|
39
|
Rivero-Müller A, Jonas KC, Hanyaloglu AC, Huhtaniemi I. Di/Oligomerization of GPCRs—Mechanisms and Functional Significance. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2013; 117:163-85. [DOI: 10.1016/b978-0-12-386931-9.00007-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
40
|
Almabouada F, Diaz-Ruiz A, Rabanal-Ruiz Y, Peinado JR, Vazquez-Martinez R, Malagon MM. Adiponectin receptors form homomers and heteromers exhibiting distinct ligand binding and intracellular signaling properties. J Biol Chem 2012; 288:3112-25. [PMID: 23255609 DOI: 10.1074/jbc.m112.404624] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Adiponectin binds to two widely expressed receptors (AdipoR1 and AdipoR2) that contain seven transmembrane domains but, unlike G-protein coupled receptors, present an extracellular C terminus and a cytosolic N terminus. Recently, AdipoR1 was found to associate in high order complexes. However, it is still unknown whether AdipoR2 may also form homomers or heteromers with AdipoR1 or if such interactions may be functionally relevant. Herein, we have analyzed the oligomerization pattern of AdipoRs by FRET and immunoprecipitation and evaluated both the internalization of AdipoRs in response to various adiponectin isoforms and the effect of adiponectin binding to different AdipoR combinations on AMP-activated protein kinase phosphorylation and peroxisome proliferator-activated receptor α activation. Transfection of HEK293AD cells with AdipoR1 and AdipoR2 showed that both receptors colocalize at both the plasma membrane and the endoplasmic reticulum. Co-transfection with the different AdipoR pairs yielded high FRET efficiencies in non-stimulated cells, which indicates that AdipoR1 and AdipoR2 form homo- and heteromeric complexes under resting conditions. Live FRET imaging suggested that both homo- and heteromeric AdipoR complexes dissociate in response to adiponectin, but heteromers separate faster than homomers. Finally, phosphorylation of AMP-activated protein kinase in response to adiponectin was delayed in cells wherein heteromer formation was favored. In sum, our findings indicate that AdipoR1 and AdipoR2 form homo- and heteromers that present unique interaction behaviors and signaling properties. This raises the possibility that the pleiotropic, tissue-dependent functions of adiponectin depend on the expression levels of AdipoR1 and AdipoR2 and, therefore, on the steady-state proportion of homo- and heteromeric complexes.
Collapse
Affiliation(s)
- Farid Almabouada
- Department of Cell Biology, Physiology, and Immunology, Instituto Maimonides de Investigacion Biomedica de Cordoba/University Hospital Reina Sofia, University of Cordoba, 14014 Cordoba, Spain
| | | | | | | | | | | |
Collapse
|
41
|
Interaction of structure-specific and promiscuous G-protein-coupled receptors mediates small-molecule signaling in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2012; 109:9917-22. [PMID: 22665789 DOI: 10.1073/pnas.1202216109] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A chemically diverse family of small-molecule signals, the ascarosides, control developmental diapause (dauer), olfactory learning, and social behaviors of the nematode model organism, Caenorhabditis elegans. The ascarosides act upstream of conserved signaling pathways, including the insulin, TGF-β, serotonin, and guanylyl cyclase pathways; however, the sensory processes underlying ascaroside function are poorly understood. Because ascarosides often are multifunctional and show strongly synergistic effects, characterization of their receptors will be essential for understanding ascaroside biology and may provide insight into molecular mechanisms that produce synergistic outcomes in small-molecule sensing. Based on DAF-8 immunoprecipitation, we here identify two G-protein-coupled receptors, DAF-37 and DAF-38, which cooperatively mediate ascaroside perception. daf-37 mutants are defective in all responses to ascr#2, one of the most potent dauer-inducing ascarosides, although this mutant responds normally to other ascarosides. In contrast, daf-38 mutants are partially defective in responses to several different ascarosides. Through cell-specific overexpression, we show that DAF-37 regulates dauer when expressed in ASI neurons and adult behavior when expressed in ASK neurons. Using a photoaffinity-labeled ascr#2 probe and amplified luminescence assays (AlphaScreen), we demonstrate that ascr#2 binds to DAF-37. Photobleaching fluorescent energy transfer assays revealed that DAF-37 and DAF-38 form heterodimers, and we show that heterodimerization strongly increases cAMP inhibition in response to ascr#2. These results suggest that that the ascarosides' intricate signaling properties result in part from the interaction of highly structure-specific G-protein-coupled receptors such as DAF-37 with more promiscuous G-protein-coupled receptors such as DAF-38.
Collapse
|
42
|
Meyer D, Voigt A, Widmayer P, Borth H, Huebner S, Breit A, Marschall S, de Angelis MH, Boehm U, Meyerhof W, Gudermann T, Boekhoff I. Expression of Tas1 taste receptors in mammalian spermatozoa: functional role of Tas1r1 in regulating basal Ca²⁺ and cAMP concentrations in spermatozoa. PLoS One 2012; 7:e32354. [PMID: 22427794 PMCID: PMC3303551 DOI: 10.1371/journal.pone.0032354] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 01/25/2012] [Indexed: 12/03/2022] Open
Abstract
BACKGROUND During their transit through the female genital tract, sperm have to recognize and discriminate numerous chemical compounds. However, our current knowledge of the molecular identity of appropriate chemosensory receptor proteins in sperm is still rudimentary. Considering that members of the Tas1r family of taste receptors are able to discriminate between a broad diversity of hydrophilic chemosensory substances, the expression of taste receptors in mammalian spermatozoa was examined. METHODOLOGY/PRINCIPAL FINDINGS The present manuscript documents that Tas1r1 and Tas1r3, which form the functional receptor for monosodium glutamate (umami) in taste buds on the tongue, are expressed in murine and human spermatozoa, where their localization is restricted to distinct segments of the flagellum and the acrosomal cap of the sperm head. Employing a Tas1r1-deficient mCherry reporter mouse strain, we found that Tas1r1 gene deletion resulted in spermatogenic abnormalities. In addition, a significant increase in spontaneous acrosomal reaction was observed in Tas1r1 null mutant sperm whereas acrosomal secretion triggered by isolated zona pellucida or the Ca²⁺ ionophore A23187 was not different from wild-type spermatozoa. Remarkably, cytosolic Ca²⁺ levels in freshly isolated Tas1r1-deficient sperm were significantly higher compared to wild-type cells. Moreover, a significantly higher basal cAMP concentration was detected in freshly isolated Tas1r1-deficient epididymal spermatozoa, whereas upon inhibition of phosphodiesterase or sperm capacitation, the amount of cAMP was not different between both genotypes. CONCLUSIONS/SIGNIFICANCE Since Ca²⁺ and cAMP control fundamental processes during the sequential process of fertilization, we propose that the identified taste receptors and coupled signaling cascades keep sperm in a chronically quiescent state until they arrive in the vicinity of the egg - either by constitutive receptor activity and/or by tonic receptor activation by gradients of diverse chemical compounds in different compartments of the female reproductive tract.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Calcium/metabolism
- Cyclic AMP/metabolism
- Female
- Gene Expression
- Humans
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Male
- Mice
- Mice, 129 Strain
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Microscopy, Confocal
- Models, Biological
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Receptors, G-Protein-Coupled/genetics
- Receptors, G-Protein-Coupled/metabolism
- Receptors, G-Protein-Coupled/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Sperm Head/metabolism
- Spermatozoa/metabolism
- Testis/cytology
- Testis/metabolism
- Red Fluorescent Protein
Collapse
Affiliation(s)
- Dorke Meyer
- Walther-Straub Institute of Pharmacology and
Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Anja Voigt
- German Institute of Nutrition,
Potsdam-Rehbruecke, Germany
- Institute for Neural Signal Transduction,
Center for Molecular Neurobiology, Hamburg, Germany
| | - Patricia Widmayer
- Institute of Physiology, University of
Hohenheim, Stuttgart, Germany
| | - Heike Borth
- Walther-Straub Institute of Pharmacology and
Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Sandra Huebner
- German Institute of Nutrition,
Potsdam-Rehbruecke, Germany
| | - Andreas Breit
- Walther-Straub Institute of Pharmacology and
Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Susan Marschall
- Institute of Experimental Genetics,
Helmholtz-Zentrum, Munich, Germany
| | | | - Ulrich Boehm
- Institute for Neural Signal Transduction,
Center for Molecular Neurobiology, Hamburg, Germany
| | | | - Thomas Gudermann
- Walther-Straub Institute of Pharmacology and
Toxicology, Ludwig-Maximilians-University, Munich, Germany
| | - Ingrid Boekhoff
- Walther-Straub Institute of Pharmacology and
Toxicology, Ludwig-Maximilians-University, Munich, Germany
| |
Collapse
|
43
|
Abstract
The LH receptor (LHR) and FSH receptor (FSHR), collectively termed the gonadotropin receptors, are members of the Family A of GPCRs. The gonadotropin receptors each contain N-linked carbohydrates that are not directly involved in hormone binding, but contribute to the proper folding, and therefore, cell surface expression of the receptor. Loss-of-function mutations of an LHR or FSHR results in decreased target cell responsiveness. Most inactivating mutations cause receptor misfolding, resulting in the retention of the mutant in its immature form in the endoplasmic reticulum. A membrane-permeable allosteric agonist of the LHR has been shown to serve as a pharmacological chaperone for misfolded and intracellularly retained LHRs by promoting their cell surface expression. Wild-type LHR and FSHR each form homodimers and heterodimers while in the ER. Therefore, when wild-type receptor is co-expressed with a misfolded mutant, the misfolded receptor dimerizes with immature wild-type receptor in the ER, causing a dominant-negative effect on cell surface expression of the mature wild-type receptor. Notably, the propensity for homodimerization is not affected by the activation status of the receptor. However, within a receptor dimer, the activity of one protomer may allosterically regulate the other protomer. Therefore, the dimerization of the gonadotropin receptors appears to be an obligate process that is part of the normal itinerary for trafficking to the cell surface and, once there, the dimerized receptors allow for additional modulations of cell signaling.
Collapse
Affiliation(s)
- Deborah L Segaloff
- Department of Molecular Physiology and Biophysics, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA,
| |
Collapse
|
44
|
Xia Y, Kellems RE. Receptor-activating autoantibodies and disease: preeclampsia and beyond. Expert Rev Clin Immunol 2011; 7:659-74. [PMID: 21895478 DOI: 10.1586/eci.11.56] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The research reviewed in this article provides examples of autoantibody-mediated receptor activation that likely contributes to disease. The classic example is Graves' hyperthyroidism, in which autoantibodies activate the thyroid-stimulating hormone receptor resulting in overproduction of thyroid hormones. Other compelling examples come from the cardiovascular literature and include agonistic autoantibodies targeting the cardiac β(1)-adrenergic receptor, which are associated with dilated cardiomyopathy. Autoantibodies capable of activating α(1)-adrenergic receptors are associated with refractory hypertension and cardiomyopathy. A prominent example is preeclampsia, a hypertensive disease of pregnancy, characterized by the presence of autoantibodies that activate the major angiotensin receptor, AT(1). AT(1) receptor-activating autoantibodies are also observed in kidney transplant recipients suffering from severe vascular rejection and malignant hypertension. AT(1) receptor-activating autoantibodies and antibodies that activate the endothelin-1 receptor, ET(A), are prevalent in individuals diagnosed with systemic sclerosis. Thus, the presence of agonistic autoantibodies directed to G protein-coupled receptors has been observed in numerous cardiovascular disease states. Rapidly emerging evidence indicates that receptor-activating autoantibodies contribute to disease, and that efforts to detect and remove these pathogenic autoantibodies or block their actions will provide promising therapeutic possibilities.
Collapse
Affiliation(s)
- Yang Xia
- Department of Biochemistry and Molecular Biology, The University of Texas Medical School, Houston, TX 77030, USA.
| | | |
Collapse
|
45
|
Zhang Y, Yang W, Chen L, Shi Y, Li G, Zhou N. Development of a novel DnaE intein-based assay for quantitative analysis of G-protein-coupled receptor internalization. Anal Biochem 2011; 417:65-72. [PMID: 21726524 DOI: 10.1016/j.ab.2011.06.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2011] [Revised: 06/01/2011] [Accepted: 06/03/2011] [Indexed: 01/12/2023]
Abstract
G-protein-coupled receptor (GPCR) internalization provides a G-protein-subtype-independent method for assaying agonist-stimulated activation of receptors. We have developed a novel assay that allows quantitative analysis of GPCR internalization based on the interaction between activated GPCRs and β-arrestin2 and on Nostoc punctiforme DnaE intein-mediated reconstitution of Renilla luciferase fragments. This assay system was validated using four functionally divergent GPCRs treated with agonists and antagonists. The EC(50) values obtained for the known agonists and antagonists are in close agreement with the results of previous reports, indicating that this assay system is sensitive enough to permit quantification of GPCR internalization. This rapid and quantitative assay, therefore, could be used universally as a functional cell-based assay for GPCR high-throughput screening during drug discovery.
Collapse
Affiliation(s)
- Yaping Zhang
- Institute of Biochemistry, College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | | | | | | | | | | |
Collapse
|
46
|
Guidolin D, Ciruela F, Genedani S, Guescini M, Tortorella C, Albertin G, Fuxe K, Agnati LF. Bioinformatics and mathematical modelling in the study of receptor–receptor interactions and receptor oligomerization. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1808:1267-83. [DOI: 10.1016/j.bbamem.2010.09.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2010] [Revised: 08/31/2010] [Accepted: 09/26/2010] [Indexed: 10/19/2022]
|
47
|
Ma AWS, Dong JY, Ma D, Wells JW. Cleavage-resistant fusion proteins of the M(2) muscarinic receptor and Gα(i1). Homotropic and heterotropic effects in the binding of ligands. Biochim Biophys Acta Gen Subj 2011; 1810:592-602. [PMID: 21397664 DOI: 10.1016/j.bbagen.2011.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Revised: 01/28/2011] [Accepted: 03/02/2011] [Indexed: 11/30/2022]
Abstract
BACKGROUND G protein-coupled receptors fused to a Gα-subunit are functionally similar to their unfused counterparts. They offer an intriguing view into the nature of the receptor-G protein complex, but their usefulness depends upon the stability of the fusion. METHODS Fusion proteins of the M(2) muscarinic receptor and the α-subunit of G(i1) were expressed in CHO and Sf9 cells, extracted in digitonin-cholate, and examined for their binding properties and their electrophoretic mobility on western blots. RESULTS Receptor fused to native α(i1) underwent proteolysis near the point of fusion to release a fragment with the mobility of α(i1). The cleavage was prevented by truncation of the α-subunit at position 18. Binding of the agonist oxotremorine-M to the stable fusion protein from Sf9 cells was biphasic, and guanylylimidodiphosphate promoted an apparent interconversion of sites from higher to lower affinity. With receptor from CHO cells, the apparent capacity for N-[(3)H]methylscopolamine was 60% of that for [(3)H]quinuclidinylbenzilate; binding at saturating concentrations of the latter was inhibited in a noncompetitive manner at low concentrations of unlabeled N-methylscopolamine. CONCLUSIONS A stable fusion protein of the M(2) receptor and truncated α(i1) resembles the native receptor-G protein complex with respect to the guanyl nucleotide-sensitive binding of agonists and the noncompetitive binding of antagonists. GENERAL SIGNIFICANCE Release of the α-subunit is likely to occur with other such fusion proteins, rendering the data ambiguous or misleading. The properties of a chemically stable fusion protein support the notion that signaling proceeds via a stable multimeric complex of receptor and G protein.
Collapse
Affiliation(s)
- Amy W-S Ma
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
48
|
Nemoto W, Fukui K, Toh H. GRIPDB - G protein coupled Receptor Interaction Partners DataBase. J Recept Signal Transduct Res 2011; 31:199-205. [DOI: 10.3109/10799893.2011.563312] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
49
|
Donnellan PD, Kimbembe CC, Reid HM, Kinsella BT. Identification of a novel endoplasmic reticulum export motif within the eighth α-helical domain (α-H8) of the human prostacyclin receptor. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2011; 1808:1202-18. [PMID: 21223948 DOI: 10.1016/j.bbamem.2011.01.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Revised: 12/20/2010] [Accepted: 01/03/2011] [Indexed: 01/20/2023]
Abstract
The human prostacyclin receptor (hIP) undergoes agonist-dependent trafficking involving a direct interaction with Rab11a GTPase. The region of interaction was localised to a 14 residue Rab11a binding domain (RBD) within the proximal carboxyl-terminal (C)-tail domain of the hIP, consisting of Val(299)-Val(307) within the eighth helical domain (α-H8) adjacent to the palmitoylated residues at Cys(308)-Cys(311). However, the factors determining the anterograde transport of the newly synthesised hIP from the endoplasmic reticulum (ER) to the plasma membrane (PM) have not been identified. The aim of the current study was to identify the major ER export motif(s) within the hIP initially by investigating the role of Lys residues in its maturation and processing. Through site-directed and Ala-scanning mutational studies in combination with analyses of protein expression and maturation, functional analyses of ligand binding, agonist-induced intracellular signalling and confocal image analyses, it was determined that Lys(297), Arg(302) and Lys(304) located within α-H8 represent the critical determinants of a novel ER export motif of the hIP. Furthermore, while substitution of those critical residues significantly impaired maturation and processing of the hIP, replacement of the positively charged Lys with Arg residues, and vice versa, was functionally permissible. Hence, this study has identified a novel 8 residue ER export motif within the functionally important α-H8 of the hIP. This ER export motif, defined by "K/R(X)(4)K/R(X)K/R," has a strict requirement for positively charged, basic Lys/Arg residues at the 1st, 6th and 8th positions and appears to be evolutionarily conserved within IP sequences from mouse to man.
Collapse
MESH Headings
- Amino Acid Motifs
- Amino Acid Sequence
- Arginine/chemistry
- Arginine/genetics
- Arginine/metabolism
- Binding Sites
- Blotting, Western
- Calcium/metabolism
- Calnexin/metabolism
- Computational Biology
- Endoplasmic Reticulum/metabolism
- HEK293 Cells
- Humans
- Lysine/chemistry
- Lysine/genetics
- Lysine/metabolism
- Microscopy, Confocal
- Models, Molecular
- Molecular Sequence Data
- Mutagenesis, Site-Directed
- Mutation
- Protein Binding
- Protein Structure, Secondary
- Protein Structure, Tertiary
- Protein Transport
- Radioligand Assay
- Receptors, Epoprostenol/chemistry
- Receptors, Epoprostenol/genetics
- Receptors, Epoprostenol/metabolism
- Sequence Homology, Amino Acid
Collapse
Affiliation(s)
- Peter D Donnellan
- School of Biomeolecular and Biomedical Sciences, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | |
Collapse
|
50
|
Parhamifar L, Sime W, Yudina Y, Vilhardt F, Mörgelin M, Sjölander A. Ligand-induced tyrosine phosphorylation of cysteinyl leukotriene receptor 1 triggers internalization and signaling in intestinal epithelial cells. PLoS One 2010; 5:e14439. [PMID: 21203429 PMCID: PMC3010979 DOI: 10.1371/journal.pone.0014439] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2010] [Accepted: 11/30/2010] [Indexed: 12/17/2022] Open
Abstract
Background Leukotriene D4 (LTD4) belongs to the bioactive lipid group known as eicosanoids and has implications in pathological processes such as inflammation and cancer. Leukotriene D4 exerts its effects mainly through two different G-protein-coupled receptors, CysLT1 and CysLT2. The high affinity LTD4 receptor CysLT1R exhibits tumor-promoting properties by triggering cell proliferation, survival, and migration in intestinal epithelial cells. In addition, increased expression and nuclear localization of CysLT1R correlates with a poorer prognosis for patients with colon cancer. Methodology/Principal Findings Using a proximity ligation assay and immunoprecipitation, this study showed that endogenous CysLT1R formed heterodimers with its counter-receptor CysLT2R under basal conditions and that LTD4 triggers reduced dimerization of CysLTRs in intestinal epithelial cells. This effect was dependent upon a parallel LTD4-induced increase in CysLT1R tyrosine phosphorylation. Leukotriene D4 also led to elevated internalization of CysLT1Rs from the plasma membrane and a simultaneous increase at the nucleus. Using sucrose, a clathrin endocytic inhibitor, dominant-negative constructs, and siRNA against arrestin-3, we suggest that a clathrin-, arrestin-3, and Rab-5-dependent process mediated the internalization of CysLT1R. Altering the CysLT1R internalization process at either the clathrin or the arrestin-3 stage led to disruption of LTD4-induced Erk1/2 activation and up-regulation of COX-2 mRNA levels. Conclusions/Significance Our data suggests that upon ligand activation, CysLT1R is tyrosine-phosphorylated and released from heterodimers with CysLT2R and, subsequently, internalizes from the plasma membrane to the nuclear membrane in a clathrin-, arrestin-3-, and Rab-5-dependent manner, thus, enabling Erk1/2 signaling and downstream transcription of the COX-2 gene.
Collapse
Affiliation(s)
- Ladan Parhamifar
- Cell and Experimental Pathology, Department of Laboratory Medicine, Clinical Research Center, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Wondossen Sime
- Cell and Experimental Pathology, Department of Laboratory Medicine, Clinical Research Center, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Yuliana Yudina
- Cell and Experimental Pathology, Department of Laboratory Medicine, Clinical Research Center, Lund University, Skåne University Hospital, Malmö, Sweden
| | - Frederik Vilhardt
- Institute of Cellular and Molecular Medicine, Panum Institute, Copenhagen University, Copenhagen, Denmark
| | - Matthias Mörgelin
- Infectious Medicine, Department of Clinical Science, Lund University, Lund, Sweden
| | - Anita Sjölander
- Cell and Experimental Pathology, Department of Laboratory Medicine, Clinical Research Center, Lund University, Skåne University Hospital, Malmö, Sweden
- * E-mail:
| |
Collapse
|