1
|
Mohammad Nezhady MA, Cagnone G, Bajon E, Chaudhari P, Modaresinejad M, Hardy P, Maggiorani D, Quiniou C, Joyal JS, Beauséjour C, Chemtob S. Unconventional receptor functions and location-biased signaling of the lactate GPCR in the nucleus. Life Sci Alliance 2025; 8:e202503226. [PMID: 39904567 PMCID: PMC11794946 DOI: 10.26508/lsa.202503226] [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: 01/15/2025] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
G-protein-coupled receptors (GPCRs) are virtually involved in every physiological process. However, mechanisms for their ability to regulate a vast array of different processes remain elusive. An unconventional functional modality could at least in part account for such diverse involvements but has yet to be explored. We found HCAR1, a multifunctional lactate GPCR, to localize at the nucleus and therein capable of initiating location-biased signaling notably nuclear-ERK and AKT. We discovered that nuclear HCAR1 (N-HCAR1) is directly involved in regulating diverse processes. Specifically, N-HCAR1 binds to protein complexes that are involved in promoting protein translation, ribosomal biogenesis, and DNA-damage repair. N-HCAR1 also interacts with chromatin remodelers to directly regulate gene expression. We show that N-HCAR1 displays a broader transcriptomic signature than its plasma membrane counterpart. Interestingly, exclusion of HCAR1 from the nucleus has the same effect as its complete cellular depletion on tumor growth and metastasis in vivo. These results reveal noncanonical functions for a cell nucleus-localized GPCR that are distinct from traditional receptor modalities and through which HCAR1 can participate in regulating various cellular processes.
Collapse
Affiliation(s)
- Mohammad Ali Mohammad Nezhady
- https://ror.org/0161xgx34 Program in Molecular Biology, Faculty of Medicine, Université de Montréal, Montreal, Canada
- https://ror.org/01gv74p78 Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Gael Cagnone
- https://ror.org/01gv74p78 Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Emmanuel Bajon
- https://ror.org/01gv74p78 Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Prabhas Chaudhari
- https://ror.org/01gv74p78 Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- Department of Experimental Medicine, McGill University, Montréal, Canada
| | - Monir Modaresinejad
- https://ror.org/01gv74p78 Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- https://ror.org/0161xgx34 Program in Biomedical Sciences, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Pierre Hardy
- https://ror.org/01gv74p78 Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Damien Maggiorani
- https://ror.org/01gv74p78 Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- https://ror.org/0161xgx34 Department of Pharmacology, Université de Montréal, Montreal, Canada
| | - Christiane Quiniou
- https://ror.org/01gv74p78 Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Jean-Sébastien Joyal
- https://ror.org/01gv74p78 Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- https://ror.org/0161xgx34 Department of Pharmacology, Université de Montréal, Montreal, Canada
| | - Christian Beauséjour
- https://ror.org/01gv74p78 Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Sylvain Chemtob
- https://ror.org/0161xgx34 Program in Molecular Biology, Faculty of Medicine, Université de Montréal, Montreal, Canada
- https://ror.org/01gv74p78 Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- https://ror.org/0161xgx34 Department of Pharmacology, Université de Montréal, Montreal, Canada
| |
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
|
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
|
4
|
Badshah M, Ibrahim J, Su N, Whiley P, Whittaker M, Exintaris B. The Effects of Age on Prostatic Responses to Oxytocin and the Effects of Antagonists. Biomedicines 2023; 11:2956. [PMID: 38001957 PMCID: PMC10669827 DOI: 10.3390/biomedicines11112956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/18/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Benign prostatic hyperplasia (BPH) is an age-related enlargement of the prostate with urethral obstruction that predominantly affects the middle-aged and older male population, resulting in disruptive lower urinary tract symptoms (LUTS), thus creating a profound impact on an individual's quality of life. The development of LUTS may be linked to overexpression of oxytocin receptors (OXTR), resulting in increased baseline myogenic tone within the prostate. Thus, it is hypothesised that targeting OXTR using oxytocin receptor antagonists (atosiban, cligosiban, and β-Mercapto-β,β-cyclopentamethylenepropionyl1, O-Me-Tyr2, Orn8]-Oxytocin (ßMßßC)), may attenuate myogenic tone within the prostate. Organ bath and immunohistochemistry techniques were conducted on prostate tissue from young and older rats. Our contractility studies demonstrated that atosiban significantly decreased the frequency of spontaneous contractions within the prostate of young rats (**** p < 0.0001), and cligosiban (* p < 0.05), and ßMßßC (**** p < 0.0001) in older rats. Additionally, immunohistochemistry findings revealed that nuclear-specific OXTR was predominantly expressed within the epithelium of the prostate of both young (*** p < 0.001) and older rats (**** p < 0.0001). In conclusion, our findings indicate that oxytocin is a key modulator of prostate contractility, and targeting OXTR is a promising avenue in the development of novel BPH drugs.
Collapse
Affiliation(s)
- Masroor Badshah
- Hudson Institute of Medical Research, Monash University, Clayton, VIC 3168, Australia;
| | - Jibriil Ibrahim
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC 3052, Australia (N.S.)
| | - Nguok Su
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC 3052, Australia (N.S.)
| | - Penny Whiley
- Hudson Institute of Medical Research, Monash University, Clayton, VIC 3168, Australia;
| | - Michael Whittaker
- Drug, Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Parkville, VIC 3052, Australia;
| | - Betty Exintaris
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Parkville, VIC 3052, Australia (N.S.)
| |
Collapse
|
5
|
Franco R, Serrano-Marín J, Navarro G, Rivas-Santisteban R. The NADPH Link between the Renin Angiotensin System and the Antioxidant Mechanisms in Dopaminergic Neurons. Antioxidants (Basel) 2023; 12:1869. [PMID: 37891948 PMCID: PMC10604245 DOI: 10.3390/antiox12101869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
The renin angiotensin system (RAS) has several components including signaling peptides, enzymes, and membrane receptors. The effort in characterizing this system in the periphery has led to the approval of a class of antihypertensives. Much less is known about RAS in the central nervous system. The production of RAS peptides and the expression of several RAS enzymes and receptors in dopaminergic neurons of the substantia nigra has raised expectations in the therapy of Parkinson's disease, a neurodegenerative condition characterized by lack of dopamine in the striatum, the motor control region of the mammalian brain. On the one hand, dopamine production requires reducing power. On the other hand, reducing power is required by mechanisms involved in REDOX homeostasis. This review focuses on the potential role of RAS in the regulation of neuronal/glial expression of glucose-6-phosphate dehydrogenase, which produces the NADPH required for dopamine synthesis and for reactive oxygen species (ROS) detoxification. It is known that transgenic expression of the gene coding for glucose-6-phosphate dehydrogenase prevents the death of dopaminergic nigral neurons. Signaling via angiotensin II G protein-coupled receptors, AT1 or AT2, leads to the activation of protein kinase A and/or protein kinase C that in turn can regulate glucose-6- phosphate dehydrogenase activity, by Ser/Thr phosphorylation/dephosphorylation events. Long-term effects of AT1 or AT2 receptor activation may also impact on the concentration of the enzyme via activation of transcription factors that participate in the regulation of gene expression in neurons (or glia). Future research is needed to determine how the system can be pharmacologically manipulated to increase the availability of NADPH to neurons degenerating in Parkinson's disease and to neuroprotective glia.
Collapse
Affiliation(s)
- Rafael Franco
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
- CiberNed, Network Center for Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28029 Madrid, Spain;
- School of Chemistry, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Joan Serrano-Marín
- Department of Biochemistry and Molecular Biomedicine, School of Biology, Universitat de Barcelona, 08028 Barcelona, Spain
| | - Gemma Navarro
- CiberNed, Network Center for Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28029 Madrid, Spain;
- Department of Biochemistry and Physiology, School of Pharmacy and Food Science, Universitat de Barcelona, 08028 Barcelona, Spain
- Institute of Neurosciences, Universitat de Barcelona, 08007 Barcelona, Spain
| | - Rafael Rivas-Santisteban
- CiberNed, Network Center for Neurodegenerative Diseases, Spanish National Health Institute Carlos III, 28029 Madrid, Spain;
- Campus Bellaterra, Autonomous University of Barcelona, Cerdanyola del Vallés, 08193 Barcelona, Spain
| |
Collapse
|
6
|
Armando I, Cuevas S, Fan C, Kumar M, Izzi Z, Jose PA, Konkalmatt PR. G Protein-Coupled Receptor 37L1 Modulates Epigenetic Changes in Human Renal Proximal Tubule Cells. Int J Mol Sci 2022; 23:ijms232214456. [PMID: 36430934 PMCID: PMC9698582 DOI: 10.3390/ijms232214456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 10/19/2022] [Accepted: 10/31/2022] [Indexed: 11/23/2022] Open
Abstract
Renal luminal sodium transport is essential for physiological blood pressure control, and abnormalities in this process are strongly implicated in the pathogenesis of essential hypertension. Renal G protein-coupled receptors (GPCRs) are critical for the regulation of the reabsorption of essential nutrients, ions, and water from the glomerular filtrate. Recently, we showed that GPCR 37L1 (GPR37L1) is expressed on the apical membrane of renal proximal tubules (RPT) and regulates luminal sodium transport and blood pressure by modulating the function of the sodium proton exchanger 3 (NHE3). However, little is known about GPR37L1 intracellular signaling. Here, we show that GPR37L1 is localized to the nuclear membrane, in addition to the plasma membrane in human RPT cells. Furthermore, GPR37L1 signals via the PI3K/AKT/mTOR pathway to decrease the expression of DNA (cytosine-5)-methyltransferase 1 (DNMT1) and enhance NHE3 transcription. Overall, we demonstrate the direct role of a nuclear membrane GPCR in the regulation of renal sodium through epigenetic gene regulation.
Collapse
|
7
|
Isaac R, Vinik Y, Mikl M, Nadav-Eliyahu S, Shatz-Azoulay H, Yaakobi A, DeForest N, Majithia AR, Webster NJ, Shav-Tal Y, Elhanany E, Zick Y. A seven-transmembrane protein-TM7SF3, resides in nuclear speckles and regulates alternative splicing. iScience 2022; 25:105270. [PMID: 36304109 PMCID: PMC9593240 DOI: 10.1016/j.isci.2022.105270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 06/08/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022] Open
Abstract
The seven-transmembrane superfamily member 3 protein (TM7SF3) is a p53-regulated homeostatic factor that attenuates cellular stress and the unfolded protein response. Here we show that TM7SF3 localizes to nuclear speckles; eukaryotic nuclear bodies enriched in splicing factors. This unexpected location for a trans -membranal protein enables formation of stable complexes between TM7SF3 and pre-mRNA splicing factors including DHX15, LARP7, HNRNPU, RBM14, and HNRNPK. Indeed, TM7SF3 regulates alternative splicing of >330 genes, mainly at the 3'end of introns by directly modulating the activity of splicing factors such as HNRNPK. These effects are observed both in cell lines and primary human pancreatic islets. Accordingly, silencing of TM7SF3 results in differential expression of 1465 genes (about 7% of the human genome); with 844 and 621 genes being up- or down-regulated, respectively. Our findings implicate TM7SF3, as a resident protein of nuclear speckles and suggest a role for seven-transmembrane proteins as regulators of alternative splicing.
Collapse
Affiliation(s)
- Roi Isaac
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Yaron Vinik
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Martin Mikl
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
- Department of Biology, University of Haifa, Haifa, Israel
| | - Shani Nadav-Eliyahu
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Hadas Shatz-Azoulay
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Adi Yaakobi
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Natalie DeForest
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Biomedical Sciences Graduate Program, University of California San Diego, La Jolla, CA 92093, USA
| | - Amit R. Majithia
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Pediatrics, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Nicholas J.G. Webster
- Department of Medicine, School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- VA San Diego Healthcare System, San Diego, CA, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, USA
| | - Yaron Shav-Tal
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat Gan 5290002, Israel
| | - Eytan Elhanany
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| | - Yehiel Zick
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
8
|
Francis AJ, Firth JM, Sanchez-Alonso JL, Gorelik J, MacLeod KT. GPER limits adverse changes to Ca 2+ signalling and arrhythmogenic activity in ovariectomised guinea pig cardiomyocytes. Front Physiol 2022; 13:1023755. [PMID: 36439245 PMCID: PMC9686394 DOI: 10.3389/fphys.2022.1023755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 10/19/2022] [Indexed: 11/11/2022] Open
Abstract
Background: The increased risk of post-menopausal women developing abnormalities of heart function emphasises the requirement to understand the effect of declining oestrogen levels on cardiac electrophysiology and structure, and investigate possible therapeutic targets, namely the G protein-coupled oestrogen receptor 1 (GPER). Methods: Female guinea pigs underwent sham or ovariectomy (OVx) surgeries. Cardiomyocytes were isolated 150-days post-operatively. Membrane structure was assessed using di-8-ANEPPs staining and scanning ion conductance microscopy. Imunnohistochemistry (IHC) determined the localisation of oestrogen receptors. The effect of GPER activation on excitation-contraction coupling mechanisms were assessed using electrophysiological and fluorescence techniques. Downstream signalling proteins were investigated by western blot. Results: IHC staining confirmed the presence of nuclear oestrogen receptors and GPER, the latter prominently localised to the peri-nuclear region and having a clear striated pattern elsewhere in the cells. Following OVx, GPER expression increased and its activation reduced Ca2+ transient amplitude (by 40%) and sarcomere shortening (by 32%). In these cells, GPER activation reduced abnormal spontaneous Ca2+ activity, shortened action potential duration and limited drug-induced early after-depolarisation formation. Conclusion: In an animal species with comparable steroidogenesis and cardiac physiology to humans, we show the expression and localisation of all three oestrogen receptors in cardiac myocytes. We found that following oestrogen withdrawal, GPER expression increased and its activation limited arrhythmogenic behaviours in this low oestrogen state, indicating a potential cardioprotective role of this receptor in post-menopausal women.
Collapse
|
9
|
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]
|
10
|
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
|
11
|
Identification of candidate genes on the basis of SNP by time-lagged heat stress interactions for milk production traits in German Holstein cattle. PLoS One 2021; 16:e0258216. [PMID: 34648531 PMCID: PMC8516222 DOI: 10.1371/journal.pone.0258216] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Accepted: 09/21/2021] [Indexed: 01/22/2023] Open
Abstract
The aim of this study was to estimate genotype by time-lagged heat stress (HS) variance components as well as main and interaction SNP-marker effects for maternal HS during the last eight weeks of cow pregnancy, considering milk production traits recorded in the offspring generation. The HS indicator was the temperature humidity index (THI) for each week. A dummy variable with the code = 1 for the respective week for THI ≥ 60 indicated HS, otherwise, for no HS, the code = 0 was assigned. The dataset included test-day and lactation production traits from 14,188 genotyped first parity Holstein cows. After genotype quality control, 41,139 SNP markers remained for the genomic analyses. Genomic animal models without (model VC_nHS) and with in-utero HS effects (model VC_wHS) were applied to estimate variance components. Accordingly, for genome-wide associations, models GWA_nHS and GWA_wHS, respectively, were applied to estimate main and interaction SNP effects. Common genomic and residual variances for the same traits were very similar from models VC_nHS and VC_wHS. Genotype by HS interaction variances varied, depending on the week with in-utero HS. Among all traits, lactation milk yield with HS from week 5 displayed the largest proportion for interaction variances (0.07). For main effects from model GWA_wHS, 380 SNPs were suggestively associated with all production traits. For the SNP interaction effects from model GWA_wHS, we identified 31 suggestive SNPs, which were located in close distance to 62 potential candidate genes. The inferred candidate genes have various biological functions, including mechanisms of immune response, growth processes and disease resistance. Two biological processes excessively represented in the overrepresentation tests addressed lymphocyte and monocyte chemotaxis, ultimately affecting immune response. The modelling approach considering time-lagged genotype by HS interactions for production traits inferred physiological mechanisms being associated with health and immunity, enabling improvements in selection of robust animals.
Collapse
|
12
|
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
|
13
|
Yu R, Lin Z, Ouyang Z, Tao Z, Fan G. Blue light induces the nuclear translocation of neuropeptide receptor PAC1-R associated with the up-regulation of PAC1-R its own in reactive oxygen species associated way. Biochim Biophys Acta Gen Subj 2021; 1865:129884. [PMID: 33647387 DOI: 10.1016/j.bbagen.2021.129884] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/17/2021] [Accepted: 02/17/2021] [Indexed: 12/12/2022]
Abstract
PAC1-R is neuropeptide PACAP (pituitary adenylate cyclase activating polypeptide) preferring receptor mediates the antioxidant and cytoprotective effects of PACAP. It was found in this research that in both PAC1R-CHO cells with high expression of PAC1R-eGFP and retinal ganglion cells (RGC-5) with natural expression of PAC1-R, blue light and hydrogen peroxide (H2O2) trigger the significant nuclear translocation of PAC1-R, and the nuclear translocation of PAC1-R was positive correlation with the up-regulation of expression level and promoter activity of PAC1-R its own, while red light worked much less efficiently than blue light. Reactive oxygen species (ROS) scavenger NAC (N-acetyl-L-cysteine) and palmitoylation inhibitor 2-bromopalmitate (2-BP) disturbed the nuclear shifting associated with the correlative up-regulation of PAC1 significantly, and PAC1-R mutant (M-PAC1-R) on Cys25/Ala25 displayed the significant decreased nuclear trafficking efficiency. Furthermore, the Western Blot results with the antibody raised against the C-terminal of PAC1-R showing PAC1-R in the nucleus was fragmentation hinting that C-terminal of PAC1-R with theoretical nuclear location signal (NLS) may be involved in activation of PAC1-R promoter in the nucleus. All above results indicated that PAC1-R makes the nuclear translocation to trigger the activation of PAC1-R itself promoter resulting into the up-regulation of of PAC1-R in response to the oxidative stress induced by blue light and ROS such as H2O2 .
Collapse
Affiliation(s)
- Rongjie Yu
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China; Guangdong Province Key Laboratory of Bioengineering Medicine, Guangdong, China; Guangdong Provincial Biotechnology Drug & Engineering Technology Research Center, Guangdong, China; National Engineering Research Center of Genetic Medicine, Guangdong, China.
| | - Zhuochao Lin
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Zehua Ouyang
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Zhengxin Tao
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| | - Guangchun Fan
- Department of Cell Biology, College of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
14
|
Zhang T, Ahn K, Emerick B, Modarai SR, Opdenaker LM, Palazzo J, Schleiniger G, Fields JZ, Boman BM. APC mutations in human colon lead to decreased neuroendocrine maturation of ALDH+ stem cells that alters GLP-2 and SST feedback signaling: Clue to a link between WNT and retinoic acid signalling in colon cancer development. PLoS One 2020; 15:e0239601. [PMID: 33112876 PMCID: PMC7592776 DOI: 10.1371/journal.pone.0239601] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/09/2020] [Indexed: 12/15/2022] Open
Abstract
APC mutations drive human colorectal cancer (CRC) development. A major contributing factor is colonic stem cell (SC) overpopulation. But, the mechanism has not been fully identified. A possible mechanism is the dysregulation of neuroendocrine cell (NEC) maturation by APC mutations because SCs and NECs both reside together in the colonic crypt SC niche where SCs mature into NECs. So, we hypothesized that sequential inactivation of APC alleles in human colonic crypts leads to progressively delayed maturation of SCs into NECs and overpopulation of SCs. Accordingly, we used quantitative immunohistochemical mapping to measure indices and proportions of SCs and NECs in human colon tissues (normal, adenomatous, malignant), which have different APC-zygosity states. In normal crypts, many cells staining for the colonic SC marker ALDH1 co-stained for chromogranin-A (CGA) and other NEC markers. In contrast, in APC-mutant tissues from familial adenomatous polyposis (FAP) patients, the proportion of ALDH+ SCs progressively increased while NECs markedly decreased. To explain how these cell populations change in FAP tissues, we used mathematical modelling to identify kinetic mechanisms. Computational analyses indicated that APC mutations lead to: 1) decreased maturation of ALDH+ SCs into progenitor NECs (not progenitor NECs into mature NECs); 2) diminished feedback signaling by mature NECs. Biological experiments using human CRC cell lines to test model predictions showed that mature GLP-2R+ and SSTR1+ NECs produce, via their signaling peptides, opposing effects on rates of NEC maturation via feedback regulation of progenitor NECs. However, decrease in this feedback signaling wouldn't explain the delayed maturation because both progenitor and mature NECs are depleted in CRCs. So the mechanism for delayed maturation must explain how APC mutation causes the ALDH+ SCs to remain immature. Given that ALDH is a key component of the retinoic acid (RA) signaling pathway, that other components of the RA pathway are selectively expressed in ALDH+ SCs, and that exogenous RA ligands can induce ALDH+ cancer SCs to mature into NECs, RA signaling must be attenuated in ALDH+ SCs in CRC. Thus, attenuation of RA signaling explains why ALDH+ SCs remain immature in APC mutant tissues. Since APC mutation causes increased WNT signaling in FAP and we found that sequential inactivation of APC in FAP patient tissues leads to progressively delayed maturation of colonic ALDH+ SCs, the hypothesis is developed that human CRC evolves due to an imbalance between WNT and RA signaling.
Collapse
Affiliation(s)
- Tao Zhang
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, United States of America
- University of Delaware, Newark, DE, United States of America
- Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Koree Ahn
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, United States of America
- University of Delaware, Newark, DE, United States of America
- Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Brooks Emerick
- Center for Applications of Mathematics in Medicine, Department of Mathematical Sciences, University of Delaware, Newark, DE, United States of America
| | - Shirin R. Modarai
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, United States of America
- University of Delaware, Newark, DE, United States of America
| | - Lynn M. Opdenaker
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, United States of America
- University of Delaware, Newark, DE, United States of America
| | - Juan Palazzo
- Thomas Jefferson University, Philadelphia, PA, United States of America
| | - Gilberto Schleiniger
- Center for Applications of Mathematics in Medicine, Department of Mathematical Sciences, University of Delaware, Newark, DE, United States of America
| | | | - Bruce M. Boman
- Cawley Center for Translational Cancer Research, Helen F. Graham Cancer Center and Research Institute, Newark, DE, United States of America
- University of Delaware, Newark, DE, United States of America
- Thomas Jefferson University, Philadelphia, PA, United States of America
- Center for Applications of Mathematics in Medicine, Department of Mathematical Sciences, University of Delaware, Newark, DE, United States of America
| |
Collapse
|
15
|
Mohammad Nezhady MA, Rivera JC, Chemtob S. Location Bias as Emerging Paradigm in GPCR Biology and Drug Discovery. iScience 2020; 23:101643. [PMID: 33103080 PMCID: PMC7569339 DOI: 10.1016/j.isci.2020.101643] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
GPCRs are the largest receptor family that are involved in virtually all biological processes. Pharmacologically, they are highly druggable targets, as they cover more than 40% of all drugs in the market. Our knowledge of biased signaling provided insight into pharmacology vastly improving drug design to avoid unwanted effects and achieve higher efficacy and selectivity. However, yet another feature of GPCR biology is left largely unexplored, location bias. Recent developments in this field show promising avenues for evolution of new class of pharmaceuticals with greater potential for higher level of precision medicine. Further consideration and understanding of this phenomenon with deep biochemical and molecular insights would pave the road to success. In this review, we critically analyze this perspective and discuss new avenues of investigation.
Collapse
Affiliation(s)
- Mohammad Ali Mohammad Nezhady
- Programmes en Biologie Moléculaire, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche du CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Corresponding author
| | | | - Sylvain Chemtob
- Programmes en Biologie Moléculaire, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche du CHU Sainte-Justine, Montreal, QC H3T 1C5, Canada
- Maisonneuve-Rosemont Hospital Research Centre, Montreal, QC, Canada
| |
Collapse
|
16
|
Rukavina Mikusic NL, Silva MG, Pineda AM, Gironacci MM. Angiotensin Receptors Heterodimerization and Trafficking: How Much Do They Influence Their Biological Function? Front Pharmacol 2020; 11:1179. [PMID: 32848782 PMCID: PMC7417933 DOI: 10.3389/fphar.2020.01179] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2020] [Accepted: 07/20/2020] [Indexed: 01/03/2023] Open
Abstract
G-protein–coupled receptors (GPCRs) are targets for around one third of currently approved and clinical prescribed drugs and represent the largest and most structurally diverse family of transmembrane signaling proteins, with almost 1000 members identified in the human genome. Upon agonist stimulation, GPCRs are internalized and trafficked inside the cell: they may be targeted to different organelles, recycled back to the plasma membrane or be degraded. Once inside the cell, the receptors may initiate other signaling pathways leading to different biological responses. GPCRs’ biological function may also be influenced by interaction with other receptors. Thus, the ultimate cellular response may depend not only on the activation of the receptor from the cell membrane, but also from receptor trafficking and/or the interaction with other receptors. This review is focused on angiotensin receptors and how their biological function is influenced by trafficking and interaction with others receptors.
Collapse
Affiliation(s)
- Natalia L Rukavina Mikusic
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Dpto. Química Biológica, IQUIFIB (UBA-CONICET), Buenos Aires, Argentina
| | - Mauro G Silva
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Dpto. Química Biológica, IQUIFIB (UBA-CONICET), Buenos Aires, Argentina
| | - Angélica M Pineda
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Dpto. Química Biológica, IQUIFIB (UBA-CONICET), Buenos Aires, Argentina
| | - Mariela M Gironacci
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Dpto. Química Biológica, IQUIFIB (UBA-CONICET), Buenos Aires, Argentina
| |
Collapse
|
17
|
Kim BH, Wang FI, Pereverzev A, Chidiac P, Dixon SJ. Toward Defining the Pharmacophore for Positive Allosteric Modulation of PTH1 Receptor Signaling by Extracellular Nucleotides. ACS Pharmacol Transl Sci 2019; 2:155-167. [PMID: 32259054 DOI: 10.1021/acsptsci.8b00053] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Indexed: 12/17/2022]
Abstract
The parathyroid hormone 1 receptor (PTH1R) is a Class B G-protein-coupled receptor that is a target for osteoporosis therapeutics. Activated PTH1R couples through Gs to the stimulation of adenylyl cyclase. As well, β-arrestin is recruited to PTH1R leading to receptor internalization and MAPK/ERK signaling. Previously, we reported that the agonist potency of PTH1R is increased in the presence of extracellular ATP, which acts as a positive allosteric modulator of PTH signaling. Another nucleotide, cytidine 5'-monophosphate (CMP), also enhances PTH1R signaling, suggesting that ATP and CMP share a moiety responsible for positive allostery, possibly ribose-5-phosphate. Therefore, we examined the effect of extracellular sugar phosphates on PTH1R signaling. cAMP levels and β-arrestin recruitment were monitored using luminescence-based assays. Alone, ribose-5-phosphate had no detectable effect on adenylyl cyclase activity in UMR-106 rat osteoblastic cells, which endogenously express PTH1R. However, ribose-5-phosphate markedly enhanced the activation of adenylyl cyclase induced by PTH. Other sugar phosphates, including glucose-1-phosphate, glucose-6-phosphate, fructose-6-phosphate, and fructose-1,6-bisphosphate, also potentiated PTH-induced adenylyl cyclase activation. As well, some sugar phosphates enhanced PTH-induced β-arrestin recruitment to human PTH1R heterologously expressed in HEK293H cells. Interestingly, the effects of glucose-1-phosphate were greater than those of its isomer glucose-6-phosphate. Our results suggest that phosphorylated monosaccharides such as ribose-5-phosphate contain the pharmacophore for positive allosteric modulation of PTH1R. At least in some cases, the extent of modulation depends on the position of the phosphate group. Knowledge of the pharmacophore may permit future development of positive allosteric modulators to increase the therapeutic efficacy of PTH1R agonists.
Collapse
Affiliation(s)
- Brandon H Kim
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry; and Bone and Joint Institute; The University of Western Ontario, London, Canada
| | - Fang I Wang
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry; and Bone and Joint Institute; The University of Western Ontario, London, Canada
| | - Alexey Pereverzev
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry; and Bone and Joint Institute; The University of Western Ontario, London, Canada
| | - Peter Chidiac
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry; and Bone and Joint Institute; The University of Western Ontario, London, Canada
| | - S Jeffrey Dixon
- Department of Physiology and Pharmacology, Schulich School of Medicine & Dentistry; and Bone and Joint Institute; The University of Western Ontario, London, Canada
| |
Collapse
|
18
|
Chandrasekharan JA, Sharma-Walia N. Arachidonic Acid Derived Lipid Mediators Influence Kaposi's Sarcoma-Associated Herpesvirus Infection and Pathogenesis. Front Microbiol 2019; 10:358. [PMID: 30915039 PMCID: PMC6422901 DOI: 10.3389/fmicb.2019.00358] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 02/11/2019] [Indexed: 12/30/2022] Open
Abstract
Kaposi’s sarcoma-associated herpesvirus (KSHV) infection, particularly latent infection is often associated with inflammation. The arachidonic acid pathway, the home of several inflammation and resolution associated lipid mediators, is widely altered upon viral infections. Several in vitro studies show that these lipid mediators help in the progression of viral pathogenesis. This review summarizes the findings related to human herpesvirus KSHV infection and arachidonic acid pathway metabolites. KSHV infection has been shown to promote inflammation by upregulating cyclooxygenase-2 (COX-2), 5 lipoxygenase (5LO), and their respective metabolites prostaglandin E2 (PGE2) and leukotriene B4 (LTB4) to promote latency and an inflammatory microenvironment. Interestingly, the anti-inflammatory lipid mediator lipoxin is downregulated during KSHV infection to facilitate infected cell survival. These studies aid in understanding the role of arachidonic acid pathway metabolites in the progression of viral infection, the host inflammatory response, and pathogenesis. With limited therapeutic options to treat KSHV infection, use of inhibitors to these inflammatory metabolites and their synthetic pathways or supplementing anti-inflammatory lipid mediators could be an effective alternative therapeutic.
Collapse
Affiliation(s)
- Jayashree A Chandrasekharan
- Department of Microbiology and Immunology, H.M. Bligh Cancer Research Laboratories, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| | - Neelam Sharma-Walia
- Department of Microbiology and Immunology, H.M. Bligh Cancer Research Laboratories, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, United States
| |
Collapse
|
19
|
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
|
20
|
Mediterranean diet and platelet-activating factor; a systematic review. Clin Biochem 2018; 60:1-10. [PMID: 30142319 DOI: 10.1016/j.clinbiochem.2018.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Revised: 08/16/2018] [Accepted: 08/16/2018] [Indexed: 12/14/2022]
Abstract
Platelet-activating factor (PAF) is a glycerylether lipid and one of the most potent endogenous mediators of inflammation. Through its binding to a well-characterized receptor it initiates a plethora of cellular pro-inflammatory actions participating by this way to the pathology of most chronic diseases, including cardiovascular and renal diseases, CNS decline and cancer. Among the variety of prudent dietary patterns, Mediterranean Diet (MD) is the dietary pattern with the strongest evidence for its ability to prevent the same chronic diseases. In addition, micronutrients and extracts from several components and characteristic food of the MD can favorably modulate PAF's actions and metabolism either directly or indirectly. However, the role of this traditional diet on PAF metabolism and actions has rarely been studied before. This systematic review summarizes, presents and discusses the outcomes of epidemiologic and intervention studies in humans, investigating the relationships between PAF status and MD. Seventeen full-text articles trying to interlink the components of MD and PAF are found and presented. The results are inconsistent due to the variability of the measured indices and methodology followed. However, preliminary results indicate that the characteristic "healthy" components of the MD, especially, cereals, legumes, vegetables, fish and wine can favorably modulate the pro-inflammatory actions of PAF and regulate its metabolism. Larger, well-controlled studies are necessary to elucidate whether the attenuation of PAF actions can mediate the preventive properties of MD against chronic diseases.
Collapse
|
21
|
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
|
22
|
Yu R, Liu H, Peng X, Cui Y, Song S, Wang L, Zhang H, Hong A, Zhou T. The palmitoylation of the N-terminal extracellular Cys37 mediates the nuclear translocation of VPAC1 contributing to its anti-apoptotic activity. Oncotarget 2018; 8:42728-42741. [PMID: 28473666 PMCID: PMC5522101 DOI: 10.18632/oncotarget.17449] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 04/14/2017] [Indexed: 12/17/2022] Open
Abstract
VPAC1 is class B G protein-coupled receptors (GPCR) shared by pituitary adenylate cyclase activating polypeptide (PACAP) and vasoactive intestinal peptide (VIP). The first cysteine (Cys37) in the N-terminal extracellular domain of mature VPAC1 is a free Cys not involved in the formation of conserved intramolecular disulfide bonds. In order to investigate the biological role of this Cys37 in VPAC1, the wild-type VPAC1 and Cys37/Ala mutant (VPAC1-C37/A) were expressed stably as fusion proteins with enhanced yellow fluorescent protein (EYFP) respectively in Chinese hamster ovary (CHO) cells. Both VPAC1-EYFP and VPAC1-C37/A-EYFP trafficked to the plasma membrane normally, and CHO cells expressing VPAC1-EYFP displayed higher anti-apoptotic activity against camptothecin (CPT) induced apoptosis than the cells expressing VPAC1-C37/A-EYFP, while VPAC1-C37/A-CHO cells showed higher proliferative activity than VPAC1-CHO cells. Confocal microscopic analysis, western blotting and fluorescence quantification assay showed VPAC1-EYFP displayed significant nuclear translocation while VPAC1-C37/A-EYFP did not transfer into nucleus under the stimulation of VIP (0.1 nM). Acyl-biotin exchange assay and click chemistry-based palmitoylation assay confirmed for the first time the palmitoylation of Cys37, which has been predicted by bioinformatics analysis. And the palmitoylation inhibitor 2-bromopalmitate significantly inhibited the nuclear translocation of VPAC1-EYFP and its anti-apoptotic activity synchronously. These results indicated the palmitoylation of the Cys37 in the N-terminal extracellular domain of VPAC1 mediates the nuclear translocation of VPAC1 contributing to its anti-apoptotic activity. These findings reveal for the first time the lipidation-mediating nuclear translocation of VPAC1 produces a novel anti-apoptotic signal pathway, which may help to promote new drug development strategy targeting VPAC1.
Collapse
Affiliation(s)
- Rongjie Yu
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Hongyu Liu
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Xinhe Peng
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Yue Cui
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Suqin Song
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Like Wang
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Huahua Zhang
- Department of Medical Genetics, Guangdong Medical University, Dongguan, Guangdong, China
| | - An Hong
- Institute of Biomedicine, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China.,National Engineering Research Center of Genetic Medicine, Jinan University, Guangzhou, Guangdong, China
| | - Tianhong Zhou
- Department of Bioengineering, School of Life Science and Technology, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
23
|
Pupo M, Bodmer A, Berto M, Maggiolini M, Dietrich PY, Picard D. A genetic polymorphism repurposes the G-protein coupled and membrane-associated estrogen receptor GPER to a transcription factor-like molecule promoting paracrine signaling between stroma and breast carcinoma cells. Oncotarget 2018; 8:46728-46744. [PMID: 28596490 PMCID: PMC5564519 DOI: 10.18632/oncotarget.18156] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 05/10/2017] [Indexed: 01/08/2023] Open
Abstract
GPER is a membrane-associated estrogen receptor of the family of G-protein coupled receptors. For breast cancer, the contribution of GPER to promoting the proliferation and migration of both carcinoma cells and cancer-associated fibroblasts (CAFs) in response to estrogen and other agonists has extensively been investigated. Intriguingly, GPER was previously found to be localized to the nucleus in one isolate of breast CAFs. Moreover, this nuclear GPER was shown to bind regulatory sequences of cancer-relevant target genes and to induce their expression. We decided to find out what induces the nuclear localization of GPER, how general this phenomenon is, and what its functional significance is. We discovered that interfering with N-linked glycosylation of GPER, either by mutation of the predicted glycosylation sites or pharmacologically with tunicamycin, drives GPER into the nucleus. Surveying a small set of CAFs from breast cancer biopsies, we found that a relatively common single nucleotide polymorphism, which results in the expression of a GPER variant with the amino acid substitution P16L, is associated with the nuclear localization of GPER. GPER with P16L fails to be glycosylated, presumably because of a conformational effect on the nearby glycosylation sites. GPER P16L is defective for membrane-associated signaling, but instead acts like an estrogen-stimulated transcription factor. In CAFs, it induces the secretion of paracrine factors that promote the migration of carcinoma cells. This raises the possibility that the GPER P16L polymorphism could be a risk factor for breast cancer.
Collapse
Affiliation(s)
- Marco Pupo
- Département de Biologie Cellulaire and Institute of Genetics and Genomics of Geneva, Université de Genève, Sciences III, CH-1211 Genève 4, Switzerland.,Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.,Current address: Areta International S.r.l., Gerenzano, Italy
| | - Alexandre Bodmer
- Département d'Oncologie, Hôpitaux Universitaires de Genève, CH - 1211 Genève 14, Switzerland
| | - Melissa Berto
- Département de Biologie Cellulaire and Institute of Genetics and Genomics of Geneva, Université de Genève, Sciences III, CH-1211 Genève 4, Switzerland
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy
| | - Pierre-Yves Dietrich
- Département d'Oncologie, Hôpitaux Universitaires de Genève, CH - 1211 Genève 14, Switzerland
| | - Didier Picard
- Département de Biologie Cellulaire and Institute of Genetics and Genomics of Geneva, Université de Genève, Sciences III, CH-1211 Genève 4, Switzerland
| |
Collapse
|
24
|
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
|
25
|
Lee HY, Jeong YS, Lee M, Kweon HS, Huh YH, Park JS, Hwang JE, Kim K, Bae YS. Intracellular formyl peptide receptor regulates naïve CD4 T cell migration. Biochem Biophys Res Commun 2018; 497:226-232. [PMID: 29427663 DOI: 10.1016/j.bbrc.2018.02.060] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 02/07/2018] [Indexed: 12/20/2022]
Abstract
We found that formyl peptide receptor (FPR) 1 and FPR3 were expressed intracellularly and/or the nucleus of naïve CD4 T cell. Activation of naïve CD4 T cells with synthetic intracellular agonists dTAT-WKYMVm and CTP-WKYMVm for FPR members stimulated CD4 T cell migration via pertussis toxin-sensitive manner. Knockdown of FPR1, but not knockdown of FPR3, blocked dTAT-WKYMVm-induced naïve CD4 T cell migration. Stimulation of naïve CD4 T cells with dTAT-WKYMVm elicited the activation of ERK, p38 MAPK, and Akt. Activation of CD4 T cells with anti-CD3 and anti-CD28 antibodies caused surface expression of FPR1 and FPR3, but not FPR2. CD4 T cells isolated from sepsis patients expressed the three members of FPR family on their cell surface. Taken together, our results suggest that intracellular FPR in naïve CD4 T cells and surface FPRs in activated CD4 T cells might regulate immune responses by regulating CD4 T cell activity.
Collapse
Affiliation(s)
- Ha Young Lee
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea.
| | - Yu Sun Jeong
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Mingyu Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea
| | - Hee-Seok Kweon
- Electron Microscopy Research Center, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Yang Hoon Huh
- Electron Microscopy Research Center, Korea Basic Science Institute, Cheongju, 28119, Republic of Korea
| | - Joon Seong Park
- Department of Hematology-Oncology, Ajou University School of Medicine, Suwon, 16499, Republic of Korea
| | - Ji Eun Hwang
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Kyuseok Kim
- Department of Emergency Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, Republic of Korea
| | - Yoe-Sik Bae
- Department of Biological Sciences, Sungkyunkwan University, Suwon, 16419, Republic of Korea; Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul, 06351, Republic of Korea.
| |
Collapse
|
26
|
Lappano R, Maggiolini M. GPER is involved in the functional liaison between breast tumor cells and cancer-associated fibroblasts (CAFs). J Steroid Biochem Mol Biol 2018; 176:49-56. [PMID: 28249728 DOI: 10.1016/j.jsbmb.2017.02.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 02/02/2017] [Accepted: 02/23/2017] [Indexed: 12/17/2022]
Abstract
The aggressiveness of breast tumors is deeply influenced by the surrounding stroma. In this regard, the functional crosstalk between cancer cells and the tumor microenvironment has received considerable attention in recent years. Cancer-associated fibroblasts (CAFs) are active components of the tumor stroma as they play a main role in the initiation, progression, metastasis and recurrence of breast malignancy. Hence, a better understanding of the mechanisms through which host stroma may contribute to cancer development would lead to novel therapeutic approaches aimed to target both tumor cells and the adjacent microenvironment. The G protein estrogen receptor (GPER/GPR30) has been involved in estrogenic signaling in normal and malignant cells, including breast cancer. It is noteworthy that the potential of GPER to mediate stimulatory effects of estrogens has been also shown in CAFs derived from patients with breast tumors, suggesting that GPER may act at the cross-road between cancer cells and these important components of the tumor microenvironment. This review recapitulates recent findings underlying the breast tumor-promoting action of CAFs, in particular their functional liaison with breast cancer cells via GPER toward the occurrence of malignant features.
Collapse
Affiliation(s)
- Rosamaria Lappano
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| | - Marcello Maggiolini
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende, Italy.
| |
Collapse
|
27
|
Dubuc C, Savard M, Bovenzi V, Lessard A, Fortier A, Côté J, Neugebauer W, Rizzolio F, Geha S, Giordano A, Chemtob S, Gobeil F. Targeting intracellular B2 receptors using novel cell-penetrating antagonists to arrest growth and induce apoptosis in human triple-negative breast cancer. Oncotarget 2018. [PMID: 29515778 PMCID: PMC5839409 DOI: 10.18632/oncotarget.24009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are integral cell-surface proteins having a central role in tumor growth and metastasis. However, several GPCRs retain an atypical intracellular/nuclear location in various types of cancer. The pathological significance of this is currently unknown. Here we extend this observation by showing that the bradykinin B2R (BK-B2R) is nuclearly expressed in the human triple-negative breast cancer (TNBC) cell line MDA-MB-231 and in human clinical specimens of TNBC. We posited that these “nuclearized” receptors could be involved in oncogenic signaling linked to aberrant growth and survival maintenance of TNBC. We used cell-penetrating BK-B2R antagonists, including FR173657 and novel transducible, cell-permeable forms of the peptide B2R antagonist HOE 140 (NG68, NG134) to demonstrate their superior efficacy over impermeable ones (HOE 140), in blocking proliferation and promoting apoptosis of MDA-MB-231 cells. Some showed an even greater antineoplastic activity over conventional chemotherapeutic drugs in vitro. The cell-permeable B2R antagonists had less to no anticancer effects on B2R shRNA-knockdown or non-B2R expressing (COS-1) cells, indicating specificity in their action. Possible mechanisms of their anticancer effects may involve activation of p38kinase/p27Kip1 pathways. Together, our data support the existence of a possible intracrine signaling pathway via internal/nuclear B2R, critical for the growth of TNBC cells, and identify new chemical entities that enable to target the corresponding intracellular GPCRs.
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, MD, United States
| | - Audrey Fortier
- 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
| | - 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
| | - Flavio Rizzolio
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, USA.,Dipartimento di Scienze Molecolari e Nanosistemi, Università Ca' Foscari Venezia, Mestre-Venezia, Italy
| | - Sameh Geha
- Department of Pathology, Centre Hospitalier Universitaire de Sherbrooke, Sherbrooke, Québec, Canada
| | - Antonio Giordano
- Department of Biology, Sbarro Institute for Cancer Research and Molecular Medicine, Temple University, Philadelphia, PA, USA
| | - Sylvain Chemtob
- Department of Pediatrics, Centre Hospitalier Universitaire 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
|
28
|
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
|
29
|
Radu BM, Osculati AMM, Suku E, Banciu A, Tsenov G, Merigo F, Di Chio M, Banciu DD, Tognoli C, Kacer P, Giorgetti A, Radu M, Bertini G, Fabene PF. All muscarinic acetylcholine receptors (M 1-M 5) are expressed in murine brain microvascular endothelium. Sci Rep 2017; 7:5083. [PMID: 28698560 PMCID: PMC5506046 DOI: 10.1038/s41598-017-05384-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Accepted: 05/30/2017] [Indexed: 12/25/2022] Open
Abstract
Clinical and experimental studies indicate that muscarinic acetylcholine receptors are potential pharmacological targets for the treatment of neurological diseases. Although these receptors have been described in human, bovine and rat cerebral microvascular tissue, a subtype functional characterization in mouse brain endothelium is lacking. Here, we show that all muscarinic acetylcholine receptors (M1-M5) are expressed in mouse brain microvascular endothelial cells. The mRNA expression of M2, M3, and M5 correlates with their respective protein abundance, but a mismatch exists for M1 and M4 mRNA versus protein levels. Acetylcholine activates calcium transients in brain endothelium via muscarinic, but not nicotinic, receptors. Moreover, although M1 and M3 are the most abundant receptors, only a small fraction of M1 is present in the plasma membrane and functions in ACh-induced Ca2+ signaling. Bioinformatic analyses performed on eukaryotic muscarinic receptors demonstrate a high degree of conservation of the orthosteric binding site and a great variability of the allosteric site. In line with previous studies, this result indicates muscarinic acetylcholine receptors as potential pharmacological targets in future translational studies. We argue that research on drug development should especially focus on the allosteric binding sites of the M1 and M3 receptors.
Collapse
Affiliation(s)
- Beatrice Mihaela Radu
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, 37134, Italy.,Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Bucharest, 050095, Romania
| | | | - Eda Suku
- Department of Biotechnology, University of Verona, Verona, 37134, Italy
| | - Adela Banciu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Bucharest, 050095, Romania.,Engineering Faculty, Constantin Brancusi' University, Calea Eroilor 30, Targu Jiu, 210135, Romania
| | - Grygoriy Tsenov
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, 37134, Italy
| | - Flavia Merigo
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, 37134, Italy
| | - Marzia Di Chio
- Department of Public Health and Community Medicine, University of Verona, Verona, 37134, Italy
| | - Daniel Dumitru Banciu
- Department of Anatomy, Animal Physiology and Biophysics, Faculty of Biology, University of Bucharest, Bucharest, 050095, Romania
| | - Cristina Tognoli
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, 37134, Italy
| | - Petr Kacer
- National Institute of Mental Health, Klecany, 25067, Czech Republic
| | | | - Mihai Radu
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, 37134, Italy. .,Department of Life and Environmental Physics, Horia Hulubei National Institute for Physics and Nuclear Engineering, PO Box MG-6, Reactorului 30, Magurele, 077125, Romania.
| | - Giuseppe Bertini
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, 37134, Italy
| | - Paolo Francesco Fabene
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Verona, 37134, Italy
| |
Collapse
|
30
|
Thomas M, Snead D, Mitchell D. An investigation into the potential role of brain angiogenesis inhibitor protein 3 (BAI3) in the tumorigenesis of small-cell carcinoma: a review of the surrounding literature. J Recept Signal Transduct Res 2017; 37:325-334. [PMID: 28537194 DOI: 10.1080/10799893.2017.1328441] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Brain angiogenesis inhibitor protein 3 (BAI3) is from the adhesion group of seven-transmembrane spanning G protein-coupled receptors (GPCRs) and has been identified via gene expression profiling as being upregulated in small-cell lung cancer (SCLC) tumors. It has subsequently been validated as a sensitive and specific immunohistochemical marker for SCLC, helping to differentiate these tumors from morphologically similar large-cell neuroendocrine (LCNEC) malignancies. It is, however, still unclear as to the role BAI3 proteins might play in SCLC and indeed how they might contribute to tumorigenesis. Interestingly, the pattern of staining observed on immunohistochemistry was in fact nuclear as opposed to the membranous staining pattern expected of transmembrane-bound molecules. This fact has lead the authors to believe that the protein receptor is structurally altered in SCLC and that this modification may confer different behavioral properties that contribute toward tumorigenesis. Nuclear localization is not unique to BAI3 and has been reported in a number of GPCRs and frequently correlates with survival outcomes. BAI3 has the potential to act as target for pharmaceutical intervention inline with developing trends in molecular pathology aiming to provide personalized, treatment regimes based on tumor-specific mutation profiles. The adhesion group of the GPCR superfamily is still poorly understood. We present a review of the existing literature regarding the role they play in both physiological and disease states and the mechanisms by which they influence a range of cellular processes.
Collapse
Affiliation(s)
- Michael Thomas
- a Department of Histopathology , University Hospitals Coventry and Warwickshire , Coventry , UK
| | - David Snead
- a Department of Histopathology , University Hospitals Coventry and Warwickshire , Coventry , UK
| | - Daniel Mitchell
- b Department of Translational Medicine , University of Warwick , Coventry , UK
| |
Collapse
|
31
|
Thomas P, Pang Y, Dong J. Membrane androgen receptor characteristics of human ZIP9 (SLC39A) zinc transporter in prostate cancer cells: Androgen-specific activation and involvement of an inhibitory G protein in zinc and MAP kinase signaling. Mol Cell Endocrinol 2017; 447:23-34. [PMID: 28219737 DOI: 10.1016/j.mce.2017.02.025] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 02/14/2017] [Accepted: 02/14/2017] [Indexed: 11/29/2022]
Abstract
Characteristics of novel human membrane androgen receptor (mAR), ZIP9 (SLC39A9), were investigated in ZIP9-transfected PC-3 cells (PC3-ZIP9). Ligand blot analysis showed plasma membrane [3H]-T binding corresponds to the position of ZIP9 on Western blots which suggests ZIP9 can bind [3H]-T alone, without a protein partner. Progesterone antagonized testosterone actions, blocking increases in zinc, Erk phosphorylation and apoptosis, further evidence that ZIP9 is specifically activated by androgens. Pre-treatment with GTPγS and pertussis toxin decreased plasma membrane [3H]-T binding and blocked testosterone-induced increases in Erk phosphorylation and intracellular zinc, indicating ZIP9 is coupled to an inhibitory G protein (Gi) that mediates both MAP kinase and zinc signaling. Testosterone treatment of nuclei and mitochondria which express ZIP9 decreased their zinc contents, suggesting ZIP9 also regulates free zinc through releasing it from these intracellular organelles. The results show ZIP9 is a specific Gi coupled-mAR mediating testosterone-induced MAP kinase and zinc signaling in PC3-ZIP9 cells.
Collapse
Affiliation(s)
- Peter Thomas
- University of Texas Marine Science Institute, 750 Channel View Drive, Port Aransas, TX, 78373, USA.
| | - Yefei Pang
- University of Texas Marine Science Institute, 750 Channel View Drive, Port Aransas, TX, 78373, USA
| | - Jing Dong
- University of Texas Marine Science Institute, 750 Channel View Drive, Port Aransas, TX, 78373, USA
| |
Collapse
|
32
|
A Review on Platelet Activating Factor Inhibitors: Could a New Class of Potent Metal-Based Anti-Inflammatory Drugs Induce Anticancer Properties? Bioinorg Chem Appl 2017; 2017:6947034. [PMID: 28458618 PMCID: PMC5387815 DOI: 10.1155/2017/6947034] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Accepted: 02/27/2017] [Indexed: 12/12/2022] Open
Abstract
In this minireview, we refer to recent results as far as the Platelet Activating Factor (PAF) inhibitors are concerned. At first, results of organic compounds (natural and synthetic ones and specific and nonspecific) as inhibitors of PAF are reported. Emphasis is given on recent results about a new class of the so-called metal-based inhibitors of PAF. A small library of 30 metal complexes has been thus created; their anti-inflammatory activity has been further evaluated owing to their inhibitory effect against PAF in washed rabbit platelets (WRPs). In addition, emphasis has also been placed on the identification of preliminary structure-activity relationships for the different classes of metal-based inhibitors.
Collapse
|
33
|
Sergin I, Jong YJI, Harmon SK, Kumar V, O'Malley KL. Sequences within the C Terminus of the Metabotropic Glutamate Receptor 5 (mGluR5) Are Responsible for Inner Nuclear Membrane Localization. J Biol Chem 2017; 292:3637-3655. [PMID: 28096465 DOI: 10.1074/jbc.m116.757724] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Revised: 01/12/2017] [Indexed: 12/19/2022] Open
Abstract
Traditionally, G-protein-coupled receptors (GPCR) are thought to be located on the cell surface where they transmit extracellular signals to the cytoplasm. However, recent studies indicate that some GPCRs are also localized to various subcellular compartments such as the nucleus where they appear required for various biological functions. For example, the metabotropic glutamate receptor 5 (mGluR5) is concentrated at the inner nuclear membrane (INM) where it mediates Ca2+ changes in the nucleoplasm by coupling with Gq/11 Here, we identified a region within the C-terminal domain (amino acids 852-876) that is necessary and sufficient for INM localization of the receptor. Because these sequences do not correspond to known nuclear localization signal motifs, they represent a new motif for INM trafficking. mGluR5 is also trafficked to the plasma membrane where it undergoes re-cycling/degradation in a separate receptor pool, one that does not interact with the nuclear mGluR5 pool. Finally, our data suggest that once at the INM, mGluR5 is stably retained via interactions with chromatin. Thus, mGluR5 is perfectly positioned to regulate nucleoplasmic Ca2+in situ.
Collapse
Affiliation(s)
- Ismail Sergin
- From the Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Yuh-Jiin I Jong
- From the Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Steven K Harmon
- From the Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Vikas Kumar
- From the Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Karen L O'Malley
- From the Department of Neuroscience, Washington University School of Medicine, St. Louis, Missouri 63110
| |
Collapse
|
34
|
Boosting Adaptive Immunity: A New Role for PAFR Antagonists. Sci Rep 2016; 6:39146. [PMID: 27966635 PMCID: PMC5155422 DOI: 10.1038/srep39146] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 11/18/2016] [Indexed: 02/02/2023] Open
Abstract
We have previously shown that the Platelet-Activating Factor Receptor (PAFR) engagement in murine macrophages and dendritic cells (DCs) promotes a tolerogenic phenotype reversed by PAFR-antagonists treatment in vitro. Here, we investigated whether a PAFR antagonist would modulate the immune response in vivo. Mice were subcutaneously injected with OVA or OVA with PAFR-antagonist WEB2170 on days 0 and 7. On day 14, OVA–specific IgG2a and IgG1 were measured in the serum. The presence of WEB2170 during immunization significantly increased IgG2a without affecting IgG1 levels. When WEB2170 was added to OVA in complete Freund’s adjuvant, enhanced IgG2a but not IgG1 production was also observed, and CD4+ FoxP3+ T cell frequency in the spleen was reduced compared to mice immunized without the antagonist. Similar results were observed in PAFR-deficient mice, along with increased Tbet mRNA expression in the spleen. Additionally, bone marrow-derived DCs loaded with OVA were transferred into naïve mice and their splenocytes were co-cultured with fresh OVA-loaded DCs. CD4+ T cell proliferation was higher in the group transferred with DCs treated with the PAFR-antagonist. We propose that the activation of PAFR by ligands present in the site of immunization is able to fine-tune the adaptive immune response.
Collapse
|
35
|
Bhosle VK, Rivera JC, Zhou TE, Omri S, Sanchez M, Hamel D, Zhu T, Rouget R, Rabea AA, Hou X, Lahaie I, Ribeiro-da-Silva A, Chemtob S. Nuclear localization of platelet-activating factor receptor controls retinal neovascularization. Cell Discov 2016; 2:16017. [PMID: 27462464 PMCID: PMC4941644 DOI: 10.1038/celldisc.2016.17] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 05/11/2016] [Indexed: 02/08/2023] Open
Abstract
Platelet-activating factor (PAF) is a pleiotropic phospholipid with proinflammatory, procoagulant and angiogenic actions on the vasculature. We and others have reported the presence of PAF receptor (Ptafr) at intracellular sites such as the nucleus. However, mechanisms of localization and physiologic functions of intracellular Ptafr remain poorly understood. We hereby identify the importance of C-terminal motif of the receptor and uncover novel roles of Rab11a GTPase and importin-5 in nuclear translocation of Ptafr in primary human retinal microvascular endothelial cells. Nuclear localization of Ptafr is independent of exogenous PAF stimulation as well as intracellular PAF biosynthesis. Moreover, nuclear Ptafr is responsible for the upregulation of unique set of growth factors, including vascular endothelial growth factor, in vitro and ex vivo. We further corroborate the intracrine PAF signaling, resulting in angiogenesis in vivo, using Ptafr antagonists with distinct plasma membrane permeability. Collectively, our findings show that nuclear Ptafr translocates in an agonist-independent manner, and distinctive functions of Ptafr based on its cellular localization point to another dimension needed for pharmacologic selectivity of drugs.
Collapse
Affiliation(s)
- Vikrant K Bhosle
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada; CHU Sainte Justine Hospital Research Centre, University of Montréal, Montréal, QC, Canada; Department of Ophthalmology, Research Centre of Hôpital Maisonneuve-Rosemont, University of Montréal, Montréal, QC, Canada
| | - José Carlos Rivera
- CHU Sainte Justine Hospital Research Centre, University of Montréal, Montréal, QC, Canada; Department of Ophthalmology, Research Centre of Hôpital Maisonneuve-Rosemont, University of Montréal, Montréal, QC, Canada
| | - Tianwei Ellen Zhou
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada; CHU Sainte Justine Hospital Research Centre, University of Montréal, Montréal, QC, Canada; Department of Ophthalmology, Research Centre of Hôpital Maisonneuve-Rosemont, University of Montréal, Montréal, QC, Canada; Department of Medicine, McGill University Health Center, Montreal, QC, Canada
| | - Samy Omri
- CHU Sainte Justine Hospital Research Centre, University of Montréal, Montréal, QC, Canada; Department of Ophthalmology, Research Centre of Hôpital Maisonneuve-Rosemont, University of Montréal, Montréal, QC, Canada
| | - Melanie Sanchez
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada; CHU Sainte Justine Hospital Research Centre, University of Montréal, Montréal, QC, Canada; Department of Ophthalmology, Research Centre of Hôpital Maisonneuve-Rosemont, University of Montréal, Montréal, QC, Canada
| | - David Hamel
- CHU Sainte Justine Hospital Research Centre, University of Montréal, Montréal, QC, Canada; Department of Pharmacology, University of Montréal, Montréal, QC, Canada
| | - Tang Zhu
- CHU Sainte Justine Hospital Research Centre, University of Montréal , Montréal, QC, Canada
| | - Raphael Rouget
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada; CHU Sainte Justine Hospital Research Centre, University of Montréal, Montréal, QC, Canada
| | - Areej Al Rabea
- Experimental Surgery, Montreal General Hospital, McGill University , Montréal, QC, Canada
| | - Xin Hou
- CHU Sainte Justine Hospital Research Centre, University of Montréal , Montréal, QC, Canada
| | - Isabelle Lahaie
- CHU Sainte Justine Hospital Research Centre, University of Montréal, Montréal, QC, Canada; Department of Ophthalmology, Research Centre of Hôpital Maisonneuve-Rosemont, University of Montréal, Montréal, QC, Canada
| | - Alfredo Ribeiro-da-Silva
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada; Alan Edwards Centre for Research on Pain, McGill University, Montréal, QC, Canada; Department of Anatomy and Cell Biology, McGill University, Montréal, QC, Canada
| | - Sylvain Chemtob
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC, Canada; CHU Sainte Justine Hospital Research Centre, University of Montréal, Montréal, QC, Canada; Department of Ophthalmology, Research Centre of Hôpital Maisonneuve-Rosemont, University of Montréal, Montréal, QC, Canada; Department of Pharmacology, University of Montréal, Montréal, QC, Canada; Departments of Pediatrics and Ophthalmology, Faculty of Medicine, University of Montréal, Montréal, QC, Canada
| |
Collapse
|
36
|
Gautier C, Barrier-Battut I, Guénon I, Goux D, Delalande C, Bouraïma-Lelong H. Implication of the estrogen receptors GPER, ESR1, ESR2 in post-testicular maturations of equine spermatozoa. Gen Comp Endocrinol 2016; 233:100-108. [PMID: 27222348 DOI: 10.1016/j.ygcen.2016.05.022] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 05/18/2016] [Accepted: 05/20/2016] [Indexed: 11/26/2022]
Abstract
Estrogen receptors ESR1, ESR2 and GPER are present on mature ejaculated horse spermatozoa, suggesting these cells as putative targets for estrogens. Indeed, spermatozoa are exposed to high level of estrogens during the transit in the male and female genital tracts but their roles are not investigated. So, we evaluated in vitro the role of 17β-estradiol during post-testicular maturations: regulation of motility, capacitation and acrosome reaction. Moreover according to the pseudo-seasonal breeder status of the stallion, we analyzed the putative seasonal variations in the presence of ESRs in spermatozoa. We showed that ESRs are more present on stallion sperm during the breeding season. We showed that capacitation and acrosome reaction are independent of estradiol action in horse. Estradiol can weakly modulate the motility and this effect is strictly associated with GPER and not with ESR1 and ESR2. The subcellular localization of GPER in the neck on stallion sperm is coherent with this effect. It seems that estrogens are not major regulators of sperm maturations associated to mare genital tract, so they could act during the epididymal maturations.
Collapse
Affiliation(s)
- Camille Gautier
- Normandie Univ, France; UNICAEN, EA2608, OeReCa, F-14032 Caen, France; USC-INRA 1377, F-14032 Caen, France
| | | | - Isabelle Guénon
- Normandie Univ, France; UNICAEN, EA2608, OeReCa, F-14032 Caen, France; USC-INRA 1377, F-14032 Caen, France
| | - Didier Goux
- Normandie Univ, France; UNICAEN, CMABIO, F-14032 Caen, France
| | - Christelle Delalande
- Normandie Univ, France; UNICAEN, EA2608, OeReCa, F-14032 Caen, France; USC-INRA 1377, F-14032 Caen, France
| | - Hélène Bouraïma-Lelong
- Normandie Univ, France; UNICAEN, EA2608, OeReCa, F-14032 Caen, France; USC-INRA 1377, F-14032 Caen, France.
| |
Collapse
|
37
|
Cattaneo F, Parisi M, Fioretti T, Sarnataro D, Esposito G, Ammendola R. Nuclear localization of Formyl-Peptide Receptor 2 in human cancer cells. Arch Biochem Biophys 2016; 603:10-9. [PMID: 27177968 DOI: 10.1016/j.abb.2016.05.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/06/2016] [Accepted: 05/06/2016] [Indexed: 12/20/2022]
Abstract
Current models of G protein-coupled receptors (GPCRs) signaling describe binding of external agonists to cell surface receptors which, in turn, trigger several biological responses. New paradigms indicate that GPCRs localize to and signal at the nucleus, thus regulating distinct signaling cascades. The formyl-peptide receptor FPR2 belongs to the GPCR super-family and is coupled to PTX-sensitive Gi proteins. We show by western blot analysis, immunofluorescence experiments and radioligand binding assays that FPR2 is expressed at nuclear level in CaLu-6 and AGS cells. Nuclear FPR2 is a functional receptor, since it participates in intra-nuclear signaling, as assessed by decreased G protein-FPR2 association and enhanced ERK2, c-Jun and c-Myc phosphorylation upon stimulation of intact nuclei with the FPR2 agonist, WKYMVm. We analyzed FPR2 sequence for the search of a nuclear localization sequence (NLS) and we found a stretch of basic aminoacids (227-KIHKK-231) in the third cytoplasmic loop of the receptor. We performed single (K230A) and multiple (H229A/K230A/K231A) mutagenesis of NLS. The constructs were individually overexpressed in HEK293 cells and immunofluorescence and western blot analysis showed that nuclear localization or translocation of FPR2 depends on the integrity of the H(229) and K(231) residues within the NLS.
Collapse
Affiliation(s)
- Fabio Cattaneo
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
| | - Melania Parisi
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy
| | - Tiziana Fioretti
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy; IRCCS SDN, Via E. Gianturco 113, Naples 80143, Italy
| | - Daniela Sarnataro
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy; CEINGE-Biotecnologie Avanzate s.c.a.r.l., Via G. Salvatore 486, Naples 80145, Italy
| | - Gabriella Esposito
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy; CEINGE-Biotecnologie Avanzate s.c.a.r.l., Via G. Salvatore 486, Naples 80145, Italy
| | - Rosario Ammendola
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Via S. Pansini 5, Naples 80131, Italy.
| |
Collapse
|
38
|
Topogenesis and cell surface trafficking of GPR34 are facilitated by positive-inside rule that effects through a tri-basic motif in the first intracellular loop. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1534-51. [PMID: 27086875 DOI: 10.1016/j.bbamcr.2016.04.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/12/2016] [Revised: 04/06/2016] [Accepted: 04/11/2016] [Indexed: 11/21/2022]
Abstract
Protein folding, topogenesis and intracellular targeting of G protein-coupled receptors (GPCRs) must be precisely coordinated to ensure correct receptor localization. To elucidate how different steps of GPCR biosynthesis work together, we investigated the process of membrane topology determination and how it relates to the acquisition of cell surface trafficking competence in human GPR34. By monitoring a fused FLAG-tag and a conformation-sensitive native epitope during the expression of GPR34 mutant panel, a tri-basic motif in the first intracellular loop was identified as the key topogenic signal that dictates the orientation of transmembrane domain-1 (TM1). Charge disruption of the motif perturbed topogenic processes and resulted in the conformational epitope loss, post-translational processing alteration, and trafficking arrest in the Golgi. The placement of a cleavable N-terminal signal sequence as a surrogate topogenic determinant overcame the effects of tri-basic motif mutations and rectified the TM1 orientation; thereby restored the conformational epitope, post-translational modifications, and cell surface trafficking altogether. Progressive N-tail truncation and site-directed mutagenesis revealed that a proline-rich segment of the N-tail and all four cysteines individually located in the four separate extracellular regions must simultaneously reside in the ER lumen to muster the conformational epitope. Oxidation of all four cysteines was necessary for the epitope formation, but the cysteine residues themselves were not required for the trafficking event. The underlying biochemical properties of the conformational epitope was therefore the key to understand mechanistic processes propelled by positive-inside rule that simultaneously regulate the topogenesis and intracellular trafficking of GPR34.
Collapse
|
39
|
Massart R, Mignon V, Stanic J, Munoz-Tello P, Becker JAJ, Kieffer BL, Darmon M, Sokoloff P, Diaz J. Developmental and adult expression patterns of the G-protein-coupled receptor GPR88 in the rat: Establishment of a dual nuclear-cytoplasmic localization. J Comp Neurol 2016; 524:2776-802. [PMID: 26918661 DOI: 10.1002/cne.23991] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 02/22/2016] [Accepted: 02/23/2016] [Indexed: 01/31/2023]
Abstract
GPR88 is a neuronal cerebral orphan G-protein-coupled receptor (GPCR) that has been linked to various psychiatric disorders. However, no extensive description of its localization has been provided so far. Here, we investigate the spatiotemporal expression of the GPR88 in prenatal and postnatal rat tissues by using in situ hybridization and immunohistochemistry. GPR88 protein was initially detected at embryonic day 16 (E16) in the striatal primordium. From E16-E20 to adulthood, the highest expression levels of both protein and mRNA were observed in striatum, olfactory tubercle, nucleus accumbens, amygdala, and neocortex, whereas in spinal cord, pons, and medulla GPR88 expression remains discrete. We observed an intracellular redistribution of GPR88 during cortical lamination. In the cortical plate of the developing cortex, GPR88 presents a classical GPCR plasma membrane/cytoplasmic localization that shifts, on the day of birth, to nuclei of neurons progressively settling in layers V to II. This intranuclear localization remains throughout adulthood and was also detected in monkey and human cortex as well as in the amygdala and hypothalamus of rats. Apart from the central nervous system, GPR88 was transiently expressed at high levels in peripheral tissues, including adrenal cortex (E16-E21) and cochlear ganglia (E19-P3), and also at moderate levels in retina (E18-E19) and spleen (E21-P7). The description of the GPR88 anatomical expression pattern may provide precious functional insights into this novel receptor. Furthermore, the GRP88 nuclear localization suggests nonclassical GPCR modes of action of the protein that could be relevant for cortical development and psychiatric disorders. J. Comp. Neurol. 524:2776-2802, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Renaud Massart
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France.,Neurology-Psychiatry Department, Pierre Fabre Research Institute, 81100, Castres, France
| | - Virginie Mignon
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France
| | - Jennifer Stanic
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France
| | - Paola Munoz-Tello
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France
| | - Jerôme A J Becker
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, INSERM, 67400, Illkirch-Graffenstaden, France
| | - Brigitte L Kieffer
- Institut de Génétique et de Biologie Moléculaire et Cellulaire, Université de Strasbourg, CNRS, INSERM, 67400, Illkirch-Graffenstaden, France
| | - Michèle Darmon
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France
| | - Pierre Sokoloff
- Neurology-Psychiatry Department, Pierre Fabre Research Institute, 81100, Castres, France
| | - Jorge Diaz
- INSERM UMR894, Centre de Psychiatrie et Neurosciences, Université Paris Descartes, 75014, Paris, France.,Université Paris Descartes, Sorbonne Paris Cité, 75006, Paris, France
| |
Collapse
|
40
|
Abstract
Although convention dictates that G protein-coupled receptors localize to and signal at the plasma membrane, accumulating evidence suggests that G protein-coupled receptors localize to and signal at intracellular membranes, most notably the nucleus. In fact, there is now significant evidence indicating that endogenous alpha-1 adrenergic receptors (α1-ARs) localize to and signal at the nuclei in adult cardiac myocytes. Cumulatively, the data suggest that α1-ARs localize to the inner nuclear membrane, activate intranuclear signaling, and regulate physiologic function in adult cardiac myocytes. Although α1-ARs signal through Gαq, unlike other Gq-coupled receptors, α1-ARs mediate important cardioprotective functions including adaptive/physiologic hypertrophy, protection from cell death (survival signaling), positive inotropy, and preconditioning. Also unlike other Gq-coupled receptors, most, if not all, functional α1-ARs localize to the nuclei in adult cardiac myocytes, as opposed to the sarcolemma. Together, α1-AR nuclear localization and cardioprotection might suggest a novel model for compartmentalization of Gq-coupled receptor signaling in which nuclear Gq-coupled receptor signaling is cardioprotective.
Collapse
|
41
|
Herrera-Luna CV, Scarlet D, Walter I, Aurich C. Effect of stallion age on the expression of LH and FSH receptors and aromatase P450 in equine male reproductive tissues. Reprod Fertil Dev 2016; 28:2016-2026. [DOI: 10.1071/rd15027] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Accepted: 06/06/2015] [Indexed: 11/23/2022] Open
Abstract
The aim of the present study was to characterise receptors for LH and FSH (LHR and FSHR, respectively) and aromatase in epididymal and testicular tissue from stallions of different ages (prepubertal, young, mature and old). Gene and protein expression were assessed by real-time quantitative polymerase chain reaction (real-time qPCR), immunohistochemistry and multiple immunofluorescence labelling. There were no differences in LHR mRNA expression in epididymal and testicular parenchyma in stallions of different age. In contrast, expression of FSHR and CYP19A1 in caput, corpus and cauda epididymis and in testicular parenchyma increased with age (P < 0.001). Immunolabelling for LHR, FSHR and aromatase was influenced by puberty. In postpubertal stallions, positive staining for LHR and aromatase was detected in Leydig cells, whereas protein expression of FSHR was present in Sertoli cells and primary spermatocytes. In prepubertal colts, staining for LHR, FSHR and aromatase was detected in seminiferous tubules. In epididymal tissue, aromatase was present in the cauda epididymis only, regardless of age. In conclusion, the results highlight the significance of gonadotropin action and oestrogen production for the maturation of male reproductive tissue in the horse. The presence of FSHR in the seminiferous tubules suggests effects of FSH on spermatogenesis in this species. The importance of oestrogen production for maintenance of testicular function in stallions was confirmed.
Collapse
|
42
|
Ronda AC, Boland RL. Intracellular Distribution and Involvement of GPR30 in the Actions of E2 on C2C12 Cells. J Cell Biochem 2015; 117:793-805. [PMID: 26359786 DOI: 10.1002/jcb.25369] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 09/09/2015] [Indexed: 12/28/2022]
Abstract
G-protein-coupled receptor 30 (GPR30) is an estrogen receptor that initiates several rapid, non-genomic signaling events triggered by E2. GPR30 has recently been identified in C2C12 cells; however, little is known about the intracelular distribution and its role in C2C12 myoblasts and myotubes. By western blotting and immunohistochemistry, we evidenced expression of GPR30. While in C2C12 myoblasts, the receptor was present in nucleus, mitochondria, and endoplasmic reticulum, in C2C12 myotubes, it was additionally found in cytoplasm. Using trypan blue uptake assay to determine cellular death and fluorescent microscopy to evaluate picnotic nuclei and mitochondrial distribution, we demonstated that treatment of C2C12 myoblasts with G1 (GPR30 agonist) did not protect the cells against apoptosis induced by H2O2 as E2. However, when G15 (GPR30 antagonist) was used, E2 could not prevent the damage caused by the oxidative stress. Further, some of the molecular mechanisms involved were investigated by wertern blot assays. Thus, E2 was able to induce AKT phosphorylation in apoptotic conditions and ERK phosphorylation in proliferating C2C12 cells but not when the cultures were incubated with G15. Additionally, using G15 antagonist we have found that GPR30 participates in the myogenin expression and creatine kinase activity stimulated by E2 in the first steps of C2C12 differentiation. Althogether these findings provide evidences showing that GPR30 is expressed in diverse intracellular compartments in undifferentiated and differentiated C2C12 cells and mediates E2 actions.
Collapse
Affiliation(s)
- Ana C Ronda
- Instituto de Investigaciones Biol, ó, gicas y Biom, é, dicas del Sur (CONICET) / Universidad Nacional del Sur, San Juan 670, Bah, í, a Blanca, 8000, Argentina
| | - Ricardo L Boland
- Instituto de Investigaciones Biol, ó, gicas y Biom, é, dicas del Sur (CONICET) / Universidad Nacional del Sur, San Juan 670, Bah, í, a Blanca, 8000, Argentina
| |
Collapse
|
43
|
Joyal JS, Bhosle VK, Chemtob S. Subcellular G-protein coupled receptor signaling hints at greater therapeutic selectivity. Expert Opin Ther Targets 2015; 19:717-21. [DOI: 10.1517/14728222.2015.1042365] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
44
|
Kim SO, Markosyan N, Pepe GJ, Duffy DM. Estrogen promotes luteolysis by redistributing prostaglandin F2α receptors within primate luteal cells. Reproduction 2015; 149:453-64. [PMID: 25687410 DOI: 10.1530/rep-14-0412] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Prostaglandin F2α (PGF2α) has been proposed as a functional luteolysin in primates. However, administration of PGF2α or prostaglandin synthesis inhibitors in vivo both initiate luteolysis. These contradictory findings may reflect changes in PGF2α receptors (PTGFRs) or responsiveness to PGF2α at a critical point during the life span of the corpus luteum. The current study addressed this question using ovarian cells and tissues from female cynomolgus monkeys and luteinizing granulosa cells from healthy women undergoing follicle aspiration. PTGFRs were present in the cytoplasm of monkey granulosa cells, while PTGFRs were localized in the perinuclear region of large, granulosa-derived monkey luteal cells by mid-late luteal phase. A PTGFR agonist decreased progesterone production in luteal cells obtained at mid-late and late luteal phases, but did not decrease progesterone production by granulosa cells or luteal cells from younger corpora lutea. These findings are consistent with a role for perinuclear PTGFRs in functional luteolysis. This concept was explored using human luteinizing granulosa cells maintained in vitro as a model for luteal cell differentiation. In these cells, PTGFRs relocated from the cytoplasm to the perinuclear area in an estrogen- and estrogen receptor-dependent manner. Similar to our findings with monkey luteal cells, human luteinizing granulosa cells with perinuclear PTGFRs responded to a PTGFR agonist with decreased progesterone production. These data support the concept that PTGFR stimulation promotes functional luteolysis only when PTGFRs are located in the perinuclear region. Estrogen receptor-mediated relocation of PTGFRs within luteal cells may be a necessary step in the initiation of luteolysis in primates.
Collapse
Affiliation(s)
- Soon Ok Kim
- Department of Physiological SciencesEastern Virginia Medical School, Norfolk, Virginia 23501, USA
| | - Nune Markosyan
- Department of Physiological SciencesEastern Virginia Medical School, Norfolk, Virginia 23501, USA
| | - Gerald J Pepe
- Department of Physiological SciencesEastern Virginia Medical School, Norfolk, Virginia 23501, USA
| | - Diane M Duffy
- Department of Physiological SciencesEastern Virginia Medical School, Norfolk, Virginia 23501, USA
| |
Collapse
|
45
|
Zhang YF, Xu QX, Liao LD, Xu XE, Wu JY, Wu ZY, Shen JH, Li EM, Xu LY. Association of mu-opioid receptor expression with lymph node metastasis in esophageal squamous cell carcinoma. Dis Esophagus 2015; 28:196-203. [PMID: 24428760 DOI: 10.1111/dote.12165] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The mu-opioid receptor (MOR), a membrane-bound G protein-coupled receptor, is the main target for opioids in the nervous system. MOR1 has been found in several types of cancer cells and reported to be involved in tumor progression and metastasis. However, the expression and clinical significance of MOR1 in esophageal squamous cell carcinoma (ESCC) remain unclear. In our study, the expression of MOR1 was confirmed in ESCC cell lines (KYSE180, KYSE150, and EC109) by Western blot. MOR1 was also detected on tissue microarrays of ESCC samples in 239 cases using immunohistochemical staining. We found that MOR1 was mainly located in the cytoplasm and occasionally occurred in the membrane or nucleus of ESCC cells. Moreover, results indicated that MOR1 expression in the cytoplasm was associated with lymph node metastasis (R = 0.164, P = 0.008, Kendall's tau-b-test). No more associations were found between MOR1 expression status and other clinical parameters. However, no statistical significant differences were found between MOR1 expression in the cytoplasm, nucleus/membrane, and the overall survival of ESCC patients (P = 0.848; P = 0.167; P = 0.428, respectively, log-rank test). Our results suggest that the cytoplasmic MOR1 may be a high-risk factor for lymph node metastasis of ESCC patients. We also hypothesize that MOR1 agonists used in ESCC patients should be prudent, and opioid receptor antagonists may be novel therapeutic drugs for ESCC patients.
Collapse
Affiliation(s)
- Y-F Zhang
- Institute of Oncologic Pathology, Shantou University Medical College, Shantou, China
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Sellers K, Raval P, Srivastava DP. Molecular signature of rapid estrogen regulation of synaptic connectivity and cognition. Front Neuroendocrinol 2015; 36:72-89. [PMID: 25159586 DOI: 10.1016/j.yfrne.2014.08.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/11/2014] [Accepted: 08/14/2014] [Indexed: 12/14/2022]
Abstract
There is now a growing appreciation that estrogens are capable of rapidly activating a number of signaling cascades within the central nervous system. In addition, there are an increasing number of studies reporting that 17β-estradiol, the major biologically active estrogen, can modulate cognition within a rapid time frame. Here we review recent studies that have begun to uncover the molecular and cellular framework which contributes to estrogens ability to rapidly modulate cognition. We first describe the mechanisms by which estrogen receptors (ERs) can couple to intracellular signaling cascades, either directly, or via the transactivation of other receptors. Subsequently, we review the evidence that estrogen can rapidly modulate both neuronal function and structure in the hippocampus and the cortex. Finally, we will discuss how estrogens may influence cognitive function through the modulation of neuronal structure, and the implications this may have on the treatment of a range of brain disorders.
Collapse
Affiliation(s)
- Katherine Sellers
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK
| | - Pooja Raval
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK
| | - Deepak P Srivastava
- Department of Basic and Clinical Neuroscience, The James Black Centre, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London SE5 9NU, UK.
| |
Collapse
|
47
|
Bhosle VK, Gobeil F, Rivera JC, Ribeiro-da-Silva A, Chemtob S. High resolution imaging and function of nuclear G protein-coupled receptors (GPCRs). Methods Mol Biol 2015; 1234:81-97. [PMID: 25304350 DOI: 10.1007/978-1-4939-1755-6_8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The traditional view of G protein-coupled receptors (GPCRs) being inactivated upon their internalization has been repeatedly challenged in recent years. GPCRs, in addition to forming the largest family of cell surface receptors, can also be found on intracellular membranes such as nuclear membranes. Since the first experimental evidence of GPCRs at the nucleus in the early 1990s, approximately 30 different GPCRs have been localized at the nucleus by independent research groups, including ours. In this chapter, we describe several techniques commonly used for immuno-detection of nuclear GPCRs focusing on subcellular fractionation of proteins based on their localization and transmission electron microscopy (TEM) using primary cultured cells as well as tissue sections. We also describe the use of confocal microscopy to study nuclear calcium currents, which can further affect downstream events such as gene transcription, nuclear envelope breakdown, or its reconstruction and nucleocytoplasmic protein transport.
Collapse
Affiliation(s)
- Vikrant K Bhosle
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada
| | | | | | | | | |
Collapse
|
48
|
Jong YJI, Sergin I, Purgert CA, O'Malley KL. Location-dependent signaling of the group 1 metabotropic glutamate receptor mGlu5. Mol Pharmacol 2014; 86:774-85. [PMID: 25326002 PMCID: PMC4244594 DOI: 10.1124/mol.114.094763] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 10/17/2014] [Indexed: 12/20/2022] Open
Abstract
Although G protein-coupled receptors are primarily known for converting extracellular signals into intracellular responses, some receptors, such as the group 1 metabotropic glutamate receptor, mGlu5, are also localized on intracellular membranes where they can mediate both overlapping and unique signaling effects. Thus, besides "ligand bias," whereby a receptor's signaling modality can shift from G protein dependence to independence, canonical mGlu5 receptor signaling can also be influenced by "location bias" (i.e., the particular membrane and/or cell type from which it signals). Because mGlu5 receptors play important roles in both normal development and in disorders such as Fragile X syndrome, autism, epilepsy, addiction, anxiety, schizophrenia, pain, dyskinesias, and melanoma, a large number of drugs are being developed to allosterically target this receptor. Therefore, it is critical to understand how such drugs might be affecting mGlu5 receptor function on different membranes and in different brain regions. Further elucidation of the site(s) of action of these drugs may determine which signal pathways mediate therapeutic efficacy.
Collapse
Affiliation(s)
- Yuh-Jiin I Jong
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| | - Ismail Sergin
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| | - Carolyn A Purgert
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| | - Karen L O'Malley
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri
| |
Collapse
|
49
|
Di Benedetto A, Sun L, Zambonin CG, Tamma R, Nico B, Calvano CD, Colaianni G, Ji Y, Mori G, Grano M, Lu P, Colucci S, Yuen T, New MI, Zallone A, Zaidi M. Osteoblast regulation via ligand-activated nuclear trafficking of the oxytocin receptor. Proc Natl Acad Sci U S A 2014; 111:16502-7. [PMID: 25378700 PMCID: PMC4246276 DOI: 10.1073/pnas.1419349111] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
We report that oxytocin (Oxt) receptors (Oxtrs), on stimulation by the ligand Oxt, translocate into the nucleus of osteoblasts, implicating this process in the action of Oxt on osteoblast maturation. Sequential immunocytochemistry of intact cells or isolated nucleoplasts stripped of the outer nuclear membrane showed progressive nuclear localization of the Oxtr; this nuclear translocation was confirmed by monitoring the movement of Oxtr-EGFP as well as by immunogold labeling. Nuclear Oxtr localization was conclusively shown by Western immunoblotting and MS of nuclear lysate proteins. We found that the passage of Oxtrs into the nucleus was facilitated by successive interactions with β-arrestins (Arrbs), the small GTPase Rab5, importin-β (Kpnb1), and transportin-1 (Tnpo1). siRNA-mediated knockdown of Arrb1, Arrb2, or Tnpo1 abrogated Oxt-induced expression of the osteoblast differentiation genes osterix (Sp7), Atf4, bone sialoprotein (Ibsp), and osteocalcin (Bglap) without affecting Erk phosphorylation. Likewise and again, without affecting pErk, inhibiting Arrb recruitment by mutating Ser rich clusters of the nuclear localization signal to Ala abolished nuclear import and Oxtr-induced gene expression. These studies define a previously unidentified mechanism for Oxtr action on bone and open possibilities for direct transcriptional modulation by nuclear G protein-coupled receptors.
Collapse
Affiliation(s)
- Adriana Di Benedetto
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy; Department of Clinical and Experimental Medicine, University of Foggia, Foggia 71122, Italy
| | - Li Sun
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | - Carlo G Zambonin
- Department of Chemistry, University of Bari Aldo Moro, Bari 70126, Italy; and
| | - Roberto Tamma
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Beatrice Nico
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Cosima D Calvano
- Department of Chemistry, University of Bari Aldo Moro, Bari 70126, Italy; and
| | - Graziana Colaianni
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Yaoting Ji
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | - Giorgio Mori
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia 71122, Italy
| | - Maria Grano
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Ping Lu
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | - Silvia Colucci
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Tony Yuen
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029
| | | | - Alberta Zallone
- Department of Basic Medical Science, Neurosciences and Sensory Organs, University of Bari Aldo Moro Medical School, Bari 70126, Italy
| | - Mone Zaidi
- Mount Sinai Bone Program and Department of Medicine, Mount Sinai School of Medicine, New York, NY 10029; Structural and Chemical Biology, Mount Sinai School of Medicine, New York, NY 10029
| |
Collapse
|
50
|
Barbarin A, Séité P, Godet J, Bensalma S, Muller JM, Chadéneau C. Atypical nuclear localization of VIP receptors in glioma cell lines and patients. Biochem Biophys Res Commun 2014; 454:524-30. [DOI: 10.1016/j.bbrc.2014.10.113] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 10/23/2014] [Indexed: 12/27/2022]
|