1
|
Yang LK, Wang W, Guo DY, Dong B. Non-canonical signaling initiated by hormone-responsive G protein-coupled receptors from subcellular compartments. Pharmacol Ther 2025; 266:108788. [PMID: 39722422 DOI: 10.1016/j.pharmthera.2024.108788] [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: 06/14/2024] [Revised: 11/13/2024] [Accepted: 12/12/2024] [Indexed: 12/28/2024]
Abstract
G protein-coupled receptors (GPCRs), the largest family of membrane receptors in the mammalian genomes, regulate almost all known physiological processes by transducing numerous extracellular stimuli including almost two-thirds of endogenous hormones and neurotransmitters. The traditional view held that GPCR signaling occurs exclusively at the cell surface, where the receptors bind with the ligands and undergo conformational changes to recruit and activate heterotrimeric G proteins. However, with the application of advanced biochemical and biophysical techniques, this conventional model is challenged by the elucidation of spatiotemporal GPCR activation with the evidence that receptors can signal from subcellular compartments to exhibit various molecular and cellular responses with physiological and pathophysiological relevance. Thus, this 'location bias' of GPCR signaling has become another layer of complexity of GPCR signal transduction. In this review, we generally introduce the development of the concept of compartmentalized GPCR signaling and comprehensively summarize the receptors reported to be localized on the membranes of different intracellular organelles. We review the physiological functions of these compartmentalized GPCRs with emphasis on some well-characterized prototypical hormone/neurotransmitter-binding receptors, including β2-adrenergic receptor, opioid receptors, parathyroid hormone type 1 receptor, thyroid-stimulating hormone receptor, cannabinoid receptor type 1, and metabotropic glutamate receptor 5, as examples. In addition, the therapeutic implications of compartmentalized GPCR signaling by introducing lipophilic or hydrophilic ligands for intracellular targeting, lipid conjugation anchor drugs, and strategy to modulate receptor internalization/resensitization, are highlighted and open new avenues in GPCR pharmacology and therapeutics.
Collapse
Affiliation(s)
- Li-Kun Yang
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China
| | - Wei Wang
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada; Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
| | - Dong-Yu Guo
- Department of Clinical Laboratory, Xiamen Huli Guoyu Clinic, Co., Ltd., Xiamen, China
| | - Bo Dong
- Fang Zongxi Center for Marine EvoDevo, MoE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, China; Insititute of Evolution & Marine Biodiversity, Ocean University of China, Qingdao 266003, China..
| |
Collapse
|
2
|
Allen BG, Merlen C, Branco AF, Pétrin D, Hébert TE. Understanding the impact of nuclear-localized GPCRs on cellular signalling. Cell Signal 2024; 123:111358. [PMID: 39181220 DOI: 10.1016/j.cellsig.2024.111358] [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: 07/24/2024] [Revised: 08/14/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
G protein-coupled receptors (GPCRs) have historically been associated with signalling events driven from the plasma membrane. More recently, signalling from endosomes has been recognized as a feature of internalizing receptors. However, there was little consideration given to the notion that GPCRs can be targeted to distinct subcellular locations that did not involve an initial trafficking to the cell surface. Here, we focus on the evidence for and the potential impact of GPCR signalling specifically initiated from the nuclear membrane. We also discuss the possibilities for selectively targeting this and other internal pools of receptors as novel venues for drug discovery.
Collapse
Affiliation(s)
- Bruce G Allen
- Montreal Heart Institute, Montréal, Québec H1T 1C8, Canada; Departments of Biochemistry and Molecular Medicine, Medicine, Pharmacology and Physiology, Université de Montréal, Montréal, Québec H3T 1J4, Canada
| | | | - Ana F Branco
- Montreal Heart Institute, Montréal, Québec H1T 1C8, Canada
| | - Darlaine Pétrin
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada
| | - Terence E Hébert
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Québec H3G 1Y6, Canada.
| |
Collapse
|
3
|
Molinaro G, Bowles JE, Croom K, Gonzalez D, Mirjafary S, Birnbaum SG, Razak KA, Gibson JR, Huber KM. Female-specific dysfunction of sensory neocortical circuits in a mouse model of autism mediated by mGluR5 and estrogen receptor α. Cell Rep 2024; 43:114056. [PMID: 38581678 PMCID: PMC11112681 DOI: 10.1016/j.celrep.2024.114056] [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: 08/17/2023] [Revised: 01/26/2024] [Accepted: 03/20/2024] [Indexed: 04/08/2024] Open
Abstract
Little is known of the brain mechanisms that mediate sex-specific autism symptoms. Here, we demonstrate that deletion of the autism spectrum disorder (ASD)-risk gene, Pten, in neocortical pyramidal neurons (NSEPten knockout [KO]) results in robust cortical circuit hyperexcitability selectively in female mice observed as prolonged spontaneous persistent activity states. Circuit hyperexcitability in females is mediated by metabotropic glutamate receptor 5 (mGluR5) and estrogen receptor α (ERα) signaling to mitogen-activated protein kinases (Erk1/2) and de novo protein synthesis. Pten KO layer 5 neurons have a female-specific increase in mGluR5 and mGluR5-dependent protein synthesis. Furthermore, mGluR5-ERα complexes are generally elevated in female cortices, and genetic reduction of ERα rescues enhanced circuit excitability, protein synthesis, and neuron size selectively in NSEPten KO females. Female NSEPten KO mice display deficits in sensory processing and social behaviors as well as mGluR5-dependent seizures. These results reveal mechanisms by which sex and a high-confidence ASD-risk gene interact to affect brain function and behavior.
Collapse
Affiliation(s)
- Gemma Molinaro
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jacob E Bowles
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Katilynne Croom
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, USA
| | - Darya Gonzalez
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Saba Mirjafary
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Shari G Birnbaum
- Department of Psychiatry, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Khaleel A Razak
- Graduate Neuroscience Program, University of California, Riverside, Riverside, CA, USA; Department of Psychology, University of California, Riverside, Riverside, CA, USA
| | - Jay R Gibson
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Kimberly M Huber
- Department of Neuroscience, O'Donnell Brain Institute, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
4
|
Klauer MJ, Willette BKA, Tsvetanova NG. Functional diversification of cell signaling by GPCR localization. J Biol Chem 2024; 300:105668. [PMID: 38272232 PMCID: PMC10882132 DOI: 10.1016/j.jbc.2024.105668] [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: 08/23/2023] [Revised: 11/13/2023] [Accepted: 11/29/2023] [Indexed: 01/27/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors and a critical class of regulators of mammalian physiology. Also known as seven transmembrane receptors (7TMs), GPCRs are ubiquitously expressed and versatile, detecting a diverse set of endogenous stimuli, including odorants, neurotransmitters, hormones, peptides, and lipids. Accordingly, GPCRs have emerged as the largest class of drug targets, accounting for upward of 30% of all prescription drugs. The view that ligand-induced GPCR responses originate exclusively from the cell surface has evolved to reflect accumulating evidence that receptors can elicit additional waves of signaling from intracellular compartments. These events in turn shape unique cellular and physiological outcomes. Here, we discuss our current understanding of the roles and regulation of compartmentalized GPCR signaling.
Collapse
Affiliation(s)
- Matthew J Klauer
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Blair K A Willette
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Nikoleta G Tsvetanova
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
5
|
Molinaro G, Bowles JE, Croom K, Gonzalez D, Mirjafary S, Birnbaum S, Razak KA, Gibson JR, Huber KM. Female specific dysfunction of sensory neocortical circuits in a mouse model of autism mediated by mGluR5 and Estrogen Receptor α. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.08.10.552857. [PMID: 37609208 PMCID: PMC10441407 DOI: 10.1101/2023.08.10.552857] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/24/2023]
Abstract
Autism manifests differently in males and females and the brain mechanisms that mediate these sex-dependent differences are unknown. Here, we demonstrate that deletion of the ASD-risk gene, Pten, in neocortical pyramidal neurons (NSE Pten KO) results in robust hyperexcitability of local neocortical circuits in female, but not male, mice, observed as prolonged, spontaneous persistent activity states (UP states). Circuit hyperexcitability in NSE Pten KO mice is mediated by enhanced and/or altered signaling of metabotropic glutamate receptor 5 (mGluR5) and estrogen receptor α (ERα) to ERK and protein synthesis selectively in Pten deleted female neurons. In support of this idea, Pten deleted Layer 5 cortical neurons have female-specific increases in mGluR5 and mGluR5-driven protein synthesis. In addition, mGluR5-ERα complexes are elevated in female cortex and genetic reduction of ERα in Pten KO cortical neurons rescues circuit excitability, protein synthesis and enlarged neurons selectively in females. Abnormal timing and hyperexcitability of neocortical circuits in female NSE Pten KO mice are associated with deficits in temporal processing of sensory stimuli and social behaviors as well as mGluR5-dependent seizures. Female-specific cortical hyperexcitability and mGluR5-dependent seizures are also observed in a human disease relevant mouse model, germline Pten +/- mice. Our results reveal molecular mechanisms by which sex and a high impact ASD-risk gene interact to affect brain function and behavior.
Collapse
|
6
|
Ahmadian Elmi M, Motamed N, Picard D. Proteomic Analyses of the G Protein-Coupled Estrogen Receptor GPER1 Reveal Constitutive Links to Endoplasmic Reticulum, Glycosylation, Trafficking, and Calcium Signaling. Cells 2023; 12:2571. [PMID: 37947649 PMCID: PMC10650109 DOI: 10.3390/cells12212571] [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: 08/25/2023] [Revised: 10/14/2023] [Accepted: 11/01/2023] [Indexed: 11/12/2023] Open
Abstract
The G protein-coupled estrogen receptor 1 (GPER1) has been proposed to mediate rapid responses to the steroid hormone estrogen. However, despite a strong interest in its potential role in cancer, whether it is indeed activated by estrogen and how this works remain controversial. To provide new tools to address these questions, we set out to determine the interactome of exogenously expressed GPER1. The combination of two orthogonal methods, namely APEX2-mediated proximity labeling and immunoprecipitation followed by mass spectrometry, gave us high-confidence results for 73 novel potential GPER1 interactors. We found that this GPER1 interactome is not affected by estrogen, a result that mirrors the constitutive activity of GPER1 in a functional assay with a Rac1 sensor. We specifically validated several hits highlighted by a gene ontology analysis. We demonstrate that CLPTM1 interacts with GPER1 and that PRKCSH and GANAB, the regulatory and catalytic subunits of α-glucosidase II, respectively, associate with CLPTM1 and potentially indirectly with GPER1. An imbalance in CLPTM1 levels induces nuclear association of GPER1, as does the overexpression of PRKCSH. Moreover, we show that the Ca2+ sensor STIM1 interacts with GPER1 and that upon STIM1 overexpression and depletion of Ca2+ stores, GPER1 becomes more nuclear. Thus, these new GPER1 interactors establish interesting connections with membrane protein maturation, trafficking, and calcium signaling.
Collapse
Affiliation(s)
- Maryam Ahmadian Elmi
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran 14155-6455, Iran
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH-1211 Genève, Switzerland
| | - Nasrin Motamed
- Department of Cellular and Molecular Biology, School of Biology, College of Science, University of Tehran, Tehran 14155-6455, Iran
| | - Didier Picard
- Département de Biologie Moléculaire et Cellulaire, Université de Genève, Sciences III, Quai Ernest-Ansermet 30, CH-1211 Genève, Switzerland
| |
Collapse
|
7
|
Yadav P, Podia M, Kumari SP, Mani I. Glutamate receptor endocytosis and signaling in neurological conditions. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 196:167-207. [PMID: 36813358 DOI: 10.1016/bs.pmbts.2022.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The non-essential amino acid glutamate acts as a major excitatory neurotransmitter and plays a significant role in the central nervous system (CNS). It binds with two different types of receptors, ionotropic glutamate receptors (iGluRs) and metabotropic glutamate receptors (mGluRs), responsible for the postsynaptic excitation of neurons. They are important for memory, neural development and communication, and learning. Endocytosis and subcellular trafficking of the receptor are essential for the regulation of receptor expression on the cell membrane and excitation of the cells. The endocytosis and trafficking of the receptor are dependent on its type, ligand, agonist, and antagonist present. This chapter discusses the types of glutamate receptors, their subtypes, and the regulation of their internalization and trafficking. The roles of glutamate receptors in neurological diseases are also briefly discussed.
Collapse
Affiliation(s)
- Prerna Yadav
- Department of Microbiology, University of Delhi, New Delhi, India
| | - Mansi Podia
- Department of Microbiology, University of Delhi, New Delhi, India
| | - Shashi Prabha Kumari
- Department of Microbiology, Ram Lal Anand College, University of Delhi, New Delhi, India
| | - Indra Mani
- Department of Microbiology, Gargi College, University of Delhi, New Delhi, India.
| |
Collapse
|
8
|
Jong YI, Harmon SK, O'Malley KL. GPCR
Signaling from Intracellular Membranes. GPCRS AS THERAPEUTIC TARGETS 2022:216-298. [DOI: 10.1002/9781119564782.ch8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2025]
|
9
|
Yang HM, Hou TZ, Zhang YN, Zhao SD, Wu YL, Zhang H. Blocked metabotropic glutamate receptor 5 enhances chemosensitivity in hepatocellular carcinoma and attenuates chemotoxicity in the normal liver by regulating DNA damage. Cancer Gene Ther 2022; 29:1487-1501. [PMID: 35396501 DOI: 10.1038/s41417-022-00465-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 03/08/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
Abstract
DNA damaging agents are used as chemotherapeutics in many cancers, including hepatocellular carcinoma (HCC). However, they are associated with problems such as low sensitivity to chemotherapy and the induction of liver injury, underscoring the need to identify new therapies. Here, we investigated the differential regulatory effect of metabotropic glutamate receptor 5 (mGlu5) on chemosensitivity in HCC and chemotoxicity to the normal liver. The expression of mGlu5 was higher in HCC than in the normal liver, and correlated with poor prognosis according to The Cancer Genome Atlas database and Integrative Molecular Database of Hepatocellular Carcinoma. Cisplatin, oxaliplatin or methyl methanesulfonate (MMS) caused cell death by decreasing mGlu5 expression in HCC cells and increased mGlu5 expression in hepatic cells. In HCC cells, inhibition of mGlu5 aggravated MMS-induced DNA damage by increasing intracellular Ca2+ overload and mitogen-activated protein kinase (MAPK) activation, thereby promoting cell death, and activation of mGlu5 rescued the effect of MMS. However, in hepatic cells, mGlu5 inhibition alleviated MMS-induced DNA damage by downregulating Ca2+-derived MAPK pathways to advance hepatic cell survival. The opposite effects of mGlu5 overexpression or knockdown on MMS-induced DNA damage supported that cell death is a result of the differential regulation of mGlu5 expression. Inhibition of mGlu5 increased chemosensitivity and decreased chemotoxicity in a rat tumor model. This study suggests that mGlu5 inhibition could act synergistically with HCC chemotherapeutics with minimal side effects, which may improve the treatment of patients with HCC in the future.
Collapse
Affiliation(s)
- Hui-Min Yang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Tian-Zhong Hou
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Ya-Nan Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
| | - Shu-Dong Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, 100083, Beijing, China
| | - Yong-Le Wu
- Center of Hepatic and Digestive Disease, Beijing YouAn Hospital, Capital Medical University, 100069, Beijing, China
| | - Hong Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Beijing Key Laboratory of Neural Regeneration and Repair, Beijing Institute for Brain Disorders, Capital Medical University, 100069, Beijing, China.
| |
Collapse
|
10
|
Fasciani I, Carli M, Petragnano F, Colaianni F, Aloisi G, Maggio R, Scarselli M, Rossi M. GPCRs in Intracellular Compartments: New Targets for Drug Discovery. Biomolecules 2022; 12:1343. [PMID: 36291552 PMCID: PMC9599219 DOI: 10.3390/biom12101343] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 08/02/2023] Open
Abstract
The architecture of eukaryotic cells is defined by extensive membrane-delimited compartments, which entails separate metabolic processes that would otherwise interfere with each other, leading to functional differences between cells. G protein-coupled receptors (GPCRs) are the largest class of cell surface receptors, and their signal transduction is traditionally viewed as a chain of events initiated from the plasma membrane. Furthermore, their intracellular trafficking, internalization, and recycling were considered only to regulate receptor desensitization and cell surface expression. On the contrary, accumulating data strongly suggest that GPCRs also signal from intracellular compartments. GPCRs localize in the membranes of endosomes, nucleus, Golgi and endoplasmic reticulum apparatuses, mitochondria, and cell division compartments. Importantly, from these sites they have shown to orchestrate multiple signals that regulate different cell pathways. In this review, we summarize the current knowledge of this fascinating phenomenon, explaining how GPCRs reach the intracellular sites, are stimulated by the endogenous ligands, and their potential physiological/pathophysiological roles. Finally, we illustrate several mechanisms involved in the modulation of the compartmentalized GPCR signaling by drugs and endogenous ligands. Understanding how GPCR signaling compartmentalization is regulated will provide a unique opportunity to develop novel pharmaceutical approaches to target GPCRs and potentially lead the way towards new therapeutic approaches.
Collapse
Affiliation(s)
- Irene Fasciani
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Marco Carli
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Francesco Petragnano
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Francesco Colaianni
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Gabriella Aloisi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Roberto Maggio
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Marco Scarselli
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Mario Rossi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
11
|
Group I Metabotropic Glutamate Receptors and Interacting Partners: An Update. Int J Mol Sci 2022; 23:ijms23020840. [PMID: 35055030 PMCID: PMC8778124 DOI: 10.3390/ijms23020840] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/09/2022] [Accepted: 01/10/2022] [Indexed: 12/21/2022] Open
Abstract
Group I metabotropic glutamate (mGlu) receptors (mGlu1/5 subtypes) are G protein-coupled receptors and are broadly expressed in the mammalian brain. These receptors play key roles in the modulation of normal glutamatergic transmission and synaptic plasticity, and abnormal mGlu1/5 signaling is linked to the pathogenesis and symptomatology of various mental and neurological disorders. Group I mGlu receptors are noticeably regulated via a mechanism involving dynamic protein-protein interactions. Several synaptic protein kinases were recently found to directly bind to the intracellular domains of mGlu1/5 receptors and phosphorylate the receptors at distinct amino acid residues. A variety of scaffolding and adaptor proteins also interact with mGlu1/5. Constitutive or activity-dependent interactions between mGlu1/5 and their interacting partners modulate trafficking, anchoring, and expression of the receptors. The mGlu1/5-associated proteins also finetune the efficacy of mGlu1/5 postreceptor signaling and mGlu1/5-mediated synaptic plasticity. This review analyzes the data from recent studies and provides an update on the biochemical and physiological properties of a set of proteins or molecules that interact with and thus regulate mGlu1/5 receptors.
Collapse
|
12
|
McMahon DB, Kuek LE, Johnson ME, Johnson PO, Horn RL, Carey RM, Adappa ND, Palmer JN, Lee RJ. The bitter end: T2R bitter receptor agonists elevate nuclear calcium and induce apoptosis in non-ciliated airway epithelial cells. Cell Calcium 2022; 101:102499. [PMID: 34839223 PMCID: PMC8752513 DOI: 10.1016/j.ceca.2021.102499] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/21/2021] [Accepted: 10/31/2021] [Indexed: 01/03/2023]
Abstract
Bitter taste receptors (T2Rs) localize to airway motile cilia and initiate innate immune responses in retaliation to bacterial quorum sensing molecules. Activation of cilia T2Rs leads to calcium-driven NO production that increases cilia beating and directly kills bacteria. Several diseases, including chronic rhinosinusitis, COPD, and cystic fibrosis, are characterized by loss of motile cilia and/or squamous metaplasia. To understand T2R function within the altered landscape of airway disease, we studied T2Rs in non-ciliated airway cell lines and primary cells. Several T2Rs localize to the nucleus in de-differentiated cells that typically localize to cilia in differentiated cells. As cilia and nuclear import utilize shared proteins, some T2Rs may target to the nucleus in the absence of motile cilia. T2R agonists selectively elevated nuclear and mitochondrial calcium through a G-protein-coupled receptor phospholipase C mechanism. Additionally, T2R agonists decreased nuclear cAMP, increased nitric oxide, and increased cGMP, consistent with T2R signaling. Furthermore, exposure to T2R agonists led to nuclear calcium-induced mitochondrial depolarization and caspase activation. T2R agonists induced apoptosis in primary bronchial and nasal cells differentiated at air-liquid interface but then induced to a squamous phenotype by apical submersion. Air-exposed well-differentiated cells did not die. This may be a last-resort defense against bacterial infection. However, it may also increase susceptibility of de-differentiated or remodeled epithelia to damage by bacterial metabolites. Moreover, the T2R-activated apoptosis pathway occurs in airway cancer cells. T2Rs may thus contribute to microbiome-tumor cell crosstalk in airway cancers. Targeting T2Rs may be useful for activating cancer cell apoptosis while sparing surrounding tissue.
Collapse
Affiliation(s)
- Derek B. McMahon
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,Correspondence: Derek B. McMahon, PhD or Robert J. Lee, PhD, Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA, 215-573-9766, (D.B.M.) or (R.J.L)
| | - Li Eon Kuek
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Madeline E. Johnson
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Paige O. Johnson
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Rachel L.J. Horn
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Ryan M. Carey
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Nithin D. Adappa
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - James N. Palmer
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Robert J. Lee
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA,Correspondence: Derek B. McMahon, PhD or Robert J. Lee, PhD, Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA USA, 215-573-9766, (D.B.M.) or (R.J.L)
| |
Collapse
|
13
|
McCullock TW, Kammermeier PJ. The evidence for and consequences of metabotropic glutamate receptor heterodimerization. Neuropharmacology 2021; 199:108801. [PMID: 34547332 DOI: 10.1016/j.neuropharm.2021.108801] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 09/07/2021] [Accepted: 09/17/2021] [Indexed: 12/15/2022]
Abstract
Metabotropic glutamate receptors (mGluRs) are an essential component of the mammalian central nervous system. These receptors modulate neuronal excitability in response to extracellular glutamate through the activation of intracellular heterotrimeric G proteins. Like most other class C G protein-coupled receptors, mGluRs function as obligate dimer proteins, meaning they need to form dimer complexes before becoming functional receptors. All mGluRs possess the ability to homodimerize, but studies over the past ten years have demonstrated these receptors are also capable of forming heterodimers in specific patterns. These mGluR heterodimers appear to have their own unique biophysical behavior and pharmacology with both native and synthetic compounds with few rules having been identified that allow for prediction of the consequences of any particular mGluR pair forming heterodimers. Here, we review the relevant literature demonstrating the existence and consequences of mGluR heterodimerization. By collecting biophysical and pharmacological data of several mGluR heterodimers we demonstrate the lack of generalizable behavior of these complexes indicating that each individual dimeric pair needs to be investigated independently. Additionally, by combining sequence alignment and structural analysis, we propose that interactions between the β4-A Helix Loop and the D Helix in the extracellular domain of these receptors are the structural components that dictate heterodimerization compatibility. Finally, we discuss the potential implications of mGluR heterodimerization from the viewpoints of further developing our understanding of neuronal physiology and leveraging mGluRs as a therapeutic target for the treatment of pathophysiology.
Collapse
Affiliation(s)
- Tyler W McCullock
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave, Box 711, Rochester, NY, 14642, USA.
| | - Paul J Kammermeier
- Department of Pharmacology and Physiology, University of Rochester Medical Center, 601 Elmwood Ave, Box 711, Rochester, NY, 14642, USA.
| |
Collapse
|
14
|
Hámor PU, Schwendt M. Metabotropic Glutamate Receptor Trafficking and its Role in Drug-Induced Neurobehavioral Plasticity. Brain Plast 2021; 7:61-76. [PMID: 34868874 PMCID: PMC8609495 DOI: 10.3233/bpl-210120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/06/2021] [Indexed: 12/18/2022] Open
Abstract
Glutamate is the major excitatory neurotransmitter in the mammalian central nervous system that guides developmental and experience-dependent changes in many cellular substrates and brain circuits, through the process collectively referred to as neurobehavioral plasticity. Regulation of cell surface expression and membrane trafficking of glutamate receptors represents an important mechanism that assures optimal excitatory transmission, and at the same time, also allows for fine-tuning neuronal responses to glutamate. On the other hand, there is growing evidence implicating dysregulated glutamate receptor trafficking in the pathophysiology of several neuropsychiatric disorders. This review provides up-to-date information on the molecular determinants regulating trafficking and surface expression of metabotropic glutamate (mGlu) receptors in the rodent and human brain and discusses the role of mGluR trafficking in maladaptive synaptic plasticity produced by addictive drugs. As substantial evidence links glutamatergic dysfunction to the progression and the severity of drug addiction, advances in our understanding of mGluR trafficking may provide opportunities for the development of novel pharmacotherapies of addiction and other neuropsychiatric disorders.
Collapse
Affiliation(s)
- Peter U. Hámor
- Department of Psychology, University of Florida, Gainesville, FL, USA
- Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| | - Marek Schwendt
- Department of Psychology, University of Florida, Gainesville, FL, USA
- Center for Addiction Research and Education, University of Florida, Gainesville, FL, USA
| |
Collapse
|
15
|
Rebeillard F, De Gois S, Pietrancosta N, Mai TH, Lai-Kuen R, Kieffer BL, Giros B, Massart R, Darmon M, Diaz J. The Orphan GPCR Receptor, GPR88, Interacts with Nuclear Protein Partners in the Cerebral Cortex. Cereb Cortex 2021; 32:479-489. [PMID: 34247243 DOI: 10.1093/cercor/bhab224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
GPR88 is an orphan G-protein-coupled receptor (GPCR) highly expressed in striatal medium spiny neurons (MSN), also found in cortical neurons at low level. In MSN, GPR88 has a canonical GPCR plasma membrane/cytoplasmic expression, whereas in cortical neurons, we previously reported an atypical intranuclear localization. Molecular size analysis suggests that GPR88, expressed in plasma membrane of MSN or in nuclear compartment of cortical neurons, corresponds to the full-length protein. By transfection of cortical neurons, we showed that GPR88 fluorescent chimeras exhibit a nuclear localization. This localization is contingent on the third intracytoplasmic loop and C-terminus domains, even though these domains do not contain any known nuclear localization signals (NLS). Using yeast two-hybrid screening with these domains, we identified the nuclear proteins ATRX, TOP2B, and BAZ2B, all involved in chromatin remodeling, as potential protein partners of GPR88. We also validated the interaction of GPR88 with these nuclear proteins by proximity ligation assay on cortical neurons in culture and coimmunoprecipitation experiments on cortical extracts from GPR88 wild-type (WT) and knockout (KO) mice. The identification of GPR88 subcellular partners may provide novel functional insights for nonclassical modes of GPCR action that could be relevant in the maturating process of neocortical neurons.
Collapse
Affiliation(s)
- Florian Rebeillard
- Cellular Biology and Molecular Pharmacology of Central Receptors, Institut de Psychiatrie et de Neurosciences de Paris, Inserm U1266, Paris 75014, France.,Université de Paris, Sorbonne Paris Cité, Paris 75005, France
| | | | - Nicolas Pietrancosta
- Laboratoire des Biomolécules, LBM, Département de chimie, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, Paris 75005, France.,Neuroscience Paris Seine-Institut de Biologie Paris Seine (NPS-IBPS) INSERM, CNRS, Sorbonne Université, Paris 75005, France
| | - Thi Hue Mai
- Cellular Biology and Molecular Pharmacology of Central Receptors, Institut de Psychiatrie et de Neurosciences de Paris, Inserm U1266, Paris 75014, France
| | - René Lai-Kuen
- Cellular and Molecular Imaging Facility, US25 Inserm-3612 CNRS, Faculté de Pharmacie de Paris, Université de Paris, Paris, France
| | | | - Bruno Giros
- Université de Paris, INCC UMR 8002, CNRS, Paris F-75006, France.,Department of Psychiatry, Douglas Hospital, Mc Gill University, Montreal, Quebec H4H 1R3, Canada
| | - Renaud Massart
- Inserm U955 Interventional NeuroPsychology Team, Ecole Normale Supérieure, Paris 75005, France
| | - Michèle Darmon
- Cellular Biology and Molecular Pharmacology of Central Receptors, Institut de Psychiatrie et de Neurosciences de Paris, Inserm U1266, Paris 75014, France
| | - Jorge Diaz
- Cellular Biology and Molecular Pharmacology of Central Receptors, Institut de Psychiatrie et de Neurosciences de Paris, Inserm U1266, Paris 75014, France.,Université de Paris, INCC UMR 8002, CNRS, Paris F-75006, France
| |
Collapse
|
16
|
Gonçalves-Monteiro S, Ribeiro-Oliveira R, Vieira-Rocha MS, Vojtek M, Sousa JB, Diniz C. Insights into Nuclear G-Protein-Coupled Receptors as Therapeutic Targets in Non-Communicable Diseases. Pharmaceuticals (Basel) 2021; 14:439. [PMID: 34066915 PMCID: PMC8148550 DOI: 10.3390/ph14050439] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) comprise a large protein superfamily divided into six classes, rhodopsin-like (A), secretin receptor family (B), metabotropic glutamate (C), fungal mating pheromone receptors (D), cyclic AMP receptors (E) and frizzled (F). Until recently, GPCRs signaling was thought to emanate exclusively from the plasma membrane as a response to extracellular stimuli but several studies have challenged this view demonstrating that GPCRs can be present in intracellular localizations, including in the nuclei. A renewed interest in GPCR receptors' superfamily emerged and intensive research occurred over recent decades, particularly regarding class A GPCRs, but some class B and C have also been explored. Nuclear GPCRs proved to be functional and capable of triggering identical and/or distinct signaling pathways associated with their counterparts on the cell surface bringing new insights into the relevance of nuclear GPCRs and highlighting the nucleus as an autonomous signaling organelle (triggered by GPCRs). Nuclear GPCRs are involved in physiological (namely cell proliferation, transcription, angiogenesis and survival) and disease processes (cancer, cardiovascular diseases, etc.). In this review we summarize emerging evidence on nuclear GPCRs expression/function (with some nuclear GPCRs evidencing atypical/disruptive signaling pathways) in non-communicable disease, thus, bringing nuclear GPCRs as targets to the forefront of debate.
Collapse
Affiliation(s)
- Salomé Gonçalves-Monteiro
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Rita Ribeiro-Oliveira
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Maria Sofia Vieira-Rocha
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Martin Vojtek
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Joana B. Sousa
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Carmen Diniz
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
17
|
Fatima A, Hoeber J, Schuster J, Koshimizu E, Maya-Gonzalez C, Keren B, Mignot C, Akram T, Ali Z, Miyatake S, Tanigawa J, Koike T, Kato M, Murakami Y, Abdullah U, Ali MA, Fadoul R, Laan L, Castillejo-López C, Liik M, Jin Z, Birnir B, Matsumoto N, Baig SM, Klar J, Dahl N. Monoallelic and bi-allelic variants in NCDN cause neurodevelopmental delay, intellectual disability, and epilepsy. Am J Hum Genet 2021; 108:739-748. [PMID: 33711248 PMCID: PMC8059333 DOI: 10.1016/j.ajhg.2021.02.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 02/21/2021] [Indexed: 12/14/2022] Open
Abstract
Neurochondrin (NCDN) is a cytoplasmatic neural protein of importance for neural growth, glutamate receptor (mGluR) signaling, and synaptic plasticity. Conditional loss of Ncdn in mice neural tissue causes depressive-like behaviors, impaired spatial learning, and epileptic seizures. We report on NCDN missense variants in six affected individuals with variable degrees of developmental delay, intellectual disability (ID), and seizures. Three siblings were found homozygous for a NCDN missense variant, whereas another three unrelated individuals carried different de novo missense variants in NCDN. We assayed the missense variants for their capability to rescue impaired neurite formation in human neuroblastoma (SH-SY5Y) cells depleted of NCDN. Overexpression of wild-type NCDN rescued the neurite-phenotype in contrast to expression of NCDN containing the variants of affected individuals. Two missense variants, associated with severe neurodevelopmental features and epilepsy, were unable to restore mGluR5-induced ERK phosphorylation. Electrophysiological analysis of SH-SY5Y cells depleted of NCDN exhibited altered membrane potential and impaired action potentials at repolarization, suggesting NCDN to be required for normal biophysical properties. Using available transcriptome data from human fetal cortex, we show that NCDN is highly expressed in maturing excitatory neurons. In combination, our data provide evidence that bi-allelic and de novo variants in NCDN cause a clinically variable form of neurodevelopmental delay and epilepsy, highlighting a critical role for NCDN in human brain development.
Collapse
|
18
|
Crilly SE, Puthenveedu MA. Compartmentalized GPCR Signaling from Intracellular Membranes. J Membr Biol 2020; 254:259-271. [PMID: 33231722 DOI: 10.1007/s00232-020-00158-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/11/2020] [Indexed: 12/21/2022]
Abstract
G protein-coupled receptors (GPCRs) are integral membrane proteins that transduce a wide array of inputs including light, ions, hormones, and neurotransmitters into intracellular signaling responses which underlie complex processes ranging from vision to learning and memory. Although traditionally thought to signal primarily from the cell surface, GPCRs are increasingly being recognized as capable of signaling from intracellular membrane compartments, including endosomes, the Golgi apparatus, and nuclear membranes. Remarkably, GPCR signaling from these membranes produces functional effects that are distinct from signaling from the plasma membrane, even though often the same G protein effectors and second messengers are activated. In this review, we will discuss the emerging idea of a "spatial bias" in signaling. We will present the evidence for GPCR signaling through G protein effectors from intracellular membranes, and the ways in which this signaling differs from canonical plasma membrane signaling with important implications for physiology and pharmacology. We also highlight the potential mechanisms underlying spatial bias of GPCR signaling, including how intracellular membranes and their associated lipids and proteins affect GPCR activity and signaling.
Collapse
Affiliation(s)
- Stephanie E Crilly
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Manojkumar A Puthenveedu
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| |
Collapse
|
19
|
Jones AJY, Gabriel F, Tandale A, Nietlispach D. Structure and Dynamics of GPCRs in Lipid Membranes: Physical Principles and Experimental Approaches. Molecules 2020; 25:E4729. [PMID: 33076366 PMCID: PMC7587580 DOI: 10.3390/molecules25204729] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 02/06/2023] Open
Abstract
Over the past decade, the vast amount of information generated through structural and biophysical studies of GPCRs has provided unprecedented mechanistic insight into the complex signalling behaviour of these receptors. With this recent information surge, it has also become increasingly apparent that in order to reproduce the various effects that lipids and membranes exert on the biological function for these allosteric receptors, in vitro studies of GPCRs need to be conducted under conditions that adequately approximate the native lipid bilayer environment. In the first part of this review, we assess some of the more general effects that a membrane environment exerts on lipid bilayer-embedded proteins such as GPCRs. This is then followed by the consideration of more specific effects, including stoichiometric interactions with specific lipid subtypes. In the final section, we survey a range of different membrane mimetics that are currently used for in vitro studies, with a focus on NMR applications.
Collapse
Affiliation(s)
| | | | | | - Daniel Nietlispach
- Department of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, UK; (A.J.Y.J.); (F.G.); (A.T.)
| |
Collapse
|
20
|
Perl S, Richter F, Richter A. Striatal and cortical metabotropic glutamate 5 receptor expression and behavioral effects of the positive allosteric modulator CDPPB in a model of DYT1 dystonia. Pharmacol Biochem Behav 2020; 196:172977. [PMID: 32615137 DOI: 10.1016/j.pbb.2020.172977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 06/09/2020] [Accepted: 06/23/2020] [Indexed: 11/19/2022]
Abstract
The metabotropic glutamate 5 (mGlu5) receptor is critically involved in corticostriatal plasticity which is disturbed in various animal models of dystonia. Recently, the positive allosteric modulator 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) exerted prodyskinetic effects in a phenotypic model of episodic dystonia. In the DYT1 knock-in (KI) mouse, a model for a persistent type of dystonia, previous ex vivo electrophysiological experiments indicated that mGlu5 receptors are involved in abnormal striatal plasticity. Therefore, in the present study we examined the mGlu5 receptor expression in the striatum and cortex of DYT1 KI mice in comparison with wildtype littermates. By immunohistochemistry (IHC) we found a lower expression of mGlu5 receptors in the cortex (16%) and ventral striatum (10%) but not in the whole striatum of DYT1 KI mice, while mRNA levels were merely lower in the striatum of DYT1 KI mice (43%). However, mGlu5 receptor protein levels measured by western blotting showed no significant differences in tissue of the whole striatum and in the cortex between both genotypes. Since DYT1 KI mice do not exhibit dystonic symptoms, we investigated if CDPPB provokes dystonia or dyskinesia. CDPPB (10, 20 and 30 mg/kg intraperitoneal, i.p.) did not induce abnormal movements and the locomotor activity did not differ between DYT1 KI and wildtype mice. The present data do not provide evidence for a crucial role of the mGlu5 receptor in the pathophysiology of DYT1 dystonia, but corticostriatal changes are in line with the hypothesis of maladaptive plasticity in dystonia.
Collapse
Affiliation(s)
- Stefanie Perl
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany.
| | - Franziska Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany; Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, 30559 Hannover, Germany.
| | - Angelika Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103 Leipzig, Germany.
| |
Collapse
|
21
|
Jong YJI, Harmon SK, O’Malley KL. Location and Cell-Type-Specific Bias of Metabotropic Glutamate Receptor, mGlu 5, Negative Allosteric Modulators. ACS Chem Neurosci 2019; 10:4558-4570. [PMID: 31609579 DOI: 10.1021/acschemneuro.9b00415] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Emerging data indicate that G-protein coupled receptor (GPCR) signaling is determined by not only the agonist and a given receptor but also a variety of cell-type-specific factors that can influence a receptor's response. For example, the metabotropic glutamate receptor, mGlu5, which is implicated in a number of neuropsychiatric disorders such as depression, anxiety, and autism, also signals from inside the cell which leads to sustained Ca2+ mobilization versus rapid transient responses. Because mGlu5 is an important drug target, many negative allosteric modulators (NAMs) have been generated to modulate its activity. Here we show that NAMs such as AFQ056, AZD2066, and RG7090 elicit very different end points when tested in postnatal neuronal cultures expressing endogenous mGlu5 receptors. For example, AFQ056 fails to block intracellular mGlu5-mediated Ca2+ increases whereas RG7090 is very effective. These differences are not due to differential receptor levels, since about the same number of mGlu5 receptors are present on neurons from the cortex, hippocampus, and striatum based on pharmacological, biochemical, and molecular data. Moreover, biotinylation studies reveal that more than 90% of the receptor is intracellular in these neurons. Taken together, these data indicate that the tested NAMs exhibit both location-dependent and cell type specific bias for mGlu5-mediated Ca2+ mobilization which may affect clinical outcomes.
Collapse
Affiliation(s)
- Yuh-Jiin Ivy Jong
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| | - Steven K. Harmon
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| | - Karen L. O’Malley
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, Missouri 63110, United States
| |
Collapse
|
22
|
Ribeiro-Oliveira R, Vojtek M, Gonçalves-Monteiro S, Vieira-Rocha MS, Sousa JB, Gonçalves J, Diniz C. Nuclear G-protein-coupled receptors as putative novel pharmacological targets. Drug Discov Today 2019; 24:2192-2201. [DOI: 10.1016/j.drudis.2019.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/15/2019] [Accepted: 09/05/2019] [Indexed: 12/14/2022]
|
23
|
Perl S, Richter F, Gericke B, Richter A. Expression of metabotropic glutamate 5 receptors in the striatum and cortex and effects of modulators on the severity of dystonia in the phenotypic dt sz model. Eur J Pharmacol 2019; 859:172527. [PMID: 31283933 DOI: 10.1016/j.ejphar.2019.172527] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 07/01/2019] [Accepted: 07/04/2019] [Indexed: 10/26/2022]
Abstract
The metabotropic glutamate 5 (mGlu5) receptor has been suggested as therapeutic target for L-Dopa-induced dyskinesia which is often associated with dystonic symptoms. Therefore, we investigated the acute effects of the non-competitive mGlu5 receptor antagonist fenobam as well as the positive modulator 3-cyano-N-(1,3-diphenyl-1H-pyrazol-5-yl)benzamide (CDPPB) on the severity of inherited dystonia in the mutant dtsz hamster, a phenotypic model with age-dependent episodes of dystonia. Fenobam did not exert significant antidystonic effects (20-50 mg/kg intraperinoneal, i.p.). CDPPB (10, 20 mg/kg i.p.) which was expected to worsen dystonia also failed to show any effects on the severity of dystonia. Interestingly, CDPPB caused axial dyskinesia in addition to the dystonic symptoms in mutant hamsters. This adverse effect could not be observed in non-dystonic control hamsters, indicating possible changes in the expression of mGlu5 receptors in dystonic hamsters. The mGlu5 receptor mRNA did not differ between the dtsz mutant and control hamsters, while immunohistochemical studies indicated that the mGlu5 receptor expression was about 35% higher in striatum and cortex of mutant hamsters at the age of high dystonia severity scores, notably not after spontaneous remission of dystonia, compared to age-matched controls. This difference in mGlu5 receptor protein may be due to altered protein conformation instead of protein level, as western blots revealed similar amounts of monomeric and dimeric protein in mutant hamsters versus control. Thus, the present data do not provide clear evidence for an important role of the mGlu5 receptor in the pathophysiology and as a therapeutic target for types of inherited dystonia.
Collapse
Affiliation(s)
- Stefanie Perl
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany
| | - Franziska Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany; Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, D-30559, Hannover, Germany
| | - Birthe Gericke
- Department of Pharmacology, Toxicology, and Pharmacy, University of Veterinary Medicine Hannover, D-30559, Hannover, Germany
| | - Angelika Richter
- Institute of Pharmacology, Pharmacy and Toxicology, Faculty of Veterinary Medicine, University of Leipzig, 04103, Leipzig, Germany.
| |
Collapse
|
24
|
Villanueva-Romero R, Gutiérrez-Cañas I, Carrión M, González-Álvaro I, Rodríguez-Frade JM, Mellado M, Martínez C, Gomariz RP, Juarranz Y. Activation of Th lymphocytes alters pattern expression and cellular location of VIP receptors in healthy donors and early arthritis patients. Sci Rep 2019; 9:7383. [PMID: 31089161 PMCID: PMC6517580 DOI: 10.1038/s41598-019-43717-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/29/2019] [Indexed: 12/15/2022] Open
Abstract
Vasoactive Intestinal Peptide (VIP) is an important immunomodulator of CD4+ cells in normal and pathological conditions, which exerts its anti-inflammatory and immunomodulatory actions through VPAC receptors, VPAC1 and VPAC2. Only a decrease in the expression of VPAC1 mRNA on Th cells upon activation has been reported. Thus, the deepening in the knowledge of the behavior of these receptors may contribute to the design of new therapies based on their activation and/or blockade. In this study, we describe the expression pattern, cellular location and functional role of VIP receptors during the activation of human Th cells in healthy conditions and in early arthritis (EA). The protein expression pattern of VPAC1 did not change with the activation of Th lymphocytes, whereas VPAC2 was up-regulated. In resting cells, VPAC1 was located on the plasma membrane and nucleus, whereas it only appeared in the nucleus in activated cells. VPAC2 was always found in plasma membrane location. VIP receptors signaled through a PKA-dependent pathway in both conditions, and also by a PKA-independent pathway in activated cells. Both receptors exhibit a potent immunomodulatory capacity by controlling the pathogenic profile and the activation markers of Th cells. These results highlight a novel translational view in inflammatory/autoimmune diseases.
Collapse
Affiliation(s)
- R Villanueva-Romero
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - I Gutiérrez-Cañas
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - M Carrión
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - I González-Álvaro
- Servicio de Reumatología, Instituto de Investigación Sanitaria Hospital La Princesa (IIS-IP), Madrid, Spain
| | - J M Rodríguez-Frade
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - M Mellado
- Departamento de Inmunología y Oncología, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - C Martínez
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - R P Gomariz
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain
| | - Y Juarranz
- Departamento de Biología Celular, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), Madrid, Spain.
| |
Collapse
|
25
|
Fitzgerald ML, Mackie K, Pickel VM. Ultrastructural localization of cannabinoid CB1 and mGluR5 receptors in the prefrontal cortex and amygdala. J Comp Neurol 2019; 527:2730-2741. [PMID: 31008528 DOI: 10.1002/cne.24704] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2018] [Revised: 04/01/2019] [Accepted: 04/17/2019] [Indexed: 12/29/2022]
Abstract
Stimulation of the postsynaptic metabotropic glutamate receptor mGluR5 triggers retrograde signaling of endocannabinoids that activate presynaptic cannabinoid CB1 receptors on juxtaposing axon terminals. To better understand the synaptic structure that supports mGluR5 mediation of CB1 activation in the prefrontal cortex (PFC) and basolateral amygdala (BLA), we examined electron microscopic dual immunolabeling of these receptors in the prelimbic PFC (prPFC) and BLA of adult male rats. CB1 immunoreactivity was detected in axon terminals that were typically large, complex, and contained dense-core and clear synaptic vesicles. Of terminals forming discernible synaptic specializations, 95% were symmetric inhibitory-type in the prPFC and 90% were inhibitory in the BLA. CB1-immunoreactive terminals frequently contacted dendrites containing mGluR5 adjacent to unlabeled terminals forming excitatory-type synapses. Because most CB1-containing terminals form inhibitory-type synapses, the unlabeled axon terminals forming asymmetric synapses are the likely source of the mGluR5 ligand glutamate. In the prPFC, serial section analysis revealed that GABAergic CB1-containing axon terminals targeted dendrites adjacent to glutamatergic axon terminals, often near dendritic bifurcations. These observations provide ultrastructural evidence that cortical CB1 receptors are strategically positioned for integration of synaptic signaling in response to stimulation of postsynaptic mGluR5 receptors and facilitation of heterosynaptic communication between multiple neurons.
Collapse
Affiliation(s)
- Megan L Fitzgerald
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, New York
| | - Ken Mackie
- Department of Psychological and Brain Sciences, Indiana University, Bloomington, Indiana
| | - Virginia M Pickel
- Weill Cornell Medicine, Feil Family Brain and Mind Research Institute, New York, New York
| |
Collapse
|
26
|
Weinberg ZY, Crilly SE, Puthenveedu MA. Spatial encoding of GPCR signaling in the nervous system. Curr Opin Cell Biol 2019; 57:83-89. [PMID: 30708280 DOI: 10.1016/j.ceb.2018.12.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 12/14/2018] [Indexed: 02/06/2023]
Abstract
Several GPCRs, including receptors previously thought to signal primarily from the cell surface, have been recently shown to signal from many intracellular compartments. This raises the idea that signaling by any given receptor is spatially encoded in the cell, with distinct sites of signal origin dictating distinct downstream consequences. We will discuss recent developments that address this novel facet of GPCR physiology, focusing on the spatial segregation of signaling from the cell surface, endosomes, and the Golgi by receptors relevant to the nervous system.
Collapse
Affiliation(s)
- Zara Y Weinberg
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Stephanie E Crilly
- Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, United States
| | - Manojkumar A Puthenveedu
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, United States; Program in Cellular and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, United States.
| |
Collapse
|
27
|
Abstract
The trillions of synaptic connections within the human brain are shaped by experience and neuronal activity, both of which underlie synaptic plasticity and ultimately learning and memory. G protein-coupled receptors (GPCRs) play key roles in synaptic plasticity by strengthening or weakening synapses and/or shaping dendritic spines. While most studies of synaptic plasticity have focused on cell surface receptors and their downstream signaling partners, emerging data point to a critical new role for the very same receptors to signal from inside the cell. Intracellular receptors have been localized to the nucleus, endoplasmic reticulum, lysosome, and mitochondria. From these intracellular positions, such receptors may couple to different signaling systems, display unique desensitization patterns, and/or show distinct patterns of subcellular distribution. Intracellular GPCRs can be activated at the cell surface, endocytosed, and transported to an intracellular site or simply activated in situ by de novo ligand synthesis, diffusion of permeable ligands, or active transport of non-permeable ligands. Current findings reinforce the notion that intracellular GPCRs play a dynamic role in synaptic plasticity and learning and memory. As new intracellular GPCR roles are defined, the need to selectively tailor agonists and/or antagonists to both intracellular and cell surface receptors may lead to the development of more effective therapeutic tools.
Collapse
Affiliation(s)
- Yuh-Jiin I. Jong
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Steven K. Harmon
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Karen L. O’Malley
- Department of Neuroscience, Washington University School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
28
|
Dubuc C, Savard M, Bovenzi V, Lessard A, Côté J, Neugebauer W, Geha S, Chemtob S, Gobeil F. Antitumor activity of cell-penetrant kinin B1 receptor antagonists in human triple-negative breast cancer cells. J Cell Physiol 2018; 234:2851-2865. [PMID: 30132865 DOI: 10.1002/jcp.27103] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 06/28/2018] [Indexed: 12/26/2022]
Abstract
High nuclear expression of G protein-coupled receptors, including kinin B1 receptors (B1R), has been observed in several human cancers, but the clinical significance of this is unknown. We put forward the hypothesis that these "nuclearized" kinin B1R contribute to tumorigenicity and can be a new target in anticancer strategies. Our initial immunostaining and ultrastructural electron microscopy analyses demonstrated high B1R expression predominantly located at internal/nuclear compartments in the MDA-MB-231 triple-negative breast cancer (TNBC) cell line as well as in clinical samples of patients with TNBC. On the basis of these findings, in the present study, we evaluated the anticancer therapeutic potential of newly identified, cell-permeable B1R antagonists in MDA-MB-231 cells (ligand-receptor binding/activity assays and LC-MS/MS analyses). We found that these compounds (SSR240612, NG67, and N2000) were more toxic to MDA-MB-231 cells in comparison with low- or non-B1R expressing MCF-10A normal human mammary epithelial cells and COS-1 cells, respectively (clonogenic, MTT proliferative/cytocidal assays, and fluorescence-activated cell-sorting (FACS)-based apoptosis analyses). By comparison, the peptide B1R antagonist R954 unable to cross cell membrane failed to produce anticancer effects. Furthermore, the putative mechanisms underlying the anticancer activities of cell-penetrant B1R antagonists were assessed by analyzing cell cycle regulation and signaling molecules related to cell survival and apoptosis (FACS and western blot). Finally, drug combination experiments showed that cell-penetrant B1R antagonists can cooperate with suboptimal doses of chemotherapeutic agents (doxorubicin and paclitaxel) to promote TNBC death. This study provides evidence on the potential value of internally acting kinin B1R antagonists in averting growth of breast cancer.
Collapse
Affiliation(s)
- Céléna Dubuc
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Martin Savard
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Veronica Bovenzi
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Andrée Lessard
- Department of Psychiatry, Maryland Psychiatric Research Center, University of Maryland School of Medicine, Baltimore, Maryland
| | - Jérôme Côté
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Witold Neugebauer
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| | - Sameh Geha
- Department of Pathology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Sylvain Chemtob
- Department of Pediatrics, Centre Hospitalier Universitaire (CHU) Sainte-Justine Research Center, Montréal, Québec, Canada
| | - Fernand Gobeil
- Department of Pharmacology and Physiology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Québec, Canada
- Institute of Pharmacology, Université de Sherbrooke, Sherbrooke, Québec, Canada
| |
Collapse
|
29
|
Wang W, Qiao Y, Li Z. New Insights into Modes of GPCR Activation. Trends Pharmacol Sci 2018; 39:367-386. [DOI: 10.1016/j.tips.2018.01.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
|
30
|
Sinreih M, Knific T, Thomas P, Frković Grazio S, Rižner TL. Membrane progesterone receptors β and γ have potential as prognostic biomarkers of endometrial cancer. J Steroid Biochem Mol Biol 2018; 178:303-311. [PMID: 29353001 DOI: 10.1016/j.jsbmb.2018.01.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Revised: 01/10/2018] [Accepted: 01/12/2018] [Indexed: 12/17/2022]
Abstract
Endometrial cancer (EC) is one of the most common malignancies in women worldwide. EC is linked to chronic exposure to estrogens that is unopposed by protective effects of progesterone. Progesterone modulates gene expression via classical nuclear receptors, and has rapid effects via the less characterized membrane-bound progesterone receptors (mPRs) of the progestin and adipoQ receptor (PAQR) family. The presence of mPRs in EC has not been investigated to date. The aims of this study were to examine PAQR7, PAQR8 and PAQR5, which encode for mPRα, mPRβ and mPRγ, respectively, for their expression and localization in EC tissue and adjacent control endometrium. Our results reveal decreased expression of PAQR7 and PAQR8, and unaltered expression of PAQR5 in EC versus control tissue. Expression of PAQR5 was decreased in EC with higher FIGO stage versus stage IA. Immunohistochemistry revealed lower levels of mPRα and mPRβ, but higher levels of mPRγ, in EC versus control tissue. There was greater decrease in mPRβ levels in tumors with lymphovascular invasion. The analysis of the expression data associates higher PAQR5 mRNA and mPRβ protein levels with favorable patient prognosis. Immunohistochemistry showed diverse localizations of mPRs in control and cancer endometrium. In control endometrium, mPRα and mPRβ were localized mostly at the cell membranes, while mPRγ was localized in the cytoplasm and/or nucleus. In cancer endometrium, mPRα and mPRβ were detected at the cell membrane or in the cytoplasm, or both, while mPRγ was only localized in the cytoplasm. Taken together, these results imply that mPRs are involved in EC pathogenesis through effects on the development or progression of cancer. The potential role of mPRβ and mPRγ as prognostic biomarkers needs to be further assessed on a larger number of samples.
Collapse
MESH Headings
- Adenocarcinoma, Mucinous/genetics
- Adenocarcinoma, Mucinous/metabolism
- Adenocarcinoma, Mucinous/pathology
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Membrane/metabolism
- Cystadenocarcinoma, Serous/genetics
- Cystadenocarcinoma, Serous/metabolism
- Cystadenocarcinoma, Serous/pathology
- Endometrial Neoplasms/genetics
- Endometrial Neoplasms/metabolism
- Endometrial Neoplasms/pathology
- Female
- Humans
- Middle Aged
- Neoplasm Invasiveness
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/metabolism
- Receptors, Progesterone/genetics
- Receptors, Progesterone/metabolism
Collapse
Affiliation(s)
- Maša Sinreih
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Slovenia
| | - Tamara Knific
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Slovenia
| | - Peter Thomas
- Marine Science Institute, University of Texas at Austin, Austin, USA
| | - Snježana Frković Grazio
- Division of Obstetrics and Gynaecology, Department of Pathology, University Medical Centre, Ljubljana, Slovenia
| | - Tea Lanišnik Rižner
- Institute of Biochemistry, Faculty of Medicine, University of Ljubljana, Slovenia.
| |
Collapse
|
31
|
Suh YH, Chang K, Roche KW. Metabotropic glutamate receptor trafficking. Mol Cell Neurosci 2018; 91:10-24. [PMID: 29604330 DOI: 10.1016/j.mcn.2018.03.014] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 03/07/2018] [Accepted: 03/26/2018] [Indexed: 01/14/2023] Open
Abstract
The metabotropic glutamate receptors (mGlu receptors) are G protein-coupled receptors that bind to the excitatory neurotransmitter glutamate and are important in the modulation of neuronal excitability, synaptic transmission, and plasticity in the central nervous system. Trafficking of mGlu receptors in and out of the synaptic plasma membrane is a fundamental mechanism modulating excitatory synaptic function through regulation of receptor abundance, desensitization, and signaling profiles. In this review, we cover the regulatory mechanisms determining surface expression and endocytosis of mGlu receptors, with particular focus on post-translational modifications and receptor-protein interactions. The literature we review broadens our insight into the precise events defining the expression of functional mGlu receptors at synapses, and will likely contribute to the successful development of novel therapeutic targets for a variety of developmental, neurological, and psychiatric disorders.
Collapse
Affiliation(s)
- Young Ho Suh
- Department of Biomedical Sciences, Neuroscience Research Institute, Seoul National University College of Medicine, Seoul 03080, South Korea.
| | - Kai Chang
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Katherine W Roche
- Receptor Biology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| |
Collapse
|
32
|
Briddon SJ, Kilpatrick LE, Hill SJ. Studying GPCR Pharmacology in Membrane Microdomains: Fluorescence Correlation Spectroscopy Comes of Age. Trends Pharmacol Sci 2017; 39:158-174. [PMID: 29277246 DOI: 10.1016/j.tips.2017.11.004] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2017] [Revised: 11/13/2017] [Accepted: 11/14/2017] [Indexed: 12/17/2022]
Abstract
G protein-coupled receptors (GPCRs) are organised within the cell membrane into highly ordered macromolecular complexes along with other receptors and signalling proteins. Understanding how heterogeneity in these complexes affects the pharmacology and functional response of these receptors is crucial for developing new and more selective ligands. Fluorescence correlation spectroscopy (FCS) and related techniques such as photon counting histogram (PCH) analysis and image-based FCS can be used to interrogate the properties of GPCRs in these membrane microdomains, as well as their interaction with fluorescent ligands. FCS analyses fluorescence fluctuations within a small-defined excitation volume to yield information about their movement, concentration and molecular brightness (aggregation). These techniques can be used on live cells with single-molecule sensitivity and high spatial resolution. Once the preserve of specialist equipment, FCS techniques can now be applied using standard confocal microscopes. This review describes how FCS and related techniques have revealed novel insights into GPCR biology.
Collapse
Affiliation(s)
- Stephen J Briddon
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK; Centre for Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, UK
| | - Laura E Kilpatrick
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK; Centre for Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, UK
| | - Stephen J Hill
- Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK; Centre for Membrane Proteins and Receptors (COMPARE), University of Birmingham and University of Nottingham, The Midlands, UK.
| |
Collapse
|
33
|
Jong YJI, Harmon SK, O'Malley KL. GPCR signalling from within the cell. Br J Pharmacol 2017; 175:4026-4035. [PMID: 28872669 DOI: 10.1111/bph.14023] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 08/08/2017] [Accepted: 08/17/2017] [Indexed: 12/22/2022] Open
Abstract
Traditionally, signal transduction from GPCRs is thought to emanate from the cell surface where receptor interactions with external stimuli can be transformed into a broad range of cellular responses. However, emergent data show that numerous GPCRs are also associated with various intracellular membranes where they may couple to different signalling systems, display unique desensitization patterns and/or exhibit distinct patterns of subcellular distribution. Although many GPCRs can be activated at the cell surface and subsequently endocytosed and transported to a unique intracellular site, other intracellular GPCRs can be activated in situ either via de novo ligand synthesis, diffusion of permeable ligands or active transport of nonpermeable ligands. Current findings reinforce the notion that intracellular GPCRs play a dynamic role in various biological functions including learning and memory, contractility and angiogenesis. As new intracellular GPCR roles are defined, the need to selectively tailor agonists and/or antagonists to both intracellular and cell surface receptors may lead to the development of more effective therapeutic tools. LINKED ARTICLES This article is part of a themed section on Molecular Pharmacology of GPCRs. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v175.21/issuetoc.
Collapse
Affiliation(s)
- Yuh-Jiin I Jong
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Steven K Harmon
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| | - Karen L O'Malley
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
34
|
Bhosle VK, Rivera JC, Chemtob S. New insights into mechanisms of nuclear translocation of G-protein coupled receptors. Small GTPases 2017; 10:254-263. [PMID: 28125336 DOI: 10.1080/21541248.2017.1282402] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The G-protein coupled receptor (GPCR) signaling was long believed to involve activation of receptor exclusively at the cell surface, followed by its binding to heterotrimeric G-proteins and arrestins to trigger various intracellular signaling cascades, and termination of signaling by internalization of the receptor. It is now accepted that many GPCRs continue to signal after internalization in the endosomes. Since the breakthrough discoveries of nuclear binding sites for their ligands in 1980s, several GPCRs have been detected at cell nuclei. But mechanisms of nuclear localization of GPCRs, many of whom contain putative nuclear localization signals, remain poorly understood to date. Nevertheless, it is known that subcellular trafficking of GPCRs is regulated by members of Ras superfamily of small GTPases, most notably by Rab and Arf GTPases. In this commentary, we highlight several recent studies which suggest novel roles of small GTPases, importins and sorting nexin proteins in the nuclear translocation of GPCRs via vesicular transport pathways. Taken together with increasing evidence for in vivo functionality of the nuclear GPCRs, better understanding of their trafficking will provide valuable clues in cell biology.
Collapse
Affiliation(s)
- Vikrant K Bhosle
- a Department of Pharmacology and Therapeutics , McGill University , Montréal , Québec , Canada.,b CHU Sainte-Justine Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,c Maisonneuve-Rosemont Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,e Cell Biology Program , Peter Gilgan Centre for Research and Learning , Toronto , Ontario , Canada
| | - José Carlos Rivera
- b CHU Sainte-Justine Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,c Maisonneuve-Rosemont Hospital Research Centre , University of Montréal , Montréal , Québec , Canada
| | - Sylvain Chemtob
- a Department of Pharmacology and Therapeutics , McGill University , Montréal , Québec , Canada.,b CHU Sainte-Justine Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,c Maisonneuve-Rosemont Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,d Departments of Pediatrics, Ophthalmology and Pharmacology , University of Montréal , Montréal , Québec , Canada
| |
Collapse
|