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
- Program in Molecular Biology, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Gael Cagnone
- Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Emmanuel Bajon
- Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Prabhas Chaudhari
- Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- Department of Experimental Medicine, McGill University, Montréal, Canada
| | - Monir Modaresinejad
- Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- Program in Biomedical Sciences, Faculty of Medicine, Université de Montréal, Montreal, Canada
| | - Pierre Hardy
- Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Damien Maggiorani
- Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Canada
| | - Christiane Quiniou
- Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Jean-Sébastien Joyal
- Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Canada
| | - Christian Beauséjour
- Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
| | - Sylvain Chemtob
- Program in Molecular Biology, Faculty of Medicine, Université de Montréal, Montreal, Canada
- Research Center of Centre Hospitalier Universitaire Sainte-Justine, Montreal, Canada
- Department of Pharmacology, Université de Montréal, Montreal, Canada
| |
Collapse
|
2
|
Li H, Peng MX, Yang RX, Chen JX, Wang YM, Wang PX, Hu YH, Pan DY, Liu PQ, Lu J. SNX3 mediates heart failure by interacting with HMGB1 and subsequently facilitating its nuclear-cytoplasmic translocation. Acta Pharmacol Sin 2025; 46:964-975. [PMID: 39753981 PMCID: PMC11950316 DOI: 10.1038/s41401-024-01436-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Accepted: 11/17/2024] [Indexed: 03/17/2025]
Abstract
Sorting nexins (SNXs) as the key regulators of sorting cargo proteins are involved in diverse diseases. SNXs can form the specific reverse vesicle transport complex (SNXs-retromer) with vacuolar protein sortings (VPSs) to sort and modulate recovery and degradation of cargo proteins. Our previous study has shown that SNX3-retromer promotes both STAT3 activation and nuclear translocation in cardiomyocytes, suggesting that SNX3 might be a critical regulator in the heart. In this study we investigated the role of SNX3 in the development of pathological cardiac hypertrophy and heart failure. We generated abdominal aortic constriction (AAC) rat model and transverse aortic constriction (TAC) mouse model; hypertrophic neonatal rat cardiomyocytes (NRCMs) were induced by exposure to isoproterenol (10 μM). We showed that the expression of SNX3 was significantly upregulated in ISO-treated NRCMs and in the failing heart of AAC rats. Overexpression of SNX3 by intramyocardial injection of Ad-SNX3 induced heart failure in rats, and increased the susceptibility of NRCMs to ISO-induced myocardial injury in vitro. In contrast, conditional knockout of SNX3 in cardiac tissue in mice rescued the detrimental heart function in TAC mice, and knockdown of SNX3 protected against ISO-induced injury in NRCMs and AAC rats. We then conducted immunoprecipitation-based mass spectrometry and localized surface plasmon resonance, and demonstrated a direct interaction between SNX3-retromer and high mobility group box 1 (HMGB1), which mediated the efflux of nuclear HMGB1. Moreover, overexpression of HMGB1 in NRCMs inhibited the pro-hypertrophic effects of SNX3, whereas knockdown of HMGB1 abolished the protective effect of SNX3-deficiency. These results suggest that HMGB1 might be a direct cargo protein of SNX3-retromer, and its interaction with SNX3 promotes its efflux from the nucleus, leading to the pathological development of heart failure.
Collapse
Affiliation(s)
- Hong Li
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- The Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Ming-Xia Peng
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Rui-Xue Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
- The Research Center of Basic Integrative Medicine, School of Basic Medical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, 510006, China
| | - Jian-Xing Chen
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yue-Mei Wang
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Pan-Xia Wang
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Yue-Huai Hu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Di-Yi Pan
- Department of Pharmacy, The Third Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, 510378, China
| | - Pei-Qing Liu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| | - Jing Lu
- National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Guangdong Provincial Key Laboratory of New Drug Design and Evaluation, Guangdong Province Engineering Laboratory for Druggability and New Drug Evaluation, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China.
| |
Collapse
|
3
|
Calebiro D, Miljus T, O'Brien S. Endomembrane GPCR signaling: 15 years on, the quest continues. Trends Biochem Sci 2025; 50:46-60. [PMID: 39532582 DOI: 10.1016/j.tibs.2024.10.006] [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: 07/08/2024] [Revised: 10/13/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
G-protein-coupled receptors (GPCRs) are the largest family of cell receptors. They mediate the effects of a multitude of endogenous and exogenous cues, are deeply involved in human physiology and disease, and are major pharmacological targets. Whereas GPCRs were long thought to signal exclusively at the plasma membrane, research over the past 15 years has revealed that they also signal via classical G-protein-mediated pathways on membranes of intracellular organelles such as endosomes and the Golgi complex. This review provides an overview of recent advances and emerging concepts related to endomembrane GPCR signaling, as well as ongoing research aimed at a better understanding of its mechanisms, physiological relevance, and potential therapeutic applications.
Collapse
Affiliation(s)
- Davide Calebiro
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK.
| | - Tamara Miljus
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| | - Shannon O'Brien
- Department of Metabolism and Systems Science, College of Medicine and Health, University of Birmingham, Birmingham, UK; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
| |
Collapse
|
4
|
Malamos P, Kalyvianaki K, Panagiotopoulos AA, Vogiatzoglou AP, Tsikalaki AA, Katifori A, Polioudaki H, Darivianaki MN, Theodoropoulos PA, Panagiotidis CA, Notas G, Castanas E, Kampa M. Nuclear translocation of the membrane oxoeicosanoid/androgen receptor, OXER1: Possible mechanisms involved. Mol Cell Endocrinol 2024; 594:112357. [PMID: 39236798 DOI: 10.1016/j.mce.2024.112357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/09/2024] [Accepted: 09/02/2024] [Indexed: 09/07/2024]
Abstract
OXER1, the receptor for the arachidonic acid metabolite 5-οxo-eicosatetraenoic acid (5-oxo-ETE), has been reported to also bind and mediate the membrane-initiated actions of androgens. Indeed, androgens antagonize the 5-oxo-ETE effects through OXER1, affecting a number of signaling pathways and inhibiting cancer cell proliferation and migration. OXER1, being a GPCR, was classically described to be localized in the plasma membrane. However, for numerous GPCRs, there is now strong evidence that they can be also found in other cellular compartments, including the nucleus. The aim of the present work was to investigate OXER1's possible localization in the nucleus and identify the mechanism(s) involved. For this purpose, we verified OXER1's nuclear presence by immunofluorescence and western blot, in whole cells and nuclei of two different prostate cancer cell lines (DU-145 and LNCaP) and in CHO cells transfected with a GFP labelled OXER1, both in untreated and OXER1 ligands' treated cells. Mutated, OXER1-tGFP expressing, CHO cells were used to verify that OXER1 agonist (5-oxo-ETE) binding is necessary for OXER1 nuclear translocation. NLS sequences were in silico identified, and a specific inhibitor, as well as, specific importins' siRNAs were also utilized to explore the mechanism involved. Moreover, we examined the role of palmitoylation in OXER1 nuclear translocation by in silico identifying possible palmitoylation sites and using a palmitoylation inhibitor. Our results clearly show that OXER1 can be localized in the nucleus, in an agonist-dependent manner, that is inhibited by androgens. We also provide evidence for two possible mechanisms for its nuclear trafficking, that involve receptor palmitoylation and importin-mediated cytoplasmic-nuclear transport. In our knowledge, it is the first time that a membrane androgen receptor is identified into the nucleus, suggesting an alternative, more direct, mode of action, involving nuclear mechanisms. Therefore, our findings provide new insights on androgen-mediated actions and androgen-lipid interactions, and reveal new possible therapeutic targets, not only for cancer, but also for other pathological conditions in which OXER1 may have an important role.
Collapse
Affiliation(s)
- Panagiotis Malamos
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 70013, Heraklion, Crete, Greece
| | - Konstantina Kalyvianaki
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 70013, Heraklion, Crete, Greece
| | - Athanasios A Panagiotopoulos
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 70013, Heraklion, Crete, Greece
| | - Amalia P Vogiatzoglou
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 70013, Heraklion, Crete, Greece
| | - Athanasia Artemis Tsikalaki
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 70013, Heraklion, Crete, Greece
| | - Anastasia Katifori
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 70013, Heraklion, Crete, Greece
| | - Hara Polioudaki
- Laboratory of Biochemistry, School of Medicine, University of Crete, Voutes Campus, 70013, Heraklion, Crete, Greece
| | - Maria N Darivianaki
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - Panayiotis A Theodoropoulos
- Laboratory of Biochemistry, School of Medicine, University of Crete, Voutes Campus, 70013, Heraklion, Crete, Greece
| | - Christos A Panagiotidis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, Thessaloniki, 54124, Greece
| | - George Notas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 70013, Heraklion, Crete, Greece
| | - Elias Castanas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 70013, Heraklion, Crete, Greece
| | - Marilena Kampa
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Voutes Campus, 70013, Heraklion, Crete, Greece.
| |
Collapse
|
5
|
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
|
6
|
Golderman V, Gofrit SG, Ivashko-Pachima Y, Gozes I, Chapman J, Shavit-Stein E. The thrombin receptor (PAR1) is associated with microtubules, mitosis and process formation in glioma cells. Heliyon 2024; 10:e33329. [PMID: 39027436 PMCID: PMC11254606 DOI: 10.1016/j.heliyon.2024.e33329] [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: 02/27/2023] [Revised: 05/31/2024] [Accepted: 06/19/2024] [Indexed: 07/20/2024] Open
Abstract
The cell surface protease-activated receptor 1 (PAR1) is overexpressed in glioblastoma multiforme (GBM). We studied the function and structure of intracellular microtubule (MT) and PAR1 in a tubulin-mediated process. We found that exposure to thrombin increased the percentage of proliferative, S, and M phases cells, affected morphology, and increased process elongation. PAR1 antagonist inversely affects these measures, increases tubulin end-binding protein 3 (EB3) mRNA expression in C6 cells, and reduces EB3 comet length, track length, and duration in neuroblastoma cells. In addition, immunofluorescence staining suggests that PAR1 is in close association with the MT α-tubulin and with coagulation cascade proteins during cell division stages. Our findings support PAR1 involvement in MT dynamics.
Collapse
Affiliation(s)
- Valery Golderman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, 52621, Israel
- Department of Neurology and Neurosurgery, Faculty of Medical and Health Sciences, Tel Aviv University, Tel-Aviv, 6997801, Israel
| | - Shany Guly Gofrit
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, 52621, Israel
| | - Yanina Ivashko-Pachima
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Illana Gozes
- Elton Laboratory for Molecular Neuroendocrinology, Department of Human Molecular Genetics and Biochemistry, Faculty of Medical and Health Sciences, Adams Super Center for Brain Studies and Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, 6997801, Israel
| | - Joab Chapman
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, 52621, Israel
- Department of Neurology and Neurosurgery, Faculty of Medical and Health Sciences, Tel Aviv University, Tel-Aviv, 6997801, Israel
- Robert and Martha Harden Chair in Mental and Neurological Diseases, Faculty of Medical and Health Sciences, Tel Aviv University, 6997801, Israel
| | - Efrat Shavit-Stein
- Department of Neurology, The Chaim Sheba Medical Center, Ramat Gan, 52621, Israel
- Department of Neurology and Neurosurgery, Faculty of Medical and Health Sciences, Tel Aviv University, Tel-Aviv, 6997801, Israel
- The TELEM Rubin Excellence in Biomedical Research Program, The Chaim Sheba Medical Center, Ramat Gan, 52621, Israel
| |
Collapse
|
7
|
Klauer MJ, Willette BKA, Tsvetanova NG. Functional diversification of cell signaling by GPCR localization. J Biol Chem 2024; 300:105668. [PMID: 38272232 PMCID: PMC10882132 DOI: 10.1016/j.jbc.2024.105668] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/13/2023] [Accepted: 11/29/2023] [Indexed: 01/27/2024] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of cell surface receptors and a critical class of regulators of mammalian physiology. Also known as seven transmembrane receptors (7TMs), GPCRs are ubiquitously expressed and versatile, detecting a diverse set of endogenous stimuli, including odorants, neurotransmitters, hormones, peptides, and lipids. Accordingly, GPCRs have emerged as the largest class of drug targets, accounting for upward of 30% of all prescription drugs. The view that ligand-induced GPCR responses originate exclusively from the cell surface has evolved to reflect accumulating evidence that receptors can elicit additional waves of signaling from intracellular compartments. These events in turn shape unique cellular and physiological outcomes. Here, we discuss our current understanding of the roles and regulation of compartmentalized GPCR signaling.
Collapse
Affiliation(s)
- Matthew J Klauer
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Blair K A Willette
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA
| | - Nikoleta G Tsvetanova
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina, USA.
| |
Collapse
|
8
|
Mastos C, Xu X, Keen AC, Halls ML. Signalling of Adrenoceptors: Canonical Pathways and New Paradigms. Handb Exp Pharmacol 2024; 285:147-184. [PMID: 38227198 DOI: 10.1007/164_2023_704] [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] [Indexed: 01/17/2024]
Abstract
The concept of G protein-coupled receptors initially arose from studies of the β-adrenoceptor, adenylyl cyclase, and cAMP signalling pathway. Since then both canonical G protein-coupled receptor signalling pathways and emerging paradigms in receptor signalling have been defined by experiments focused on adrenoceptors. Here, we discuss the evidence for G protein coupling specificity of the nine adrenoceptor subtypes. We summarise the ability of each of the adrenoceptors to activate proximal signalling mediators including cAMP, calcium, mitogen-activated protein kinases, and protein kinase C pathways. Finally, we highlight the importance of precise spatial and temporal control of adrenoceptor signalling that is controlled by the localisation of receptors at intracellular membranes and in larger protein complexes.
Collapse
Affiliation(s)
- Chantel Mastos
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Xiaomeng Xu
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Alastair C Keen
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia
| | - Michelle L Halls
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.
| |
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
|
Fasciani I, Carli M, Petragnano F, Colaianni F, Aloisi G, Maggio R, Scarselli M, Rossi M. GPCRs in Intracellular Compartments: New Targets for Drug Discovery. Biomolecules 2022; 12:1343. [PMID: 36291552 PMCID: PMC9599219 DOI: 10.3390/biom12101343] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 08/02/2023] Open
Abstract
The architecture of eukaryotic cells is defined by extensive membrane-delimited compartments, which entails separate metabolic processes that would otherwise interfere with each other, leading to functional differences between cells. G protein-coupled receptors (GPCRs) are the largest class of cell surface receptors, and their signal transduction is traditionally viewed as a chain of events initiated from the plasma membrane. Furthermore, their intracellular trafficking, internalization, and recycling were considered only to regulate receptor desensitization and cell surface expression. On the contrary, accumulating data strongly suggest that GPCRs also signal from intracellular compartments. GPCRs localize in the membranes of endosomes, nucleus, Golgi and endoplasmic reticulum apparatuses, mitochondria, and cell division compartments. Importantly, from these sites they have shown to orchestrate multiple signals that regulate different cell pathways. In this review, we summarize the current knowledge of this fascinating phenomenon, explaining how GPCRs reach the intracellular sites, are stimulated by the endogenous ligands, and their potential physiological/pathophysiological roles. Finally, we illustrate several mechanisms involved in the modulation of the compartmentalized GPCR signaling by drugs and endogenous ligands. Understanding how GPCR signaling compartmentalization is regulated will provide a unique opportunity to develop novel pharmaceutical approaches to target GPCRs and potentially lead the way towards new therapeutic approaches.
Collapse
Affiliation(s)
- Irene Fasciani
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Marco Carli
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Francesco Petragnano
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Francesco Colaianni
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Gabriella Aloisi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Roberto Maggio
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| | - Marco Scarselli
- Department of Translational Research on New Technologies in Medicine and Surgery, University of Pisa, 56126 Pisa, Italy
| | - Mario Rossi
- Department of Biotechnological and Applied Clinical Sciences, University of L’Aquila, 67100 L’Aquila, Italy
| |
Collapse
|
11
|
Zhang J, Wang X, Lin Z, Zhu T. Pro-angiogenic activity of isofuran. Biosci Biotechnol Biochem 2022; 86:1506-1514. [PMID: 36066914 DOI: 10.1093/bbb/zbac146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/29/2022] [Indexed: 11/12/2022]
Abstract
Isofurans (IsoFs) are a series of novel discovered lipid peroxidation products. This study focused on the investigation of angiogenic property of IsoF. MTT stain assay indicated that 1 μM IsoF had the most bioactivity in RBECs. IsoF significantly promoted cellular proliferation and migration and remarkably decreased staurosporine-induced apoptosis by TUNEL assay in the RBECs. It successfully up-regulated rat aortic vascularization and choroid explant sprouting, extracellular regulated protein kinases (ERK)1/2, and triggered calcium release. RT-PCR examination indicated that IsoF up-regulated tumor necrosis factor (TNF)α, angiopoietin-1 receptor (Tie2), and vascular endothelial growth factor (VEGF)-A, but did not interfere with caspase 2 and VEGF-C in the RBECs. IsoF has pro-angiogenic activity. Calcium release and ERK1/2 phosphorylation may be involved in the signaling of the IsoF-induced up-regulation of TNFα, Tie2, and VEGF-A, which could be the molecular mechanism of the pro-angiogenic activity of the IsoF.
Collapse
Affiliation(s)
- Jingxia Zhang
- Key Laboratory of Translational Tumor Medicine in Fujian Province, School of Basic Medical Science Putian University, Putian, Fujian, China
| | - Xiangjiang Wang
- Key Laboratory of Translational Tumor Medicine in Fujian Province, School of Basic Medical Science Putian University, Putian, Fujian, China
| | - Zhiping Lin
- Key Laboratory of Translational Tumor Medicine in Fujian Province, School of Basic Medical Science Putian University, Putian, Fujian, China
| | - Tang Zhu
- Key Laboratory of Translational Tumor Medicine in Fujian Province, School of Basic Medical Science Putian University, Putian, Fujian, China.,Yujia Biotech., Guangzhou, Guangdong, China
| |
Collapse
|
12
|
Sanchez M, Hamel D, Bajon E, Duhamel F, Bhosle VK, Zhu T, Rivera JC, Dabouz R, Nadeau-Vallée M, Sitaras N, Tremblay DÉ, Omri S, Habelrih T, Rouget R, Hou X, Gobeil F, Joyal JS, Sapieha P, Mitchell G, Ribeiro-Da-Silva A, Mohammad Nezhady MA, Chemtob S. The Succinate Receptor SUCNR1 Resides at the Endoplasmic Reticulum and Relocates to the Plasma Membrane in Hypoxic Conditions. Cells 2022; 11:2185. [PMID: 35883628 PMCID: PMC9321536 DOI: 10.3390/cells11142185] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 12/24/2022] Open
Abstract
The GPCR SUCNR1/GPR91 exerts proangiogenesis upon stimulation with the Krebs cycle metabolite succinate. GPCR signaling depends on the surrounding environment and intracellular localization through location bias. Here, we show by microscopy and by cell fractionation that in neurons, SUCNR1 resides at the endoplasmic reticulum (ER), while being fully functional, as shown by calcium release and the induction of the expression of the proangiogenic gene for VEGFA. ER localization was found to depend upon N-glycosylation, particularly at position N8; the nonglycosylated mutant receptor localizes at the plasma membrane shuttled by RAB11. This SUCNR1 glycosylation is physiologically regulated, so that during hypoxic conditions, SUCNR1 is deglycosylated and relocates to the plasma membrane. Downstream signal transduction of SUCNR1 was found to activate the prostaglandin synthesis pathway through direct interaction with COX-2 at the ER; pharmacologic antagonism of the PGE2 EP4 receptor (localized at the nucleus) was found to prevent VEGFA expression. Concordantly, restoring the expression of SUCNR1 in the retina of SUCNR1-null mice renormalized vascularization; this effect is markedly diminished after transfection of the plasma membrane-localized SUCNR1 N8A mutant, emphasizing that ER localization of the succinate receptor is necessary for proper vascularization. These findings uncover an unprecedented physiologic process where GPCR resides at the ER for signaling function.
Collapse
Affiliation(s)
- Melanie Sanchez
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - David Hamel
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Emmanuel Bajon
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - François Duhamel
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Vikrant K. Bhosle
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
- Cell Biology Program, The Hospital for Sick Children Research Institute, Toronto, ON M5G 1X8, Canada
| | - Tang Zhu
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Jose Carlos Rivera
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Rabah Dabouz
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Mathieu Nadeau-Vallée
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Nicholas Sitaras
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - David-Étienne Tremblay
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Samy Omri
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Tiffany Habelrih
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
| | - Raphael Rouget
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Xin Hou
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Fernand Gobeil
- Department of Pharmacology, Université de Sherbrooke, Sherbrooke, QC J1H 5N4, Canada;
| | - Jean-Sébastien Joyal
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Przemyslaw Sapieha
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
| | - Grant Mitchell
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
| | - Alfredo Ribeiro-Da-Silva
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
| | - Mohammad Ali Mohammad Nezhady
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Program of Molecular Biology, Faculty of Medicine, Université de Montréal, Montréal, QC H3T 1J4, Canada
- Research Center-CHU Ste-Justine, Departments of Pediatrics, Ophthalmology, and Pharmacology, Faculty of Medicine, Université de Montréal, 3175, Chemin Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| | - Sylvain Chemtob
- Department of Pharmacology and Therapeutics, McGill University, Montréal, QC H3A 1A3, Canada; (M.S.); (V.K.B.); (R.D.); (R.R.); (A.R.-D.-S.)
- Department of Pharmacology, Université de Montréal, Montréal, QC H3C 3J7, Canada; (D.H.); (F.D.); (M.N.-V.); (N.S.); (D.-É.T.); (T.H.)
- Department of Pediatrics, Research Center-CHU Ste-Justine, Montréal, QC H3T 1C5, Canada; (E.B.); (T.Z.); (X.H.); (J.-S.J.); (G.M.)
- Department of Ophthalmology, Research Center of Hôpital Maisonneuve-Rosemont, Université de Montréal, Montréal, QC H1T 2M4, Canada; (J.C.R.); (S.O.); (P.S.)
- Research Center-CHU Ste-Justine, Departments of Pediatrics, Ophthalmology, and Pharmacology, Faculty of Medicine, Université de Montréal, 3175, Chemin Côte Ste-Catherine, Montréal, QC H3T 1C5, Canada
| |
Collapse
|
13
|
Positive allosteric regulation of PAC1-R up-regulates PAC1-R and its specific ligand PACAP. Acta Biochim Biophys Sin (Shanghai) 2022; 54:657-672. [PMID: 35593471 PMCID: PMC9828401 DOI: 10.3724/abbs.2022041] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PAC1-R is a recognized preferential receptor for the neuropeptide of pituitary adenylate cyclase-activating polypeptide (PACAP), which mediates neuroprotective and nerve regenerative activities of PACAP. In this study, we found that in both PAC1R-CHO cells with high expression of PAC1R-eGFP and retinal ganglion cells (RGC-5) with the natural expression of PAC1-R, oligo-peptide PACAP(28-38) and the positively charged arginine-rich penetrating peptide TAT, as positive allosteric modulators of PAC1-R, significantly trigger the nuclear translocation of PAC1-R. The chromatin immunoprecipitation (ChIP)-PCR results show that the nuclear translocated PAC1-R binds with the promoter regions of PAC1-R and its specific ligand PACAP. The up-regulated promoter activities of PAC1-R and PACAP induced by PACAP(28-38) or TAT are positively correlative with the increase of the expression levels of PAC1-R and PACAP. Moreover, the nuclear translocation of PAC1-R induced by PACAP(28-38) or TAT is significantly inhibited by the mutation of PAC1-R on Cys25 and the palmitoylation inhibitor 2-bromopalmitate. Meanwhile, the increase in both PAC1-R and PACAP levels and the neuroprotective activities of PACAP(28-38) and TAT in MPP-induced cell model of Parkinson ' s disease are synchronously inhibited by 2-bromopalmitate, which are positively correlated with the nuclear translocation of PAC1-R induced by PACAP(28-38) or TAT. Bioinformatics analysis and motif enrichment analysis following ChIP-sequencing show that the transcription factors including SP1, Zic2, GATA1, REST and YY1 may be recruited by nuclear PAC1-R and involved in regulating the promoter activities of PAC1-R and PACAP. ChIP-sequencing and related bioinformatics analysis show that the downstream target genes regulated by the nuclear PAC1-R are mostly involved in the process of cellular stress and related to neuroprotection, neuronal genesis and development.
Collapse
|
14
|
LncRNAS—modulators of neurovascular units in diabetic retinopathy. Eur J Pharmacol 2022; 925:174937. [DOI: 10.1016/j.ejphar.2022.174937] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/14/2022] [Accepted: 03/30/2022] [Indexed: 01/08/2023]
|
15
|
Zheng HC, Jiang HM. Shuttling of cellular proteins between the plasma membrane and nucleus (Review). Mol Med Rep 2021; 25:14. [PMID: 34779504 PMCID: PMC8600410 DOI: 10.3892/mmr.2021.12530] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/09/2021] [Indexed: 11/23/2022] Open
Abstract
Recently accumulated evidence has indicated that the nucleomembrane shuttling of cellular proteins is common, which provides new insight into the subcellular translocation and biological functions of proteins synthesized in the cytoplasm. The present study aimed to clarify the trafficking of proteins between the plasma membrane and nucleus. These proteins primarily consist of transmembrane receptors, membrane adaptor proteins, adhesive proteins, signal proteins and nuclear proteins, which contribute to proliferation, apoptosis, chemoresistance, adhesion, migration and gene expression. The proteins frequently undergo cross-talk, such as the interaction of transmembrane proteins with signal proteins. The transmembrane proteins undergo endocytosis, infusion into organelles or proteolysis into soluble forms for import into the nucleus, while nuclear proteins interact with membrane proteins or act as receptors. The nucleocytosolic translocation involves export or import through nuclear membrane pores by importin or exportin. Nuclear proteins generally interact with other transcription factors, and then binding to the promoter for gene expression, while membrane proteins are responsible for signal initiation by binding to other membrane and/or adaptor proteins. Protein translocation occurs in a cell-specific manner and is closely linked to cellular biological events. The present review aimed to improve understanding of cytosolic protein shuttling between the plasma membrane and nucleus and the associated signaling pathways.
Collapse
Affiliation(s)
- Hua-Chuan Zheng
- Department of Oncology, The Affiliated Hospital of Chengde Medical University, Chengde, Hebei 067000, P.R. China
| | - Hua-Mao Jiang
- Department of Urology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121001, P.R. China
| |
Collapse
|
16
|
Liu H, Yang J, Zhang Y, Han J, Yang Y, Zhao Z, Dai X, Wang H, Ding X, Liu Y, Zhong W, Gao W, Sun T. Psychologic Stress Drives Progression of Malignant Tumors via DRD2/HIF1α Signaling. Cancer Res 2021; 81:5353-5365. [PMID: 34321238 PMCID: PMC9306299 DOI: 10.1158/0008-5472.can-21-1043] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 04/16/2021] [Accepted: 07/23/2021] [Indexed: 01/07/2023]
Abstract
Although it is established that the sustained psychologic stress conditions under which patients with tumors often reside accelerates malignant progression of tumors, the molecular mechanism behind this association is unclear. In this work, the effect of psychologic stress on tumor progression was verified using a stress-stimulated tumor-bearing mouse model (Str-tumor). Both D2 dopamine receptor (DRD2) and hypoxia-inducible factor-1α (HIF1α) were highly expressed in the nucleus of Str-tumors. Treatment with trifluoperazine (TFP), a DRD2 inhibitor, elicited better antitumor effects in Str-tumors than the control group. These results indicate that DRD2 may mediate stress-induced malignant tumor progression. DRD2 interacted with von Hippel-Lindau (VHL) in the nucleus, and competitive binding of DRD2 and HIF1α to VHL resulted in reduced ubiquitination-mediated degradation of HIF1α, enhancing the epithelial-mesenchymal transition of tumor cells. TFP acted as an interface inhibitor between DRD2 and VHL to promote the degradation of HIF1α. In conclusion, DRD2 may promote the progression of malignant tumors induced by psychologic stress via activation of the oxygen-independent HIF1α pathway, and TFP may serve as a therapeutic strategy for stress management in patients with cancer. SIGNIFICANCE: This work identifies DRD2 regulation of HIF1α as a mechanism underlying the progression of malignant tumors stimulated by psychologic stress and suggests that DRD2 inhibition can mitigate these stress conditions in patients.See related commentary by Bernabé, p. 5144.
Collapse
Affiliation(s)
- Huijuan Liu
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China.,Department of Anesthesiology, Tianjin Fourth Central Hospital, Tianjin, China
| | - Jiahuan Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Yang Zhang
- Department of Pathology, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, Shandong, China
| | - Jingxia Han
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Yuyan Yang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Zihan Zhao
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Xintong Dai
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Hongqi Wang
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Xiujuan Ding
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China
| | - Yanrong Liu
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Weilong Zhong
- Department of Gastroenterology and Hepatology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wenqing Gao
- Tianjin Key Laboratory of Early Druggability Evaluation of Innovative Drugs and Tianjin Key Laboratory of Molecular Drug Research, Tianjin International Joint Academy of Biomedicine, Tianjin, China.,Corresponding Authors: Tao Sun, Nankai University, State Key Laboratory of Medicinal Chemical Biology, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300450 China. Phone: 13512922691; E-mail: ; and Wenqing Gao, Phone: 18512215515; E-mail:
| | - Tao Sun
- State Key Laboratory of Medicinal Chemical Biology and College of Pharmacy, Nankai University, Tianjin, China.,Corresponding Authors: Tao Sun, Nankai University, State Key Laboratory of Medicinal Chemical Biology, No. 38 Tongyan Road, Haihe River Education Park, Jinnan District, Tianjin, 300450 China. Phone: 13512922691; E-mail: ; and Wenqing Gao, Phone: 18512215515; E-mail:
| |
Collapse
|
17
|
Wang W, Bian J, Sun Y, Li Z. The new fate of internalized membrane receptors: Internalized activation. Pharmacol Ther 2021; 233:108018. [PMID: 34626676 DOI: 10.1016/j.pharmthera.2021.108018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 10/02/2021] [Accepted: 10/04/2021] [Indexed: 12/23/2022]
Abstract
Classically, the fate of internalized membrane receptors includes receptor degradation and receptor recycling. However, recent findings have begun to challenge these views. Much research demonstrated that many internalized membrane receptors can trigger distinct signal activation rather than being desensitized inside the cell. Here, we introduce the concept of "internalized activation" which not only represents a new mode of receptor activation, but also endows the new fate for receptor internalization (from death to life). The new activation mode and fate of membrane receptor are ubiquitous and have unique theoretical significance. We systematically put forward the features, process, and regulation of "internalized activation" and its significance in signal transduction and diseases. "Internalized activation" will provide a completely new understanding for the theory of receptor activation, internalization and novel drug targets for precision medicine.
Collapse
Affiliation(s)
- Wenjing Wang
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Jingwei Bian
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Yang Sun
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China
| | - Zijian Li
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing Key Laboratory of Cardiovascular Receptors Research, Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Ministry of Health, Key Laboratory of Molecular Cardiovascular Sciences, Ministry of Education, Beijing 100191, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
18
|
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
|
19
|
Jones I, Hägglund AC, Carlsson L. Reduced mTORC1-signaling in retinal ganglion cells leads to vascular retinopathy. Dev Dyn 2021; 251:321-335. [PMID: 34148274 DOI: 10.1002/dvdy.389] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND The coordinated wiring of neurons, glia and endothelial cells into neurovascular units is critical for central nervous system development. This is best exemplified in the mammalian retina where interneurons, astrocytes and retinal ganglion cells sculpt their vascular environment to meet the metabolic demands of visual function. Identifying the molecular networks that underlie neurovascular unit formation is an important step towards a deeper understanding of nervous system development and function. RESULTS Here, we report that cell-to-cell mTORC1-signaling is essential for neurovascular unit formation during mouse retinal development. Using a conditional knockout approach we demonstrate that reduced mTORC1 activity in asymmetrically positioned retinal ganglion cells induces a delay in postnatal vascular network formation in addition to the production of rudimentary and tortuous vessel networks in adult animals. The severity of this vascular phenotype is directly correlated to the degree of mTORC1 down regulation within the neighboring retinal ganglion cell population. CONCLUSIONS This study establishes a cell nonautonomous role for mTORC1-signaling during retinal development. These findings contribute to our current understanding of neurovascular unit formation and demonstrate how ganglion cells actively sculpt their local environment to ensure that the retina is perfused with an appropriate supply of oxygen and nutrients.
Collapse
Affiliation(s)
- Iwan Jones
- Umeå Center for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
| | | | - Leif Carlsson
- Umeå Center for Molecular Medicine (UCMM), Umeå University, Umeå, Sweden
| |
Collapse
|
20
|
Nassar E, Hassan N, El-Ghonaimy EA, Hassan H, Abdullah MS, Rottke TV, Kiesel L, Greve B, Ibrahim SA, Götte M. Syndecan-1 Promotes Angiogenesis in Triple-Negative Breast Cancer through the Prognostically Relevant Tissue Factor Pathway and Additional Angiogenic Routes. Cancers (Basel) 2021; 13:cancers13102318. [PMID: 34066023 PMCID: PMC8150756 DOI: 10.3390/cancers13102318] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/07/2021] [Accepted: 05/07/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Triple-negative breast cancer is an aggressive subtype of breast cancer characterized by tumor angiogenesis and poor patient survival. Here, we analyzed the function of the cell surface molecule Syndecan-1 in tumor angiogenesis in a 3D cell culture system. As a novel finding, we demonstrate that downregulation of Syndecan-1 reduces angiogenesis by decreasing the amount of angiogenesis factors of the tissue factor pathway. Furthermore, we show that the components of this pathway are associated with the prognosis of breast cancer patients. Our study identifies Syndecan-1 and the tissue factor pathway as novel potential therapeutic targets in the aggressive triple-negative subtype of breast cancer, for which no targeted therapies are currently available. Abstract Triple-negative breast cancer (TNBC) is characterized by increased angiogenesis, metastasis, and poor survival. Dysregulation of the cell surface heparan sulfate proteoglycan and signaling co-receptor Syndecan-1 is linked to poor prognosis. To study its role in angiogenesis, we silenced Syndecan-1 in TNBC cell lines using a 3D human umbilical vein endothelial cell (HUVEC) co-culture system. Syndecan-1 siRNA depletion in SUM-149, MDA-MB-468, and MDA-MB-231 cells decreased HUVEC tubule network formation. Angiogenesis array revealed reduced VEGF-A and tissue factor (TF) in the Syndecan-1-silenced secretome. qPCR independently confirmed altered expression of F3, F7, F2R/PAR1, F2RL1/PAR2, VEGF-A, EDN1, IGFBP1, and IGFBP2 in SUM-149, MDA-MB-231, and MDA-MB-468 cells. ELISA revealed reduced secreted endothelin-1 (SUM-149, MDA-MB-468) and TF (all cell lines) upon Syndecan-1 depletion, while TF pathway inhibitor treatment impaired angiogenesis. Survival analysis of 3951 patients demonstrated that high expression of F3 and F7 are associated with better relapse-free survival, whereas poor survival was observed in TNBC and p53 mutant basal breast cancer (F3) and in ER-negative and HER2-positive breast cancer (F2R, F2RL1). STRING protein network analysis revealed associations of Syndecan-1 with VEGF-A and IGFBP1, further associated with the TF and ET-1 pathways. Our study suggests that TNBC Syndecan-1 regulates angiogenesis via the TF and additional angiogenic pathways and marks its constituents as novel prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Eyyad Nassar
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
| | - Nourhan Hassan
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, 12613 Giza, Egypt;
| | - Eslam A. El-Ghonaimy
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
- Department of Zoology, Faculty of Science, Cairo University, 12613 Giza, Egypt;
| | - Hebatallah Hassan
- Department of Zoology, Faculty of Science, Cairo University, 12613 Giza, Egypt;
| | - Mahmoud Salah Abdullah
- Biotechnology/Biomolecular Chemistry Program, Faculty of Science, Cairo University, 12613 Giza, Egypt;
| | - Theresa V. Rottke
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
| | - Ludwig Kiesel
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
| | - Burkhard Greve
- Department of Radiotherapy and Radiooncology, University Hospital Münster, 48149 Münster, Germany;
| | - Sherif Abdelaziz Ibrahim
- Department of Zoology, Faculty of Science, Cairo University, 12613 Giza, Egypt;
- Correspondence: (S.A.I.); (M.G.)
| | - Martin Götte
- Department of Gynecology and Obstetrics, Münster University Hospital, Albert-Schweitzer-Campus 1, D11, 48149 Münster, Germany; (E.N.); (N.H.); (E.A.E.-G.); (T.V.R.); (L.K.)
- Correspondence: (S.A.I.); (M.G.)
| |
Collapse
|
21
|
Gonçalves-Monteiro S, Ribeiro-Oliveira R, Vieira-Rocha MS, Vojtek M, Sousa JB, Diniz C. Insights into Nuclear G-Protein-Coupled Receptors as Therapeutic Targets in Non-Communicable Diseases. Pharmaceuticals (Basel) 2021; 14:439. [PMID: 34066915 PMCID: PMC8148550 DOI: 10.3390/ph14050439] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 04/27/2021] [Accepted: 04/30/2021] [Indexed: 12/14/2022] Open
Abstract
G-protein-coupled receptors (GPCRs) comprise a large protein superfamily divided into six classes, rhodopsin-like (A), secretin receptor family (B), metabotropic glutamate (C), fungal mating pheromone receptors (D), cyclic AMP receptors (E) and frizzled (F). Until recently, GPCRs signaling was thought to emanate exclusively from the plasma membrane as a response to extracellular stimuli but several studies have challenged this view demonstrating that GPCRs can be present in intracellular localizations, including in the nuclei. A renewed interest in GPCR receptors' superfamily emerged and intensive research occurred over recent decades, particularly regarding class A GPCRs, but some class B and C have also been explored. Nuclear GPCRs proved to be functional and capable of triggering identical and/or distinct signaling pathways associated with their counterparts on the cell surface bringing new insights into the relevance of nuclear GPCRs and highlighting the nucleus as an autonomous signaling organelle (triggered by GPCRs). Nuclear GPCRs are involved in physiological (namely cell proliferation, transcription, angiogenesis and survival) and disease processes (cancer, cardiovascular diseases, etc.). In this review we summarize emerging evidence on nuclear GPCRs expression/function (with some nuclear GPCRs evidencing atypical/disruptive signaling pathways) in non-communicable disease, thus, bringing nuclear GPCRs as targets to the forefront of debate.
Collapse
Affiliation(s)
- Salomé Gonçalves-Monteiro
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Rita Ribeiro-Oliveira
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Maria Sofia Vieira-Rocha
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Martin Vojtek
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Joana B. Sousa
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| | - Carmen Diniz
- Laboratory of Pharmacology, Department of Drug Sciences, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; (S.G.-M.); (R.R.-O.); (M.S.V.-R.); (M.V.)
- LAQV/REQUIMTE, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
22
|
Lu J, Xu S, Huo Y, Sun D, Hu Y, Wang J, Zhang X, Wang P, Li Z, Liang M, Wu Z, Liu P. Sorting nexin 3 induces heart failure via promoting retromer-dependent nuclear trafficking of STAT3. Cell Death Differ 2021; 28:2871-2887. [PMID: 33947971 DOI: 10.1038/s41418-021-00789-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/08/2021] [Accepted: 04/15/2021] [Indexed: 12/18/2022] Open
Abstract
Sorting nexins (SNXs), the retromer-associated cargo binding proteins, have emerged as critical regulators of the trafficking of proteins involved in the pathogenesis of diverse diseases. However, studies of SNXs in the development of cardiovascular diseases, especially cardiac hypertrophy and heart failure, are lacking. Here, we ask whether SNX3, the simplest structured isoform in the SNXs family, may act as a key inducer of myocardial injury. An increased level of SNX3 was observed in failing hearts from human patients and mice. Cardiac-specific Snx3 knockout (Snx3-cKO) mice and Snx3 transgenic (Snx3-cTg) mice were generated to evaluate the role of Snx3 in myocardial hypertrophy, fibrosis, and heart function by morphology, echocardiography, histological staining, and hypertrophic biomarkers. We report that Snx3-cKO in mice significantly protected against isoproterenol (ISO)-induced cardiac hypertrophy at 12 weeks. Conversely, Snx3-cTg mice were more susceptible to ISO-induced cardiac hypertrophy at 12 weeks and showed aggravated cardiac injury even heart failure at 24 weeks. Immunoprecipitation-based mass spectrometry, immunofluorescent staining, co-immunoprecipitation, localized surface plasmon resonance, and proximity ligation assay were performed to examine the direct interaction of SNX3-retromer with signal transducer and activator of transcription 3 (STAT3). We discovered that STAT3 was a new interacting partner of SNX3-retromer, and SNX3-retromer served as an essential platform for assembling gp130/JAK2/STAT3 complexes and subsequent phosphorylation of STAT3 by direct combination at EE. SNX3-retromer and STAT3 complexes were transiently imported into the nucleus after hypertrophic stimuli. The pharmacological inhibition or knockdown of STAT3 reversed SNX3 overexpression-induced myocardial injury. STAT3 overexpression blunts the beneficial function of SNX3 knockdown on hypertrophic cardiomyocytes. We show that SNX3-retromer promoted importin α3-mediated STAT3 nuclear trafficking and ultimately leading to cardiac injury. Taken together, our study reveals that SNX3 plays a key role in cardiac function and implicates SNX3 as a potential therapeutic target for cardiac hypertrophy and heart failure.
Collapse
Affiliation(s)
- Jing Lu
- School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Suowen Xu
- Aab Cardiovascular Research Institute, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA.,Department of Endocrinology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, P. R. China
| | - Yuqing Huo
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Vascular Biology Center, Augusta University, Augusta, GA, USA
| | - Duanping Sun
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, P.R. China
| | - Yuehuai Hu
- School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Junjian Wang
- School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Xiaolei Zhang
- School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Panxia Wang
- School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Zhuoming Li
- School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, P. R. China
| | - Mengya Liang
- Department of Cardiac Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China
| | - Zhongkai Wu
- Department of Cardiac Surgery, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, P.R. China.
| | - Peiqing Liu
- School of Pharmaceutical Sciences, National and Local United Engineering Lab of Druggability and New Drugs Evaluation, Sun Yat-sen University, Guangzhou, P. R. China.
| |
Collapse
|
23
|
Ren Y, Zhang Y, Wang L, He F, Yan M, Liu X, Ou Y, Wu Q, Bi T, Wang S, Liu J, Ding BS, Wang L, Qing J. Selective Targeting of Vascular Endothelial YAP Activity Blocks EndMT and Ameliorates Unilateral Ureteral Obstruction-Induced Kidney Fibrosis. ACS Pharmacol Transl Sci 2021; 4:1066-1074. [PMID: 34151201 DOI: 10.1021/acsptsci.1c00010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Indexed: 02/08/2023]
Abstract
Kidney fibrosis is accompanied by vascular dysfunction. Discovering new ways to ameliorate dysfunctional angiogenesis may bypass kidney fibrosis. YAP (Yes-associated protein) plays a multifaceted role during angiogenesis. Here, we found that selectively targeting YAP signaling in the endothelium ameliorates unilateral ureteral obstruction (UUO)-induced kidney fibrosis. Genetic deletion of Yap1, encoding YAP protein, in VE-cadherin+ endothelial cells inhibited endothelial-to-mesenchymal transition (EndMT) and dysfunctional angiogenesis and improved obstructive nephropathy and kidney fibrosis. Treatment with the systemic YAP inhibitor verteporfin worsened kidney fibrosis symptoms because of its lack of cell specificity. In an attempt to identify endothelial-specific YAP modulators, we found that G-protein-coupled receptor coagulation factor II receptor-like 1 (F2RL1) was highly expressed in vessels after UUO-induced kidney fibrosis. The F2RL1 peptide antagonist FSLLRY-NH2 selectively blocked YAP activity in endothelial cells and ameliorated kidney fibrosis. Thus, selective antagonization of endothelial YAP activity might bypass kidney fibrosis and provide new avenues for the design of antifibrotic therapies.
Collapse
Affiliation(s)
- Yafeng Ren
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Yuwei Zhang
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Lu Wang
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Fuqian He
- The Center of Gerontology and Geriatrics and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Mengli Yan
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| | - Xiaoheng Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Yangying Ou
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Qinkai Wu
- Michael Smith Laboratories, University of British Columbia, Vancouver V6T1Z4, Canada
| | - Tao Bi
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Shiyuan Wang
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Jian Liu
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Bi-Sen Ding
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China.,Fibrosis Research Center, Icahn School of Medicine at Mount Sinai, New York, New York 10128, United States.,Ansary Stem Cell Institute, Weill Cornell Medicine, New York, New York 10065, United States
| | - Li Wang
- Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China
| | - Jie Qing
- National Traditional Chinese Medicine Clinical Research Base, Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Research Center of Integrated Traditional Chinese and Western Medicine, Affiliated Traditional Medicine Hospital, Southwest Medical University, Luzhou 646000, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu 610064, China
| |
Collapse
|
24
|
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: 5] [Impact Index Per Article: 1.3] [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
|
25
|
Calebiro D, Koszegi Z, Lanoiselée Y, Miljus T, O'Brien S. G protein-coupled receptor-G protein interactions: a single-molecule perspective. Physiol Rev 2020; 101:857-906. [PMID: 33331229 DOI: 10.1152/physrev.00021.2020] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
G protein-coupled receptors (GPCRs) regulate many cellular and physiological processes, responding to a diverse range of extracellular stimuli including hormones, neurotransmitters, odorants, and light. Decades of biochemical and pharmacological studies have provided fundamental insights into the mechanisms of GPCR signaling. Thanks to recent advances in structural biology, we now possess an atomistic understanding of receptor activation and G protein coupling. However, how GPCRs and G proteins interact in living cells to confer signaling efficiency and specificity remains insufficiently understood. The development of advanced optical methods, including single-molecule microscopy, has provided the means to study receptors and G proteins in living cells with unprecedented spatio-temporal resolution. The results of these studies reveal an unexpected level of complexity, whereby GPCRs undergo transient interactions among themselves as well as with G proteins and structural elements of the plasma membrane to form short-lived signaling nanodomains that likely confer both rapidity and specificity to GPCR signaling. These findings may provide new strategies to pharmaceutically modulate GPCR function, which might eventually pave the way to innovative drugs for common diseases such as diabetes or heart failure.
Collapse
Affiliation(s)
- Davide Calebiro
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, United Kingdom
| | - Zsombor Koszegi
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, United Kingdom
| | - Yann Lanoiselée
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, United Kingdom
| | - Tamara Miljus
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, United Kingdom
| | - Shannon O'Brien
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Nottingham and Birmingham, Birmingham, United Kingdom
| |
Collapse
|
26
|
Crilly SE, Puthenveedu MA. Compartmentalized GPCR Signaling from Intracellular Membranes. J Membr Biol 2020; 254:259-271. [PMID: 33231722 DOI: 10.1007/s00232-020-00158-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 11/11/2020] [Indexed: 12/21/2022]
Abstract
G protein-coupled receptors (GPCRs) are integral membrane proteins that transduce a wide array of inputs including light, ions, hormones, and neurotransmitters into intracellular signaling responses which underlie complex processes ranging from vision to learning and memory. Although traditionally thought to signal primarily from the cell surface, GPCRs are increasingly being recognized as capable of signaling from intracellular membrane compartments, including endosomes, the Golgi apparatus, and nuclear membranes. Remarkably, GPCR signaling from these membranes produces functional effects that are distinct from signaling from the plasma membrane, even though often the same G protein effectors and second messengers are activated. In this review, we will discuss the emerging idea of a "spatial bias" in signaling. We will present the evidence for GPCR signaling through G protein effectors from intracellular membranes, and the ways in which this signaling differs from canonical plasma membrane signaling with important implications for physiology and pharmacology. We also highlight the potential mechanisms underlying spatial bias of GPCR signaling, including how intracellular membranes and their associated lipids and proteins affect GPCR activity and signaling.
Collapse
Affiliation(s)
- Stephanie E Crilly
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Manojkumar A Puthenveedu
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, MI, 48109, USA.,Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| |
Collapse
|
27
|
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: 39] [Impact Index Per Article: 7.8] [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
| | | | - 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
|
28
|
Protease-activated receptor 2 contributes to placental development and fetal growth in mice. Thromb Res 2020; 193:173-179. [PMID: 32717642 DOI: 10.1016/j.thromres.2020.06.039] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/17/2020] [Accepted: 06/23/2020] [Indexed: 11/20/2022]
Abstract
BACKGROUND Protease-activated receptor 2 (PAR2) is activated by serine proteases such as coagulation tissue factor/VIIa complex, factor Xa or trypsin and is pro-angiogenic in several disease models. Impaired angiogenesis in placenta causes placental dysfunction and fetal growth restriction. PAR2 is expressed in the placenta trophoblast. However, the role of PAR2 in pregnancy remains unknown. OBJECTIVE The present study aimed to examine the role of PAR2 in placental development and fetal growth using a murine model. METHODS PAR2-/- or PAR2+/+ mice in the ICR background were used. Female PAR2-/- mice were mated with male PAR2-/- mice, and female PAR2+/+ mice were mated with male PAR2+/+ mice to obtain PAR2-/- and PAR2+/+ fetuses, respectively. The day a virginal plug was observed in the morning was determined as 0.5-day post-coitum (dpc). Pregnant mice were sacrificed on 13.5 or 18.5 dpc to collect samples. RESULTS A deficiency of PAR2 significantly reduced the fetal and placental weight and impaired placental labyrinth development in mice on 18.5 dpc. Collagen IV expression in placenta labyrinth was smaller in PAR2 knockout mice compared to that of wild-type mice. A deficiency of PAR2 also reduced the expression levels of genes related to angiogenesis and coagulation in placenta. CONCLUSION Our data suggest that PAR2 is required for fetal growth and angiogenesis in the placenta and is thus important for a normal pregnancy.
Collapse
|
29
|
Okabe K, Fukada H, Tai-Nagara I, Ando T, Honda T, Nakajima K, Takeda N, Fong GH, Ema M, Kubota Y. Neuron-derived VEGF contributes to cortical and hippocampal development independently of VEGFR1/2-mediated neurotrophism. Dev Biol 2020; 459:65-71. [PMID: 31790655 DOI: 10.1016/j.ydbio.2019.11.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 11/07/2019] [Accepted: 11/28/2019] [Indexed: 12/13/2022]
Abstract
Vascular endothelial growth factor (VEGF) is a potent mitogen critical for angiogenesis and organogenesis. Deletion or inhibition of VEGF during development not only profoundly suppresses vascular outgrowth, but significantly affects the development and function of various organs. In the brain, VEGF is thought to not only promote vascular growth, but also directly act on neurons as a neurotrophic factor by activating VEGF receptors. In the present study, we demonstrated that deletion of VEGF using hGfap-Cre line, which recombines genes specifically in cortical and hippocampal neurons, severely impaired brain organization and vascularization of these regions. The mutant mice had motor deficits, with lethality around the time of weaning. Multiple reporter lines indicated that VEGF was highly expressed in neurons, but that its cognate receptors, VEGFR1 and 2 were exclusive to endothelial cells in the brain. In accordance, mice lacking neuronal VEGFR1 and VEGFR2 did not exhibit neuronal deformities or lethality. Taken together, our data suggest that neuron-derived VEGF contributes to cortical and hippocampal development likely through angiogenesis independently of direct neurotrophic effects mediated by VEGFR1 and 2.
Collapse
Affiliation(s)
- Keisuke Okabe
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Plastic Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| | - Hugh Fukada
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Ikue Tai-Nagara
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Tomofumi Ando
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan; Department of Surgery, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Takao Honda
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan
| | - Norihiko Takeda
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan
| | - Guo-Hua Fong
- Center for Vascular Biology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06032, USA; Department of Cell Biology, University of Connecticut School of Medicine, 263 Farmington Avenue, Farmington, CT, 06032, USA
| | - Masatsugu Ema
- Department of Stem Cells and Human Disease Models, Research Center for Animal Life Science, Shiga University of Medical Science, Seta, Tsukinowa-cho, Otsu, Shiga, 520-2192, Japan
| | - Yoshiaki Kubota
- Department of Anatomy, Keio University School of Medicine, 35 Shinanomachi, Shinjuku-ku, Tokyo, 160-8582, Japan.
| |
Collapse
|
30
|
Sayah DN, Zhou TE, Omri S, Mazzaferri J, Quiniou C, Wirth M, Côté F, Dabouz R, Desjarlais M, Costantino S, Chemtob S. Novel Anti-Interleukin-1β Therapy Preserves Retinal Integrity: A Longitudinal Investigation Using OCT Imaging and Automated Retinal Segmentation in Small Rodents. Front Pharmacol 2020; 11:296. [PMID: 32226385 PMCID: PMC7081735 DOI: 10.3389/fphar.2020.00296] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 02/27/2020] [Indexed: 12/16/2022] Open
Abstract
Retinopathy of prematurity (ROP) is the leading cause of blindness in neonates. Inflammation, in particular interleukin-1β (IL-1β), is increased in early stages of the disorder, and contributes to inner and outer retinal vasoobliteration in the oxygen-induced retinopathy (OIR) model of ROP. A small peptide antagonist of IL-1 receptor, composed of the amino acid sequence, rytvela, has been shown to exert beneficial anti-inflammatory effects without compromising immunovigilance-related NF-κB in reproductive tissues. We conducted a longitudinal study to determine the efficacy of “rytvela” in preserving the integrity of the retina in OIR model, using optical coherence tomography (OCT) which provides high-resolution cross-sectional imaging of ocular structures in vivo. Sprague–Dawley rats subjected to OIR and treated or not with “rytvela” were compared to IL-1 receptor antagonist (Kineret). OCT imaging and custom automated segmentation algorithm used to measure retinal thickness (RT) were obtained at P14 and P30; gold-standard immunohistochemistry (IHC) was used to confirm retinal anatomical changes. OCT revealed significant retinal thinning in untreated animals by P30, confirmed by IHC; these changes were coherently associated with increased apoptosis. Both rytvela and Kineret subsided apoptosis and preserved RT. As anticipated, Kineret diminished both SAPK/JNK and NF-κB axes, whereas rytvela selectively abated the former which resulted in preserved monocyte phagocytic function. Altogether, OCT imaging with automated segmentation is a reliable non-invasive approach to study longitudinally retinal pathology in small animal models of retinopathy.
Collapse
Affiliation(s)
- Diane N Sayah
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Tianwei E Zhou
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Samy Omri
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | | | - Christiane Quiniou
- Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| | - Maëlle Wirth
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - France Côté
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Rabah Dabouz
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Faculty of Medicine, McGill University, Montreal, QC, Canada
| | - Michel Desjarlais
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Santiago Costantino
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Sylvain Chemtob
- Hopital Maisonneuve-Rosemont Research Center, Montreal, QC, Canada.,Department of Ophthalmology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada.,Department of Pediatrics, Centre Hospitalier Universitaire Sainte-Justine Research Center, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
31
|
Zhang X, Fan S, Zhang L, Shi Y. Glucagon-like peptide-1 receptor undergoes importin-α-dependent nuclear localization in rat aortic smooth muscle cells. FEBS Lett 2020; 594:1506-1516. [PMID: 32010959 DOI: 10.1002/1873-3468.13751] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 01/21/2020] [Accepted: 01/29/2020] [Indexed: 11/08/2022]
Abstract
Glucagon-like peptide 1 receptor (GLP-1R) belongs to the family B of G protein-coupled receptors (GPCRs) and has antidiabetic and cardioprotective effects. Classical GLP-1R at the plasma membrane undergoes desensitization and internalization and is recycled back to the plasma membrane under the control of GLP-1 in islet β-cells. However, the subcellular localization of GLP-1R in the vascular system remains unclear. Here, we find that GLP-1R is localized in the nucleus of rat aortic smooth muscle cells (RASMCs) and in the tunica media. We identify a functional nuclear localization signal (NLS; 412-442aa) at the C-terminal region of GLP-1R. Nuclear import of GLP-1R is mediated by an importin-α-dependent pathway and regulated by phosphorylation of Ser416 in the NLS. Upon leaving the nucleus, GLP-1R promotes cell proliferation in RASMCs. These findings may provide insights into the cardiovascular functions of GLP-1R.
Collapse
Affiliation(s)
- Xin Zhang
- Key Laboratory of Chemical Biology and Molecular Engineering, Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, China
| | - Shaohua Fan
- Key Laboratory of Chemical Biology and Molecular Engineering, Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, China
| | - Lihui Zhang
- Shanxi Bethune Hospital, Taiyuan, Shanxi, China
| | - Yawei Shi
- Key Laboratory of Chemical Biology and Molecular Engineering, Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, China
| |
Collapse
|
32
|
Ribeiro-Oliveira R, Vojtek M, Gonçalves-Monteiro S, Vieira-Rocha MS, Sousa JB, Gonçalves J, Diniz C. Nuclear G-protein-coupled receptors as putative novel pharmacological targets. Drug Discov Today 2019; 24:2192-2201. [DOI: 10.1016/j.drudis.2019.09.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 08/15/2019] [Accepted: 09/05/2019] [Indexed: 12/14/2022]
|
33
|
O'Mara TA, Spurdle AB, Glubb DM. Analysis of Promoter-Associated Chromatin Interactions Reveals Biologically Relevant Candidate Target Genes at Endometrial Cancer Risk Loci. Cancers (Basel) 2019; 11:cancers11101440. [PMID: 31561579 PMCID: PMC6826789 DOI: 10.3390/cancers11101440] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/19/2019] [Accepted: 09/23/2019] [Indexed: 02/06/2023] Open
Abstract
The identification of target genes at genome-wide association study (GWAS) loci is a major obstacle for GWAS follow-up. To identify candidate target genes at the 16 known endometrial cancer GWAS risk loci, we performed HiChIP chromatin looping analysis of endometrial cell lines. To enrich for enhancer-promoter interactions, a mechanism through which GWAS variation may target genes, we captured chromatin loops associated with H3K27Ac histone, characteristic of promoters and enhancers. Analysis of HiChIP loops contacting promoters revealed enrichment for endometrial cancer GWAS heritability and intersection with endometrial cancer risk variation identified 103 HiChIP target genes at 13 risk loci. Expression of four HiChIP target genes (SNX11, SRP14, HOXB2 and BCL11A) was associated with risk variation, providing further evidence for their targeting. Network analysis functionally prioritized a set of proteins that interact with those encoded by HiChIP target genes, and this set was enriched for pan-cancer and endometrial cancer drivers. Lastly, HiChIP target genes and prioritized interacting proteins were over-represented in pathways related to endometrial cancer development. In summary, we have generated the first global chromatin looping data from normal and tumoral endometrial cells, enabling analysis of all known endometrial cancer risk loci and identifying biologically relevant candidate target genes.
Collapse
Affiliation(s)
- Tracy A O'Mara
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane QLD 4006, Australia.
| | - Amanda B Spurdle
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane QLD 4006, Australia.
| | - Dylan M Glubb
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane QLD 4006, Australia.
| |
Collapse
|
34
|
Fang Q, Tian M, Wang F, Zhang Z, Du T, Wang W, Yang Y, Li X, Chen G, Xiao L, Wei H, Wang Y, Chen C, Wang DW. Amlodipine induces vasodilation via Akt2/Sp1-activated miR-21 in smooth muscle cells. Br J Pharmacol 2019; 176:2306-2320. [PMID: 30927374 DOI: 10.1111/bph.14679] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 02/15/2019] [Accepted: 03/13/2019] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE The calcium antagonist amlodipine exerts important cardioprotective effects by modulating smooth muscle and endothelial functions. However, the mechanisms underlying these effects are incompletely understood. EXPERIMENTAL APPROACH Western blotting was used to compare the expression of key genes involved in vascular smooth muscle cell (VSMC) phenotype conversion. Recombinant adeno-associated virus system was used to regulate miRNA expression in rats via tail vein. Bioinformatics was used to predict the transcriptional regulation of miR-21 upstream followed by biochemical validation using quantitative real-time polymerase chain reaction, ChIP-qPCR and EMSA assays. KEY RESULTS Only the calcium antagonist amlodipine, and no other type of anti-hypertensive drug, induced miR-21 overexpression in plasma and aortic vessels in the animal model. Real-time PCR and luciferase assays showed that amlodipine induced miR-21 overexpression in vascular smooth muscle cells. Western blot and immunofluorescence assays demonstrated that amlodipine activated Akt2, rather than Akt1, followed by activation of transcription factor Sp1, which regulated VSMC phenotype conversion via binding to the miR-21 promoter. Furthermore, bioinformatic analyses and luciferase assays demonstrated that amlodipine activated miR-21 transcription at the -2034/-2027 Sp1-binding site, which was further demonstrated by ChIP-qPCR and EMSA assays. Consistently, small-interfering RNA-mediated knockdown of Akt2 and Sp1 significantly attenuated the effects of amlodipine on miR-21 expression in smooth muscle cells. CONCLUSION AND IMPLICATIONS These results indicate that amlodipine induces smooth muscle cell differentiation via miR-21, which is regulated by p-Akt2 and Sp1 nuclear translocation, thereby providing a novel target for cardiovascular diseases.
Collapse
Affiliation(s)
- Qin Fang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Min Tian
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Wang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zhihao Zhang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Tingyi Du
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Wang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yong Yang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xianqing Li
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Guangzhi Chen
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Lei Xiao
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Haoran Wei
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Yan Wang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chen Chen
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Dao Wen Wang
- Division of Cardiology, Department of Internal Medicine, Hubei Key Laboratory of Genetics and Molecular Mechanisms of Cardiological Disorders, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Heteromerization fingerprints between bradykinin B2 and thromboxane TP receptors in native cells. PLoS One 2019; 14:e0216908. [PMID: 31086419 PMCID: PMC6516669 DOI: 10.1371/journal.pone.0216908] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 04/30/2019] [Indexed: 12/12/2022] Open
Abstract
Bradykinin (BK) and thromboxane-A2 (TX-A2) are two vasoactive mediators that modulate vascular tone and inflammation via binding to their cognate "class A" G-protein coupled receptors (GPCRs), BK-B2 receptors (B2R) and TX-prostanoid receptors (TP), respectively. Both BK and TX-A2 lead to ERK1/2-mediated vascular smooth muscle cell (VSMC) proliferation and/or hypertrophy. While each of B2R and TP could form functional dimers with various GPCRs, the likelihood that B2R-TP heteromerization could contribute to their co-regulation has never been investigated. The main objective of this study was to investigate the mode of B2R and TP interaction in VSMC, and its possible impact on downstream signaling. Our findings revealed synergistically activated ERK1/2 following co-stimulation of rat VSMC with a subthreshold dose of BK and effective doses of the TP stable agonist, IBOP, possibly involving biased agonist signaling. Single detection of each of B2R and TP in VSMC, using in-situ proximity ligation assay (PLA), provided evidence of the constitutive expression of nuclear and extranuclear B2R and TP. Moreover, inspection of B2R-TP PLA signals in VSMC revealed agonist-modulated nuclear and extranuclear proximity between B2R and TP, whose quantification varied substantially following single versus dual agonist stimulations. B2R-TP interaction was further verified by the findings of co-immunoprecipitation (co-IP) analysis of VSMC lysates. To our knowledge, this is the first study that provides evidence supporting the existence of B2R-TP heteromerization fingerprints in primary cultured VSMC.
Collapse
|
36
|
Bencheikh L, Diop MK, Rivière J, Imanci A, Pierron G, Souquere S, Naimo A, Morabito M, Dussiot M, De Leeuw F, Lobry C, Solary E, Droin N. Dynamic gene regulation by nuclear colony-stimulating factor 1 receptor in human monocytes and macrophages. Nat Commun 2019; 10:1935. [PMID: 31028249 PMCID: PMC6486619 DOI: 10.1038/s41467-019-09970-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 04/10/2019] [Indexed: 02/06/2023] Open
Abstract
Despite their location at the cell surface, several receptor tyrosine kinases (RTK) are also found in the nucleus, as either intracellular domains or full length proteins. However, their potential nuclear functions remain poorly understood. Here we find that a fraction of full length Colony Stimulating Factor-1 Receptor (CSF-1R), an RTK involved in monocyte/macrophage generation, migrates to the nucleus upon CSF-1 stimulation in human primary monocytes. Chromatin-immunoprecipitation identifies the preferential recruitment of CSF-1R to intergenic regions, where it co-localizes with H3K4me1 and interacts with the transcription factor EGR1. When monocytes are differentiated into macrophages with CSF-1, CSF-1R is redirected to transcription starting sites, colocalizes with H3K4me3, and interacts with ELK and YY1 transcription factors. CSF-1R expression and chromatin recruitment is modulated by small molecule CSF-1R inhibitors and altered in monocytes from chronic myelomonocytic leukemia patients. Unraveling this dynamic non-canonical CSF-1R function suggests new avenues to explore the poorly understood functions of this receptor and its ligands. Receptor tyrosine kinases localize to the cell surface and have been suggested to also have nuclear function. Here the authors provide evidence that Colony Stimulating Factor-1 Receptor (CSF-1R) migrates to the nucleus upon CSF-1 stimulation in monocytes and that upon differentiation into macrophages, CSF-1R localizes to TSS, co-localizes with H3K4me3, and interacts with ELK and YY1.
Collapse
Affiliation(s)
- Laura Bencheikh
- INSERM U1170, Gustave Roussy Cancer Center, 94805, Villejuif, France.,Faculté de Médecine, Université Paris-Sud, 94270, Le Kremlin-Bicêtre, France
| | | | - Julie Rivière
- INSERM U1170, Gustave Roussy Cancer Center, 94805, Villejuif, France
| | - Aygun Imanci
- INSERM U1170, Gustave Roussy Cancer Center, 94805, Villejuif, France.,Faculté de Médecine, Université Paris-Sud, 94270, Le Kremlin-Bicêtre, France
| | - Gerard Pierron
- CNRS UMR9196, Gustave Roussy Cancer Center, 94805, Villejuif, France
| | - Sylvie Souquere
- CNRS UMR9196, Gustave Roussy Cancer Center, 94805, Villejuif, France
| | - Audrey Naimo
- INSERM US23, CNRS UMS 3655, AMMICa, Genomic platform, Gustave Roussy Cancer Center, 94805, Villejuif, France
| | - Margot Morabito
- INSERM U1170, Gustave Roussy Cancer Center, 94805, Villejuif, France
| | - Michaël Dussiot
- INSERM U1163, CNRS UMR8254, Institut Imagine, Hôpital Necker Enfants Malades, 75015, Paris, France.,Institut Imagine, Hôpital Necker Enfants Malades, Université Sorbonne-Paris-Cité, 75015, Paris, France.,Laboratoire d'excellence GR-Ex, Institut Imagine, Hôpital Necker Enfants Malades, 75015, Paris, France
| | - Frédéric De Leeuw
- INSERM US23, CNRS UMS 3655, AMMICa, Imaging and Cytometry Platform, Gustave Roussy Cancer Center, 94805, Villejuif, France
| | - Camille Lobry
- INSERM U1170, Gustave Roussy Cancer Center, 94805, Villejuif, France.,Faculté de Médecine, Université Paris-Sud, 94270, Le Kremlin-Bicêtre, France
| | - Eric Solary
- INSERM U1170, Gustave Roussy Cancer Center, 94805, Villejuif, France. .,Faculté de Médecine, Université Paris-Sud, 94270, Le Kremlin-Bicêtre, France. .,Department of Hematology, Gustave Roussy Cancer Center, 94805, Villejuif, France.
| | - Nathalie Droin
- INSERM U1170, Gustave Roussy Cancer Center, 94805, Villejuif, France. .,Faculté de Médecine, Université Paris-Sud, 94270, Le Kremlin-Bicêtre, France. .,INSERM US23, CNRS UMS 3655, AMMICa, Genomic platform, Gustave Roussy Cancer Center, 94805, Villejuif, France.
| |
Collapse
|
37
|
Protease-activated receptor 2 protects against VEGF inhibitor-induced glomerular endothelial and podocyte injury. Sci Rep 2019; 9:2986. [PMID: 30814628 PMCID: PMC6393426 DOI: 10.1038/s41598-019-39914-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 01/31/2019] [Indexed: 02/02/2023] Open
Abstract
Vascular endothelial growth factor (VEGF) inhibitors cause glomerular injury. We have recently shown that activation of protease-activated receptor 2 (PAR2) by factor Xa exacerbated diabetic kidney disease. However, the role of PAR2 in glomerular injury induced by VEGF blockade is not known. Herein, we investigated the effect of the lack of PAR2 on VEGF inhibitor-induced glomerular injury. Although administering an anti-VEGF antibody by itself did not show renal phenotype in wild type mice, its administration to mice lacking endothelial nitric oxide synthase (eNOS) caused glomerular injury. Different from what we expected, administration of an anti-VEGF antibody in mice lacking PAR2 and eNOS exacerbated albuminuria and reduced the expression levels of CD31, pro-angiogenic VEGF, and angiogenesis-related chemokines in their kidneys. Podocyte injury was also evident in this model of mice lacking PAR2. Our results suggest that PAR2 is protective against VEGF inhibitor-induced glomerular endothelial and podocyte injury.
Collapse
|
38
|
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
|
39
|
Beaudry-Richard A, Nadeau-Vallée M, Prairie É, Maurice N, Heckel É, Nezhady M, Pundir S, Madaan A, Boudreault A, Hou X, Quiniou C, Sierra EM, Beaulac A, Lodygensky G, Robertson SA, Keelan J, Adams Waldorf KM, Olson DM, Rivera JC, Lubell WD, Joyal JS, Bouchard JF, Chemtob S. Antenatal IL-1-dependent inflammation persists postnatally and causes retinal and sub-retinal vasculopathy in progeny. Sci Rep 2018; 8:11875. [PMID: 30089839 PMCID: PMC6082873 DOI: 10.1038/s41598-018-30087-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 07/23/2018] [Indexed: 12/13/2022] Open
Abstract
Antenatal inflammation as seen with chorioamnionitis is harmful to foetal/neonatal organ development including to eyes. Although the major pro-inflammatory cytokine IL-1β participates in retinopathy induced by hyperoxia (a predisposing factor to retinopathy of prematurity), the specific role of antenatal IL-1β associated with preterm birth (PTB) in retinal vasculopathy (independent of hyperoxia) is unknown. Using a murine model of PTB induced with IL-1β injection in utero, we studied consequent retinal and choroidal vascular development; in this process we evaluated the efficacy of IL-1R antagonists. Eyes of foetuses exposed only to IL-1β displayed high levels of pro-inflammatory genes, and a persistent postnatal infiltration of inflammatory cells. This prolonged inflammatory response was associated with: (1) a marked delay in retinal vessel growth; (2) long-lasting thinning of the choroid; and (3) long-term morphological and functional alterations of the retina. Antenatal administration of IL-1R antagonists - 101.10 (a modulator of IL-1R) more so than Kineret (competitive IL-1R antagonist) - prevented all deleterious effects of inflammation. This study unveils a key role for IL-1β, a major mediator of chorioamnionitis, in causing sustained ocular inflammation and perinatal vascular eye injury, and highlights the efficacy of antenatal 101.10 to suppress deleterious inflammation.
Collapse
Affiliation(s)
- Alexandra Beaudry-Richard
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Mathieu Nadeau-Vallée
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
- Department of Pharmacology, Université de Montréal, Montréal, Canada
| | - Élizabeth Prairie
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Noémie Maurice
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Émilie Heckel
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Mohammad Nezhady
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Sheetal Pundir
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Ankush Madaan
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Amarilys Boudreault
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Xin Hou
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Christiane Quiniou
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Estefania Marin Sierra
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | - Alexandre Beaulac
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Gregory Lodygensky
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
| | - Sarah A Robertson
- Department of Obstetrics and Gynaecology, University of Adelaide, Adelaide, South Australia, 5005, Australia
| | - Jeffrey Keelan
- Div Obstetrics & Gynaecology, University of Western Australia King Edward Memorial Hospital, Perth, Australia
| | | | - David M Olson
- Departments of Obstetrics and Gynaecology, Pediatrics and Physiology, University of Alberta, Edmonton, AB, Canada
| | - Jose-Carlos Rivera
- Department of Ophthalmology, Maisonneuve-Rosemont Hospital Research Centre, Montréal, Canada
| | - William D Lubell
- Department of Chemistry, Université de Montréal, Montréal, Canada
| | - Jean-Sebastien Joyal
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada
- Department of Pharmacology, Université de Montréal, Montréal, Canada
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada
| | | | - Sylvain Chemtob
- Departments of Pediatrics, Ophthalmology and Pharmacology, CHU Sainte-Justine Research Centre, Montréal, Canada.
- Department of Pharmacology, Université de Montréal, Montréal, Canada.
- Department of Pharmacology and Therapeutics, McGill University, Montréal, Canada.
| |
Collapse
|
40
|
Paredes I, Himmels P, Ruiz de Almodóvar C. Neurovascular Communication during CNS Development. Dev Cell 2018; 45:10-32. [PMID: 29634931 DOI: 10.1016/j.devcel.2018.01.023] [Citation(s) in RCA: 151] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 11/22/2017] [Accepted: 01/08/2018] [Indexed: 12/11/2022]
Abstract
A precise communication between the nervous and the vascular systems is crucial for proper formation and function of the central nervous system (CNS). Interestingly, this communication does not only occur by neural cells regulating the growth and properties of the vasculature, but new studies show that blood vessels actively control different neurodevelopmental processes. Here, we review the current knowledge on how neurons in particular influence growing blood vessels during CNS development and on how vessels participate in shaping the neural compartment. We also review the identified molecular mechanisms of this bidirectional communication.
Collapse
Affiliation(s)
- Isidora Paredes
- Biochemistry Center, Heidelberg University, 69120 Heidelberg, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Patricia Himmels
- Biochemistry Center, Heidelberg University, 69120 Heidelberg, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany
| | - Carmen Ruiz de Almodóvar
- Biochemistry Center, Heidelberg University, 69120 Heidelberg, Germany; Interdisciplinary Center for Neurosciences, Heidelberg University, 69120 Heidelberg, Germany.
| |
Collapse
|
41
|
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
|
42
|
Wang W, Qiao Y, Li Z. New Insights into Modes of GPCR Activation. Trends Pharmacol Sci 2018; 39:367-386. [DOI: 10.1016/j.tips.2018.01.001] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
|
43
|
Fortmann SD, Rao SV, Tanguay JF, Lordkipanidze M, Hanley DF, Can M, Kim MH, Marciniak TA, Serebruany VL. Vorapaxar and diplopia: Possible off-target PAR-receptor mismodulation. Thromb Haemost 2017; 115:905-10. [DOI: 10.1160/th15-11-0882] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/05/2016] [Indexed: 11/05/2022]
Abstract
SummaryVorapaxar, a novel antiplatelet thrombin PAR-1 inhibitor, has been evaluated in the successful TRA2P trial and the failed TRACER trial. The drug is currently approved for post myocardial infarction and peripheral artery disease indications with concomitant use of clopidogrel and/or aspirin. The FDA ruled that the vorapaxar safety profile is acceptable. However, both trials revealed excess diplopia (double vision) usually reversible after vorapaxar. The diplopia risk appears to be small (about 1 extra case per 1,000 treated subjects), but real. Overall, there were 10 placebo and 34 vorapaxar diplopia cases (p=0.018) consistent for TRACER (2 vs 13 cases; p=0.010) and for TRA2P (8 vs 21 cases; p=0.018). Hence, we review the FDA-confirmed evidence and discuss potential causes and implications of such a surprising adverse association, which may be related to off-target PAR receptor mismodulation in the eye.
Collapse
|
44
|
Joyal JS, Gantner ML, Smith LEH. Retinal energy demands control vascular supply of the retina in development and disease: The role of neuronal lipid and glucose metabolism. Prog Retin Eye Res 2017; 64:131-156. [PMID: 29175509 DOI: 10.1016/j.preteyeres.2017.11.002] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/11/2017] [Accepted: 11/15/2017] [Indexed: 12/15/2022]
Affiliation(s)
- Jean-Sébastien Joyal
- Department of Pediatrics, Pharmacology and Ophthalmology, CHU Sainte-Justine Research Center, Université de Montréal, Montreal, Qc, Canada; Department of Pharmacology and Therapeutics, McGill University, Montreal, Qc, Canada.
| | - Marin L Gantner
- The Lowy Medical Research Institute, La Jolla, United States
| | - Lois E H Smith
- Department of Ophthalmology, Harvard Medical School, Boston Children's Hospital, 300 Longwood Avenue, Boston MA 02115, United States.
| |
Collapse
|
45
|
Retinal vasculature development in health and disease. Prog Retin Eye Res 2017; 63:1-19. [PMID: 29129724 DOI: 10.1016/j.preteyeres.2017.11.001] [Citation(s) in RCA: 225] [Impact Index Per Article: 28.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Revised: 11/02/2017] [Accepted: 11/06/2017] [Indexed: 12/17/2022]
Abstract
Development of the retinal vasculature is based on highly coordinated signalling between different cell types of the retina, integrating internal metabolic requirements with external influences such as the supply of oxygen and nutrients. The developing mouse retinal vasculature is a useful model system to study these interactions because it is experimentally accessible for intra ocular injections and genetic manipulations, can be easily imaged and develops in a similar fashion to that of humans. Research using this model has provided insights about general principles of angiogenesis as well as pathologies that affect the developing retinal vasculature. In this review, we discuss recent advances in our understanding of the molecular and cellular mechanisms that govern the interactions between neurons, glial and vascular cells in the developing retina. This includes a review of mechanisms that shape the retinal vasculature, such as sprouting angiogenesis, vascular network remodelling and vessel maturation. We also explore how the disruption of these processes in mice can lead to pathology - such as oxygen induced retinopathy - and how this translates to human retinopathy of prematurity.
Collapse
|
46
|
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: 85] [Impact Index Per Article: 10.6] [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
|
47
|
Grajales-Esquivel E, Luz-Madrigal A, Bierly J, Haynes T, Reis ES, Han Z, Gutierrez C, McKinney Z, Tzekou A, Lambris JD, Tsonis PA, Del Rio-Tsonis K. Complement component C3aR constitutes a novel regulator for chick eye morphogenesis. Dev Biol 2017; 428:88-100. [PMID: 28576690 PMCID: PMC5726978 DOI: 10.1016/j.ydbio.2017.05.019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/05/2016] [Accepted: 05/17/2017] [Indexed: 12/22/2022]
Abstract
Complement components have been implicated in a wide variety of functions including neurogenesis, proliferation, cell migration, differentiation, cancer, and more recently early development and regeneration. Following our initial observations indicating that C3a/C3aR signaling induces chick retina regeneration, we analyzed its role in chick eye morphogenesis. During eye development, the optic vesicle (OV) invaginates to generate a bilayer optic cup (OC) that gives rise to the retinal pigmented epithelium (RPE) and neural retina. We show by immunofluorescence staining that C3 and the receptor for C3a (the cleaved and active form of C3), C3aR, are present in chick embryos during eye morphogenesis in the OV and OC. Interestingly, C3aR is mainly localized in the nuclear compartment at the OC stage. Loss of function studies at the OV stage using morpholinos or a blocking antibody targeting the C3aR (anti-C3aR Ab), causes eye defects such as microphthalmia and defects in the ventral portion of the eye that result in coloboma. Such defects were not observed when C3aR was disrupted at the OC stage. Histological analysis demonstrated that microphthalmic eyes were unable to generate a normal optic stalk or a closed OC. The dorsal/ventral patterning defects were accompanied by an expansion of the ventral markers Pax2, cVax and retinoic acid synthesizing enzyme raldh-3 (aldh1a3) domains, an absence of the dorsal expression of Tbx5 and raldh-1 (aldh1a1) and a re-specification of the ventral RPE to neuroepithelium. In addition, the eyes showed overall decreased expression of Gli1 and a change in distribution of nuclear β-catenin, suggesting that Shh and Wnt pathways have been affected. Finally, we observed prominent cell death along with a decrease in proliferating cells, indicating that both processes contribute to the microphthalmic phenotype. Together our results show that C3aR is necessary for the proper morphogenesis of the OC. This is the first report implicating C3aR in eye development, revealing an unsuspected hitherto regulator for proper chick eye morphogenesis.
Collapse
Affiliation(s)
- Erika Grajales-Esquivel
- Department of Biology, Miami University and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA.
| | - Agustin Luz-Madrigal
- Department of Biology, Miami University and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA; Department of Biology, University of Dayton and Center for Tissue Regeneration and Engineering at the University of Dayton (TREND), Dayton, OH 45469, USA.
| | - Jeffrey Bierly
- Department of Biology, Miami University and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA.
| | - Tracy Haynes
- Department of Biology, Miami University and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA.
| | - Edimara S Reis
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Zeyu Han
- Department of Biology, Miami University and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA.
| | - Christian Gutierrez
- Department of Biology, Miami University and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA.
| | - Zachary McKinney
- Department of Biology, Miami University and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA.
| | - Apostolia Tzekou
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - John D Lambris
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Panagiotis A Tsonis
- Department of Biology, University of Dayton and Center for Tissue Regeneration and Engineering at the University of Dayton (TREND), Dayton, OH 45469, USA.
| | - Katia Del Rio-Tsonis
- Department of Biology, Miami University and Center for Visual Sciences at Miami University (CVSMU), Oxford, OH 45056, USA.
| |
Collapse
|
48
|
Toivanen PI, Nieminen T, Laakkonen JP, Heikura T, Kaikkonen MU, Ylä-Herttuala S. Snake venom VEGF Vammin induces a highly efficient angiogenic response in skeletal muscle via VEGFR-2/NRP specific signaling. Sci Rep 2017; 7:5525. [PMID: 28717175 PMCID: PMC5514023 DOI: 10.1038/s41598-017-05876-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 06/05/2017] [Indexed: 01/13/2023] Open
Abstract
Vascular Endothelial Growth Factors (VEGFs) are promising molecules for the treatment of ischemic diseases by pro-angiogenic therapy. Snake venom VEGFs are a novel subgroup with unique receptor binding profiles and as such are potential new therapeutic agents. We determined the ligand-receptor interactions, gene regulation and angiogenic properties of Vipera ammodytes venom VEGF, Vammin, and compared it to the canonical angiogenic factor VEGF-A to evaluate the use of Vammin for therapeutic angiogenesis. Vammin efficiently induced VEGFR-2 mediated proliferation and expression of genes associated with proliferation, migration and angiogenesis. VEGF-A165 and especially VEGF-A109 induced less pronounced effects. Vammin regulates a number of signaling pathways by inducing the expression of NR4A family nuclear receptors and regulators of calcium signaling and MAP kinase pathways. Interestingly, MARC1, which encodes an enzyme discovered to catalyze reduction of nitrate to NO, was identified as a novel VEGFR-2 regulated gene. In rabbit skeletal muscle adenoviral delivery of Vammin induced prominent angiogenic responses. Both the vector dose and the co-receptor binding of the ligand were critical parameters controlling the type of angiogenic response from sprouting angiogenesis to vessel enlargement. Vammin induced VEGFR-2/NRP-1 mediated signaling more effectively than VEGF-A, consequently it is a promising candidate for development of pro-angiogenic therapies.
Collapse
Affiliation(s)
- Pyry I Toivanen
- Department of Biotechnology and Molecular Medicine, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tiina Nieminen
- Department of Biotechnology and Molecular Medicine, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Johanna P Laakkonen
- Department of Biotechnology and Molecular Medicine, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Tommi Heikura
- Department of Biotechnology and Molecular Medicine, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Minna U Kaikkonen
- Department of Biotechnology and Molecular Medicine, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Seppo Ylä-Herttuala
- Department of Biotechnology and Molecular Medicine, A.I.Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland. .,Heart Center and Gene Therapy Unit, Kuopio University Hospital, Kuopio, Finland.
| |
Collapse
|
49
|
Hartnett ME. Advances in understanding and management of retinopathy of prematurity. Surv Ophthalmol 2017; 62:257-276. [PMID: 28012875 PMCID: PMC5401801 DOI: 10.1016/j.survophthal.2016.12.004] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 12/11/2016] [Accepted: 12/12/2016] [Indexed: 12/16/2022]
Abstract
The understanding, diagnosis, and treatment of retinopathy of prematurity have changed in the 70 years since the original description of retrolental fibroplasia associated with high oxygenation. It is now recognized that retinopathy of prematurity differs in appearance worldwide and as ever smaller and younger premature infants survive. New methods are being evaluated to image the retina, diagnose severe retinopathy of prematurity, and determine windows of time for treatment to save eyes and improve visual and neural outcomes. New treatments to promote physiologic retinal vascular development, vascular repair, and inhibit vasoproliferation by regulating proteins involved in vascular endothelial growth factor, insulin-like growth factor, or erythropoietin signaling. Reducing excessive oxidative/nitrosative stress and understanding progenitor cells and neurovascular and glial vascular interactions are being studied.
Collapse
Affiliation(s)
- Mary Elizabeth Hartnett
- Department of Ophthalmology and Visual Sciences, Moran Eye Center, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
50
|
Bhosle VK, Rivera JC, Chemtob S. New insights into mechanisms of nuclear translocation of G-protein coupled receptors. Small GTPases 2017; 10:254-263. [PMID: 28125336 DOI: 10.1080/21541248.2017.1282402] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The G-protein coupled receptor (GPCR) signaling was long believed to involve activation of receptor exclusively at the cell surface, followed by its binding to heterotrimeric G-proteins and arrestins to trigger various intracellular signaling cascades, and termination of signaling by internalization of the receptor. It is now accepted that many GPCRs continue to signal after internalization in the endosomes. Since the breakthrough discoveries of nuclear binding sites for their ligands in 1980s, several GPCRs have been detected at cell nuclei. But mechanisms of nuclear localization of GPCRs, many of whom contain putative nuclear localization signals, remain poorly understood to date. Nevertheless, it is known that subcellular trafficking of GPCRs is regulated by members of Ras superfamily of small GTPases, most notably by Rab and Arf GTPases. In this commentary, we highlight several recent studies which suggest novel roles of small GTPases, importins and sorting nexin proteins in the nuclear translocation of GPCRs via vesicular transport pathways. Taken together with increasing evidence for in vivo functionality of the nuclear GPCRs, better understanding of their trafficking will provide valuable clues in cell biology.
Collapse
Affiliation(s)
- Vikrant K Bhosle
- a Department of Pharmacology and Therapeutics , McGill University , Montréal , Québec , Canada.,b CHU Sainte-Justine Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,c Maisonneuve-Rosemont Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,e Cell Biology Program , Peter Gilgan Centre for Research and Learning , Toronto , Ontario , Canada
| | - José Carlos Rivera
- b CHU Sainte-Justine Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,c Maisonneuve-Rosemont Hospital Research Centre , University of Montréal , Montréal , Québec , Canada
| | - Sylvain Chemtob
- a Department of Pharmacology and Therapeutics , McGill University , Montréal , Québec , Canada.,b CHU Sainte-Justine Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,c Maisonneuve-Rosemont Hospital Research Centre , University of Montréal , Montréal , Québec , Canada.,d Departments of Pediatrics, Ophthalmology and Pharmacology , University of Montréal , Montréal , Québec , Canada
| |
Collapse
|