51
|
Nickoloff-Bybel EA, Festa L, Meucci O, Gaskill PJ. Co-receptor signaling in the pathogenesis of neuroHIV. Retrovirology 2021; 18:24. [PMID: 34429135 PMCID: PMC8385912 DOI: 10.1186/s12977-021-00569-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/11/2021] [Indexed: 12/13/2022] Open
Abstract
The HIV co-receptors, CCR5 and CXCR4, are necessary for HIV entry into target cells, interacting with the HIV envelope protein, gp120, to initiate several signaling cascades thought to be important to the entry process. Co-receptor signaling may also promote the development of neuroHIV by contributing to both persistent neuroinflammation and indirect neurotoxicity. But despite the critical importance of CXCR4 and CCR5 signaling to HIV pathogenesis, there is only one therapeutic (the CCR5 inhibitor Maraviroc) that targets these receptors. Moreover, our understanding of co-receptor signaling in the specific context of neuroHIV is relatively poor. Research into co-receptor signaling has largely stalled in the past decade, possibly owing to the complexity of the signaling cascades and functions mediated by these receptors. Examining the many signaling pathways triggered by co-receptor activation has been challenging due to the lack of specific molecular tools targeting many of the proteins involved in these pathways and the wide array of model systems used across these experiments. Studies examining the impact of co-receptor signaling on HIV neuropathogenesis often show activation of multiple overlapping pathways by similar stimuli, leading to contradictory data on the effects of co-receptor activation. To address this, we will broadly review HIV infection and neuropathogenesis, examine different co-receptor mediated signaling pathways and functions, then discuss the HIV mediated signaling and the differences between activation induced by HIV and cognate ligands. We will assess the specific effects of co-receptor activation on neuropathogenesis, focusing on neuroinflammation. We will also explore how the use of substances of abuse, which are highly prevalent in people living with HIV, can exacerbate the neuropathogenic effects of co-receptor signaling. Finally, we will discuss the current state of therapeutics targeting co-receptors, highlighting challenges the field has faced and areas in which research into co-receptor signaling would yield the most therapeutic benefit in the context of HIV infection. This discussion will provide a comprehensive overview of what is known and what remains to be explored in regard to co-receptor signaling and HIV infection, and will emphasize the potential value of HIV co-receptors as a target for future therapeutic development. ![]()
Collapse
Affiliation(s)
- E A Nickoloff-Bybel
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA
| | - L Festa
- Department of Basic and Translational Sciences, School of Dental Medicine, University of Pennsylvania, 240 S. 40th Street, Philadelphia, PA, 19104, USA
| | - O Meucci
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.,Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
| | - P J Gaskill
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 N. 15th Street, Philadelphia, PA, 19102, USA.
| |
Collapse
|
52
|
Watkins JM, Ross-Elliott TJ, Shan X, Lou F, Dreyer B, Tunc-Ozdemir M, Jia H, Yang J, Oliveira CC, Wu L, Trusov Y, Schwochert TD, Krysan P, Jones AM. Differential regulation of G protein signaling in Arabidopsis through two distinct pathways that internalize AtRGS1. Sci Signal 2021; 14:14/695/eabe4090. [PMID: 34376571 DOI: 10.1126/scisignal.abe4090] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In animals, endocytosis of a seven-transmembrane GPCR is mediated by arrestins to propagate or arrest cytoplasmic G protein-mediated signaling, depending on the bias of the receptor or ligand, which determines how much one transduction pathway is used compared to another. In Arabidopsis thaliana, GPCRs are not required for G protein-coupled signaling because the heterotrimeric G protein complex spontaneously exchanges nucleotide. Instead, the seven-transmembrane protein AtRGS1 modulates G protein signaling through ligand-dependent endocytosis, which initiates derepression of signaling without the involvement of canonical arrestins. Here, we found that endocytosis of AtRGS1 initiated from two separate pools of plasma membrane: sterol-dependent domains and a clathrin-accessible neighborhood, each with a select set of discriminators, activators, and candidate arrestin-like adaptors. Ligand identity (either the pathogen-associated molecular pattern flg22 or the sugar glucose) determined the origin of AtRGS1 endocytosis. Different trafficking origins and trajectories led to different cellular outcomes. Thus, in this system, compartmentation with its associated signalosome architecture drives biased signaling.
Collapse
Affiliation(s)
- Justin M Watkins
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Timothy J Ross-Elliott
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Xiaoyi Shan
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Fei Lou
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bernd Dreyer
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Meral Tunc-Ozdemir
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Haiyan Jia
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Jing Yang
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Celio Cabral Oliveira
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA.,Department of Biochemistry and Molecular Biology/Bioagro, Universidade Federal de Viçosa, Viçosa, MG, Brazil
| | - Luguang Wu
- School of Agriculture and Food Science, University of Queensland, St. Lucia, Queensland Q4072, Australia
| | - Yuri Trusov
- School of Agriculture and Food Science, University of Queensland, St. Lucia, Queensland Q4072, Australia
| | - Timothy D Schwochert
- Department of Chemistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Patrick Krysan
- Department of Horticulture, University of Wisconsin Madison, Madison, WI 53706, USA
| | - Alan M Jones
- Department of Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA. .,Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
53
|
Miningou Zobon NT, Jędrzejewska-Szmek J, Blackwell KT. Temporal pattern and synergy influence activity of ERK signaling pathways during L-LTP induction. eLife 2021; 10:e64644. [PMID: 34374340 PMCID: PMC8363267 DOI: 10.7554/elife.64644] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 08/03/2021] [Indexed: 01/21/2023] Open
Abstract
Long-lasting long-term potentiation (L-LTP) is a cellular mechanism of learning and memory storage. Studies have demonstrated a requirement for extracellular signal-regulated kinase (ERK) activation in L-LTP produced by a diversity of temporal stimulation patterns. Multiple signaling pathways converge to activate ERK, with different pathways being required for different stimulation patterns. To answer whether and how different temporal patterns select different signaling pathways for ERK activation, we developed a computational model of five signaling pathways (including two novel pathways) leading to ERK activation during L-LTP induction. We show that calcium and cAMP work synergistically to activate ERK and that stimuli given with large intertrial intervals activate more ERK than shorter intervals. Furthermore, these pathways contribute to different dynamics of ERK activation. These results suggest that signaling pathways with different temporal sensitivities facilitate ERK activation to diversity of temporal patterns.
Collapse
Affiliation(s)
| | - Joanna Jędrzejewska-Szmek
- Laboratory of Neuroinformatic, Nencki Institute of Experimental Biology of Polish Academy of SciencesWarsawPoland
| | - Kim T Blackwell
- Interdisciplinary Program in Neuroscience, Bioengineering Department, George Mason UniversityFairfaxUnited States
- Krasnow Institute for Advanced Study, George Mason UniversityFairfaxUnited States
| |
Collapse
|
54
|
Liu Y, Shao YT, Ward R, Ma L, Gui HX, Hao Q, Mu X, Yang Y, An S, Guo XX, Xu TR. The C-terminal of the α1b-adreneroceptor is a key determinant for its structure integrity and biological functions. Biosci Biotechnol Biochem 2021; 85:1128-1139. [PMID: 33693487 DOI: 10.1093/bbb/zbab034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 02/19/2021] [Indexed: 12/13/2022]
Abstract
The C-terminal of G protein-coupled receptors is now recognized as being important for G protein activation and signaling function. To detect the role of C-terminal tail in receptor activation, we used the α1b-AR, which has a long C-terminal of 164 amino acids. We constructed the intramolecular FRET sensors, in which the C-terminal was truncated to 10 (∆C-10), 20 (∆C-20), 30 (∆C-30), 50 (∆C-50), 70 (∆C-70), or 90 (∆C-90). The truncated mutants of ∆C-10, ∆C-20, or ∆C-30 cannot induce FRET signal changes and downstream ERK1/2 phosphorylation. However, the truncated mutants of ∆C-50, ∆C-70, or ∆C-90 induce significant FRET signal changes and downstream ERK1/2 phosphorylation, especially ∆C-90. This is particularly true in the case of the ∆C-90, ∆C-70, or ∆C-50 which retained the potential phosphorylation sites (Ser401, Ser404, Ser408, or Ser410). The ∆C-90 showed an increase in agonist-induced FRET signal changes and ERK1/2 phosphorylation in PKC- or endocytosis-dependent and EGFR-, src-, or β-arrestin2-independent.
Collapse
Affiliation(s)
- Ying Liu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China.,Institute of Life Sciences, Jinzhou Medical University, Jinzhou, Liaoning, China
| | - Yu-Ting Shao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Richard Ward
- Centre for Translational Pharmacology, Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Li Ma
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Hao-Xin Gui
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qian Hao
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xi Mu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yang Yang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Su An
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xiao-Xi Guo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Tian-Rui Xu
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
55
|
Subramaniam S, Ruf W, Bosmann M. Advocacy of targeting protease-activated receptors in severe coronavirus disease 2019. Br J Pharmacol 2021; 179:2086-2099. [PMID: 34235728 PMCID: PMC8794588 DOI: 10.1111/bph.15587] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/19/2021] [Accepted: 05/27/2021] [Indexed: 12/23/2022] Open
Abstract
Identifying drug targets mitigating vascular dysfunction, thrombo-inflammation and thromboembolic complications in COVID-19 is essential. COVID-19 coagulopathy differs from sepsis coagulopathy. Factors that drive severe lung pathology and coagulation abnormalities in COVID-19 are not understood. Protein-protein interaction studies indicate that the tagged viral bait protein ORF9c directly interacts with PAR2, which modulates host cell IFN and inflammatory cytokines. In addition to direct interaction of SARS-CoV-2 viral protein with PARs, we speculate that activation of PAR by proteases plays a role in COVID-19-induced hyperinflammation. In COVID-19-associated coagulopathy elevated levels of activated coagulation proteases may cleave PARs in association with TMPRSS2. PARs activation enhances the release of cytokines, chemokines and tissue factor expression to propagate IFN-dependent inflammation, leukocyte-endothelial interaction, vascular permeability and coagulation responses. This hypothesis, corroborated by in vitro findings and emerging clinical evidence, will focus targeted studies of PAR1/2 blockers as adjuvant drugs against cytokine release syndrome and COVID-19-associated coagulopathy.
Collapse
Affiliation(s)
- Saravanan Subramaniam
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany.,Department of Immunology and Microbiology, Scripps Research, La Jolla, California, USA
| | - Markus Bosmann
- Pulmonary Center, Department of Medicine, Boston University School of Medicine, Boston, Massachusetts, USA.,Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
56
|
Ganzen L, Ko MJ, Zhang M, Xie R, Chen Y, Zhang L, James R, Mumm J, van Rijn RM, Zhong W, Pang CP, Zhang M, Tsujikawa M, Leung YF. Drug screening with zebrafish visual behavior identifies carvedilol as a potential treatment for an autosomal dominant form of retinitis pigmentosa. Sci Rep 2021; 11:11432. [PMID: 34075074 PMCID: PMC8169685 DOI: 10.1038/s41598-021-89482-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 04/23/2021] [Indexed: 02/04/2023] Open
Abstract
Retinitis Pigmentosa (RP) is a mostly incurable inherited retinal degeneration affecting approximately 1 in 4000 individuals globally. The goal of this work was to identify drugs that can help patients suffering from the disease. To accomplish this, we screened drugs on a zebrafish autosomal dominant RP model. This model expresses a truncated human rhodopsin transgene (Q344X) causing significant rod degeneration by 7 days post-fertilization (dpf). Consequently, the larvae displayed a deficit in visual motor response (VMR) under scotopic condition. The diminished VMR was leveraged to screen an ENZO SCREEN-WELL REDOX library since oxidative stress is postulated to play a role in RP progression. Our screening identified a beta-blocker, carvedilol, that ameliorated the deficient VMR of the RP larvae and increased their rod number. Carvedilol may directly on rods as it affected the adrenergic pathway in the photoreceptor-like human Y79 cell line. Since carvedilol is an FDA-approved drug, our findings suggest that carvedilol can potentially be repurposed to treat autosomal dominant RP patients.
Collapse
Affiliation(s)
- Logan Ganzen
- grid.169077.e0000 0004 1937 2197Department of Biological Sciences, Purdue University, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Purdue University Life Sciences Program, Purdue University, West Lafayette, IN 47907 USA
| | - Mee Jung Ko
- grid.169077.e0000 0004 1937 2197Purdue University Life Sciences Program, Purdue University, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907 USA
| | - Mengrui Zhang
- grid.213876.90000 0004 1936 738XDepartment of Statistics, University of Georgia, Athens, GA 30602 USA
| | - Rui Xie
- grid.170430.10000 0001 2159 2859Department of Statistics and Data Science, University of Central Florida, Orlando, FL 32816 USA
| | - Yongkai Chen
- grid.213876.90000 0004 1936 738XDepartment of Statistics, University of Georgia, Athens, GA 30602 USA
| | - Liyun Zhang
- grid.21107.350000 0001 2171 9311Wilmer Eye Institute, John Hopkins School of Medicine, Baltimore, MD 21205 USA
| | - Rebecca James
- grid.169077.e0000 0004 1937 2197Department of Biological Sciences, Purdue University, West Lafayette, IN 47907 USA
| | - Jeff Mumm
- grid.21107.350000 0001 2171 9311Wilmer Eye Institute, John Hopkins School of Medicine, Baltimore, MD 21205 USA
| | - Richard M. van Rijn
- grid.169077.e0000 0004 1937 2197Purdue University Life Sciences Program, Purdue University, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Purdue Institute for Integrative Neuroscience, Purdue University, 610 Purdue Mall, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Purdue Institute for Drug Discovery, Purdue University, 610 Purdue Mall, West Lafayette, IN 47907 USA
| | - Wenxuan Zhong
- grid.213876.90000 0004 1936 738XDepartment of Statistics, University of Georgia, Athens, GA 30602 USA
| | - Chi Pui Pang
- grid.10784.3a0000 0004 1937 0482Department of Ophthalmology and Visual Sciences, Chinese University of Hong Kong, Hong Kong, China ,grid.263451.70000 0000 9927 110XJoint Shantou International Eye Center, Shantou University and the Chinese University of Hong Kong, Shantou, China
| | - Mingzhi Zhang
- grid.263451.70000 0000 9927 110XJoint Shantou International Eye Center, Shantou University and the Chinese University of Hong Kong, Shantou, China
| | - Motokazu Tsujikawa
- grid.136593.b0000 0004 0373 3971Department of Ophthalmology, Osaka University Graduate School of Medicine, Osaka, Japan ,grid.136593.b0000 0004 0373 3971Department of Clinical Laboratory and Biomedical Sciences, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuk Fai Leung
- grid.169077.e0000 0004 1937 2197Department of Biological Sciences, Purdue University, West Lafayette, IN 47907 USA ,grid.257413.60000 0001 2287 3919Department of Biochemistry and Molecular Biology, Indiana University School of Medicine Lafayette, 625 Harrison Street, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Purdue Institute for Integrative Neuroscience, Purdue University, 610 Purdue Mall, West Lafayette, IN 47907 USA ,grid.169077.e0000 0004 1937 2197Purdue Institute for Drug Discovery, Purdue University, 610 Purdue Mall, West Lafayette, IN 47907 USA
| |
Collapse
|
57
|
Alhosaini K, Azhar A, Alonazi A, Al-Zoghaibi F. GPCRs: The most promiscuous druggable receptor of the mankind. Saudi Pharm J 2021; 29:539-551. [PMID: 34194261 PMCID: PMC8233523 DOI: 10.1016/j.jsps.2021.04.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/13/2021] [Indexed: 12/11/2022] Open
Abstract
All physiological events in living organisms originated as specific chemical/biochemical signals on the cell surface and transmitted into the cytoplasm. This signal is translated within milliseconds-hours to a specific and unique order required to maintain optimum performance and homeostasis of living organisms. Examples of daily biological functions include neuronal communication and neurotransmission in the process of learning and memory, secretion (hormones, sweat, and saliva), muscle contraction, cellular growth, differentiation and migration during wound healing, and immunity to fight infections. Among the different transducers for such life-dependent signals is the large family of G protein-coupled receptors (GPCRs). GPCRs constitute roughly 800 genes, corresponding to 2% of the human genome. While GPCRs control a plethora of pathophysiological disorders, only approximately one-third of GPCR families have been deorphanized and characterized. Recent drug data show that around 40% of the recommended drugs available in the market target mainly GPCRs. In this review, we presented how such system signals, either through G protein or via other players, independent of G protein, function within the biological system. We also discussed drugs in the market or clinical trials targeting mainly GPCRs in various diseases, including cancer.
Collapse
Key Words
- AC, Adenylyl Cyclase
- Arrestin
- CCR, Chemokine Receptor
- COX, Cyclooxygenase
- DAG, Diacylglycerol
- Drugs
- ERK, Extracellular signal-Regulated Kinase
- G proteins
- GIP, Gastric Inhibitory Peptide
- GLP1R, Glucagon-Like Peptide-1 Receptor
- GPCR
- GRKs
- GRKs, G protein-coupled Receptor Kinases
- Heterodimerization
- IP3, Inositol 1,4,5-triphosphate
- MAPK, Mitogen-Activated Protein Kinase
- NMDA, N-Methyl D-Aspartate
- Nbs, Nanobodies
- PAR-1, Protease Activated Receptor 1
- PIP2, Phosphatidylinositol-4,5-bisphosphate
- PKA, Protein Kinase A
- Signaling
- cAMP, cyclic AMP
Collapse
Affiliation(s)
- Khaled Alhosaini
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Post Box 2457, Riyadh 11451, Saudi Arabia
| | - Asim Azhar
- Interdisciplinary Biotechnology Unit, AMU Aligarh, UP, India
| | - Asma Alonazi
- Department of Pharmacology & Toxicology, College of Pharmacy, King Saud University, Post Box 2457, Riyadh 11451, Saudi Arabia
| | - F Al-Zoghaibi
- Molecular BioMedicine Program, Research Centre, King Faisal Specialist Hospital and Research Centre, P.O.Box: 3354, MBC:03, Riyadh 11211, Saudi Arabia
| |
Collapse
|
58
|
Crilly SE, Ko W, Weinberg ZY, Puthenveedu MA. Conformational specificity of opioid receptors is determined by subcellular location irrespective of agonist. eLife 2021; 10:67478. [PMID: 34013886 PMCID: PMC8208814 DOI: 10.7554/elife.67478] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 05/19/2021] [Indexed: 12/13/2022] Open
Abstract
The prevailing model for the variety in drug responses is that different drugs stabilize distinct active states of their G protein-coupled receptor (GPCR) targets, allowing coupling to different effectors. However, whether the same ligand generates different GPCR active states based on the immediate environment of receptors is not known. Here we address this question using spatially resolved imaging of conformational biosensors that read out distinct active conformations of the δ-opioid receptor (DOR), a physiologically relevant GPCR localized to Golgi and the surface in neuronal cells. We have shown that Golgi and surface pools of DOR both inhibit cAMP, but engage distinct conformational biosensors in response to the same ligand in rat neuroendocrine cells. Further, DOR recruits arrestins on the surface but not on the Golgi. Our results suggest that the local environment determines the active states of receptors for any given drug, allowing GPCRs to couple to different effectors at different subcellular locations.
Collapse
Affiliation(s)
- Stephanie E Crilly
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, United States.,Department of Pharmacology University of Michigan Medical School, Ann Arbor, United States
| | - Wooree Ko
- Department of Pharmacology University of Michigan Medical School, Ann Arbor, United States
| | - Zara Y Weinberg
- Department of Pharmacology University of Michigan Medical School, Ann Arbor, United States
| | - Manojkumar A Puthenveedu
- Cellular and Molecular Biology Program, University of Michigan, Ann Arbor, United States.,Department of Pharmacology University of Michigan Medical School, Ann Arbor, United States
| |
Collapse
|
59
|
Gottesman-Katz L, Latorre R, Vanner S, Schmidt BL, Bunnett NW. Targeting G protein-coupled receptors for the treatment of chronic pain in the digestive system. Gut 2021; 70:970-981. [PMID: 33272979 PMCID: PMC9716638 DOI: 10.1136/gutjnl-2020-321193] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 10/21/2020] [Accepted: 11/07/2020] [Indexed: 12/14/2022]
Abstract
Chronic pain is a hallmark of functional disorders, inflammatory diseases and cancer of the digestive system. The mechanisms that initiate and sustain chronic pain are incompletely understood, and available therapies are inadequate. This review highlights recent advances in the structure and function of pronociceptive and antinociceptive G protein-coupled receptors (GPCRs) that provide insights into the mechanisms and treatment of chronic pain. This knowledge, derived from studies of somatic pain, can guide research into visceral pain. Mediators from injured tissues transiently activate GPCRs at the plasma membrane of neurons, leading to sensitisation of ion channels and acute hyperexcitability and nociception. Sustained agonist release evokes GPCR redistribution to endosomes, where persistent signalling regulates activity of channels and genes that control chronic hyperexcitability and nociception. Endosomally targeted GPCR antagonists provide superior pain relief in preclinical models. Biased agonists stabilise GPCR conformations that favour signalling of beneficial actions at the expense of detrimental side effects. Biased agonists of µ-opioid receptors (MOPrs) can provide analgesia without addiction, respiratory depression and constipation. Opioids that preferentially bind to MOPrs in the acidic microenvironment of diseased tissues produce analgesia without side effects. Allosteric modulators of GPCRs fine-tune actions of endogenous ligands, offering the prospect of refined pain control. GPCR dimers might function as distinct therapeutic targets for nociception. The discovery that GPCRs that control itch also mediate irritant sensation in the colon has revealed new targets. A deeper understanding of GPCR structure and function in different microenvironments offers the potential of developing superior treatments for GI pain.
Collapse
Affiliation(s)
- Lena Gottesman-Katz
- Molecular Pathobiology, New York University, New York, New York, USA,Division of Pediatric Gastroenterology, Columbia University Medical Center/New York Presbyterian, New York, New York, USA
| | - Rocco Latorre
- Molecular Pathobiology, New York University, New York, New York, USA
| | - Stephen Vanner
- Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queens University, Kingston, Ontario, Canada
| | - Brian L Schmidt
- Bluestone Center, New York University, New York, New York, USA
| | - Nigel W Bunnett
- Molecular Pathobiology, New York University, New York, New York, USA
| |
Collapse
|
60
|
Chandrabalan A, Ramachandran R. Molecular mechanisms regulating Proteinase‐Activated Receptors (PARs). FEBS J 2021; 288:2697-2726. [DOI: 10.1111/febs.15829] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/10/2021] [Accepted: 03/18/2021] [Indexed: 12/13/2022]
Affiliation(s)
- Arundhasa Chandrabalan
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology Schulich School of Medicine and Dentistry University of Western Ontario London Canada
| |
Collapse
|
61
|
McMahon DB, Carey RM, Kohanski MA, Adappa ND, Palmer JN, Lee RJ. PAR-2-activated secretion by airway gland serous cells: role for CFTR and inhibition by Pseudomonas aeruginosa. Am J Physiol Lung Cell Mol Physiol 2021; 320:L845-L879. [PMID: 33655758 DOI: 10.1152/ajplung.00411.2020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Airway submucosal gland serous cells are important sites of fluid secretion in conducting airways. Serous cells also express the cystic fibrosis (CF) transmembrane conductance regulator (CFTR). Protease-activated receptor 2 (PAR-2) is a G protein-coupled receptor that activates secretion from intact airway glands. We tested if and how human nasal serous cells secrete fluid in response to PAR-2 stimulation using Ca2+ imaging and simultaneous differential interference contrast imaging to track isosmotic cell shrinking and swelling reflecting activation of solute efflux and influx pathways, respectively. During stimulation of PAR-2, serous cells exhibited dose-dependent increases in intracellular Ca2+. At stimulation levels >EC50 for Ca2+, serous cells simultaneously shrank ∼20% over ∼90 s due to KCl efflux reflecting Ca2+-activated Cl- channel (CaCC, likely TMEM16A)-dependent secretion. At lower levels of PAR-2 stimulation (<EC50 for Ca2+), shrinkage was not evident due to failure to activate CaCC. Low levels of cAMP-elevating VIP receptor (VIPR) stimulation, also insufficient to activate secretion alone, synergized with low-level PAR-2 stimulation to elicit fluid secretion dependent on both cAMP and Ca2+ to activate CFTR and K+ channels, respectively. Polarized cultures of primary serous cells also exhibited synergistic fluid secretion. Pre-exposure to Pseudomonas aeruginosa conditioned media inhibited PAR-2 activation by proteases but not peptide agonists in primary nasal serous cells, Calu-3 bronchial cells, and primary nasal ciliated cells. Disruption of synergistic CFTR-dependent PAR-2/VIPR secretion may contribute to reduced airway surface liquid in CF. Further disruption of the CFTR-independent component of PAR-2-activated secretion by P. aeruginosa may also be important to CF pathophysiology.
Collapse
Affiliation(s)
- Derek B McMahon
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Ryan M Carey
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Michael A Kohanski
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Nithin D Adappa
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - James N Palmer
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Robert J Lee
- Department of Otorhinolaryngology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania.,Department of Physiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
62
|
Smith JS, Pack TF. Noncanonical interactions of G proteins and β‐arrestins: from competitors to companions. FEBS J 2021; 288:2550-2561. [DOI: 10.1111/febs.15749] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 12/02/2020] [Accepted: 02/02/2021] [Indexed: 12/30/2022]
Affiliation(s)
- Jeffrey S. Smith
- Department of Dermatology Massachusetts General Hospital Boston MA USA
- Department of Dermatology Brigham and Women's Hospital Boston MA USA
- Department of Dermatology Beth Israel Deaconess Medical Center Boston MA USA
- Dermatology Program Boston Children's Hospital Boston MA USA
- Harvard Medical School Boston MA USA
| | | |
Collapse
|
63
|
Spinal PAR2 Activation Contributes to Hypersensitivity Induced by Peripheral Inflammation in Rats. Int J Mol Sci 2021; 22:ijms22030991. [PMID: 33498178 PMCID: PMC7863954 DOI: 10.3390/ijms22030991] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 01/16/2021] [Accepted: 01/17/2021] [Indexed: 11/17/2022] Open
Abstract
The mechanisms of inflammatory pain need to be identified in order to find new superior treatments. Protease-activated receptors 2 (PAR2) and transient receptor potential vanilloid 1 (TRPV1) are highly co-expressed in dorsal root ganglion neurons and implicated in pain development. Here, we examined the role of spinal PAR2 in hyperalgesia and the modulation of synaptic transmission in carrageenan-induced peripheral inflammation, using intrathecal (i.t.) treatment in the behavioral experiments and recordings of spontaneous, miniature and dorsal root stimulation-evoked excitatory postsynaptic currents (sEPSCs, mEPSCs and eEPSCs) in spinal cord slices. Intrathecal PAR2-activating peptide (AP) administration aggravated the carrageenan-induced thermal hyperalgesia, and this was prevented by a TRPV1 antagonist (SB 366791) and staurosporine i.t. pretreatment. Additionally, the frequency of the mEPSC and sEPSC and the amplitude of the eEPSC recorded from the superficial dorsal horn neurons were enhanced after acute PAR2 AP application, while prevented with SB 366791 or staurosporine pretreatment. PAR2 antagonist application reduced the thermal hyperalgesia and decreased the frequency of mEPSC and sEPSC and the amplitude of eEPSC. Our findings highlight the contribution of spinal PAR2 activation to carrageenan-induced hyperalgesia and the importance of dorsal horn PAR2 and TRPV1 receptor interactions in the modulation of nociceptive synaptic transmission.
Collapse
|
64
|
Dissecting the structural features of β-arrestins as multifunctional proteins. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140603. [PMID: 33421644 DOI: 10.1016/j.bbapap.2021.140603] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 12/21/2020] [Accepted: 01/04/2021] [Indexed: 02/08/2023]
Abstract
β-arrestins bind active G protein-coupled receptors (GPCRs) and play a crucial role in receptor desensitization and internalization. The classical paradigm of arrestin function has been expanded with the identification of many non-receptor-binding partners, which indicated the multifunctional role of β-arrestins in cellular functions. To elucidate the molecular mechanism of β-arrestin-mediated signaling, the structural features of β-arrestins were investigated using X-ray crystallography and cryogenic electron microscopy (cryo-EM). However, the intrinsic conformational flexibility of β-arrestins hampers the elucidation of structural interactions between β-arrestins and their binding partners using conventional structure determination tools. Therefore, structural information obtained using complementary structure analysis techniques would be necessary in combination with X-ray crystallography and cryo-EM data. In this review, we describe how β-arrestins interact with their binding partners from a structural point of view, as elucidated by both traditional methods (X-ray crystallography and cryo-EM) and complementary structure analysis techniques.
Collapse
|
65
|
Neurokinin receptors and their implications in various autoimmune diseases. CURRENT RESEARCH IN IMMUNOLOGY 2021; 2:66-78. [PMID: 35492389 PMCID: PMC9040085 DOI: 10.1016/j.crimmu.2021.06.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/18/2022] Open
Abstract
Neurokinin receptors belong to the GPCRs family and are ubiquitously expressed throughout the nervous and immune systems. Neurokinin receptors in coordination with neurokinins playing an important role in many physiological processes, including smooth muscle contraction, secretion, proliferation, and nociception. They also contribute to various disease conditions such as inflammatory bowel disease, rheumatoid arthritis, multiple sclerosis, psoriasis, and cancer. Neurokinin receptors antagonist are potent and highly selective and showing success in treating chemotherapy-induced nausea and vomiting. In this review, discuss the various neurokinin receptor expression on immune cells and their importance in various inflammatory and autoimmune diseases and their therapeutic importance. The Neurokinin receptor is an important regulatory mechanism to control the neuronal and immune systems. Various neurokinin receptors (NK1R, NK2R, and NK3R) are expressed in neurons and cells of the immune system. Substance P (SP) controls the differentiation and function of immune cells. SP-NK1R receptor signaling shows substantial cross-talk between neuronal and immune systems in inflammation and autoimmunity.
Collapse
|
66
|
Pierre O, Fouchard M, Buscaglia P, Le Goux N, Leschiera R, Mignen O, Fluhr JW, Misery L, Le Garrec R. Calcium Increase and Substance P Release Induced by the Neurotoxin Brevetoxin-1 in Sensory Neurons: Involvement of PAR2 Activation through Both Cathepsin S and Canonical Signaling. Cells 2020; 9:E2704. [PMID: 33348659 PMCID: PMC7767211 DOI: 10.3390/cells9122704] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/14/2020] [Accepted: 12/15/2020] [Indexed: 12/17/2022] Open
Abstract
Red tides involving Karenia brevis expose humans to brevetoxins (PbTxs). Oral exposition triggers neurotoxic shellfish poisoning, whereas inhalation induces a respiratory syndrome and sensory disturbances. No curative treatment is available and the pathophysiology is not fully elucidated. Protease-activated receptor 2 (PAR2), cathepsin S (Cat-S) and substance P (SP) release are crucial mediators of the sensory effects of ciguatoxins (CTXs) which are PbTx analogs. This work explored the role of PAR2 and Cat-S in PbTx-1-induced sensory effects and deciphered the signaling pathway involved. We performed calcium imaging, PAR2 immunolocalization and SP release experiments in monocultured sensory neurons or co-cultured with keratinocytes treated with PbTx-1 or P-CTX-2. We demonstrated that PbTx-1-induced calcium increase and SP release involved Cat-S, PAR2 and transient receptor potential vanilloid 4 (TRPV4). The PbTx-1-induced signaling pathway included protein kinase A (PKA) and TRPV4, which are compatible with the PAR2 biased signaling induced by Cat-S. Internalization of PAR2 and protein kinase C (PKC), inositol triphosphate receptor and TRPV4 activation evoked by PbTx-1 are compatible with the PAR2 canonical signaling. Our results suggest that PbTx-1-induced sensory disturbances involve the PAR2-TRPV4 pathway. We identified PAR2, Cat-S, PKA, and PKC that are involved in TRPV4 sensitization induced by PbTx-1 in sensory neurons.
Collapse
Affiliation(s)
- Ophélie Pierre
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| | - Maxime Fouchard
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
| | - Paul Buscaglia
- InsermUMR1227, Lymphocytes B et Autoimmunity, University Brest, F-29200 Brest, France; (P.B.); (N.L.G.); (O.M.)
| | - Nelig Le Goux
- InsermUMR1227, Lymphocytes B et Autoimmunity, University Brest, F-29200 Brest, France; (P.B.); (N.L.G.); (O.M.)
| | - Raphaël Leschiera
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| | - Olivier Mignen
- InsermUMR1227, Lymphocytes B et Autoimmunity, University Brest, F-29200 Brest, France; (P.B.); (N.L.G.); (O.M.)
| | - Joachim W. Fluhr
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
- Department of Dermatology and Allergology, Universitaetsmedizin Charit Berlin, D-10117 Berlin, Germany
| | - Laurent Misery
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
- Department of Dermatology, University Hospital of Brest, F-29200 Brest, France
| | - Raphaële Le Garrec
- EA4685 Laboratory of Interactions Neurons-Keratinocytes (LIEN), Faculty of Medicine and Health Sciences, University Brest, F-29200 Brest, France; (M.F.); (R.L.); (J.W.F.); (L.M.); (R.L.G.)
| |
Collapse
|
67
|
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: 50] [Impact Index Per Article: 12.5] [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
|
68
|
Wang M, Thyagarajan B. Pain pathways and potential new targets for pain relief. Biotechnol Appl Biochem 2020; 69:110-123. [PMID: 33316085 DOI: 10.1002/bab.2086] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 12/06/2020] [Indexed: 12/15/2022]
Abstract
Pain is an unpleasant sensory and emotional experience that affects a sizable percentage of people on a daily basis. Sensory neurons known as nociceptors built specifically to detect damaging stimuli can be found throughout the body. They transmit information about noxious stimuli from mechanical, thermal, and chemical sources to the central nervous system and higher brain centers via electrical signals. Nociceptors express various channels and receptors such as voltage-gated sodium and calcium channels, transient receptor potential channels, and opioid receptors that allow them to respond in a highly specific manner to noxious stimuli. Attenuating the pain response can be achieved by inhibiting or altering the expression of these pain targets. Achieving a deeper understanding of how these receptors can be affected at the molecular level can lead to the development of novel pain therapies. This review will discuss the mechanisms of pain, introduce the various receptors that are responsible for detecting pain, and future directions in pharmacological therapies.
Collapse
Affiliation(s)
- Menglan Wang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| | - Baskaran Thyagarajan
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Wyoming, Laramie, WY, USA
| |
Collapse
|
69
|
Crilly SE, Puthenveedu MA. Compartmentalized GPCR Signaling from Intracellular Membranes. J Membr Biol 2020; 254:259-271. [PMID: 33231722 DOI: 10.1007/s00232-020-00158-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [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
|
70
|
Interactions and Signal Transduction Pathways Involved during Central Nervous System Entry by Neisseria meningitidis across the Blood-Brain Barriers. Int J Mol Sci 2020; 21:ijms21228788. [PMID: 33233688 PMCID: PMC7699760 DOI: 10.3390/ijms21228788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 11/18/2020] [Indexed: 11/20/2022] Open
Abstract
The Gram-negative diplococcus Neisseria meningitidis, also called meningococcus, exclusively infects humans and can cause meningitis, a severe disease that can lead to the death of the afflicted individuals. To cause meningitis, the bacteria have to enter the central nervous system (CNS) by crossing one of the barriers protecting the CNS from entry by pathogens. These barriers are represented by the blood–brain barrier separating the blood from the brain parenchyma and the blood–cerebrospinal fluid (CSF) barriers at the choroid plexus and the meninges. During the course of meningococcal disease resulting in meningitis, the bacteria undergo several interactions with host cells, including the pharyngeal epithelium and the cells constituting the barriers between the blood and the CSF. These interactions are required to initiate signal transduction pathways that are involved during the crossing of the meningococci into the blood stream and CNS entry, as well as in the host cell response to infection. In this review we summarize the interactions and pathways involved in these processes, whose understanding could help to better understand the pathogenesis of meningococcal meningitis.
Collapse
|
71
|
Legumain Induces Oral Cancer Pain by Biased Agonism of Protease-Activated Receptor-2. J Neurosci 2020; 41:193-210. [PMID: 33172978 DOI: 10.1523/jneurosci.1211-20.2020] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is one of the most painful cancers, which interferes with orofacial function including talking and eating. We report that legumain (Lgmn) cleaves protease-activated receptor-2 (PAR2) in the acidic OSCC microenvironment to cause pain. Lgmn is a cysteine protease of late endosomes and lysosomes that can be secreted; it exhibits maximal activity in acidic environments. The role of Lgmn in PAR2-dependent cancer pain is unknown. We studied Lgmn activation in human oral cancers and oral cancer mouse models. Lgmn was activated in OSCC patient tumors, compared with matched normal oral tissue. After intraplantar, facial or lingual injection, Lgmn evoked nociception in wild-type (WT) female mice but not in female mice lacking PAR2 in NaV1.8-positive neurons (Par2Nav1.8), nor in female mice treated with a Lgmn inhibitor, LI-1. Inoculation of an OSCC cell line caused mechanical and thermal hyperalgesia that was reversed by LI-1. Par2Nav1.8 and Lgmn deletion attenuated mechanical allodynia in female mice with carcinogen-induced OSCC. Lgmn caused PAR2-dependent hyperexcitability of trigeminal neurons from WT female mice. Par2 deletion, LI-1, and inhibitors of adenylyl cyclase or protein kinase A (PKA) prevented the effects of Lgmn. Under acidified conditions, Lgmn cleaved within the extracellular N terminus of PAR2 at Asn30↓Arg31, proximal to the canonical trypsin activation site. Lgmn activated PAR2 by biased mechanisms in HEK293 cells to induce Ca2+ mobilization, cAMP formation, and PKA/protein kinase D (PKD) activation, but not β-arrestin recruitment or PAR2 endocytosis. Thus, in the acidified OSCC microenvironment, Lgmn activates PAR2 by biased mechanisms that evoke cancer pain.SIGNIFICANCE STATEMENT Oral squamous cell carcinoma (OSCC) is one of the most painful cancers. We report that legumain (Lgmn), which exhibits maximal activity in acidic environments, cleaves protease-activated receptor-2 (PAR2) on neurons to produce OSCC pain. Active Lgmn was elevated in OSCC patient tumors, compared with matched normal oral tissue. Lgmn evokes pain-like behavior through PAR2 Exposure of pain-sensing neurons to Lgmn decreased the current required to generate an action potential through PAR2 Inhibitors of adenylyl cyclase and protein kinase A (PKA) prevented the effects of Lgmn. Lgmn activated PAR2 to induce calcium mobilization, cAMP formation, and activation of protein kinase D (PKD) and PKA, but not β-arrestin recruitment or PAR2 endocytosis. Thus, Lgmn is a biased agonist of PAR2 that evokes cancer pain.
Collapse
|
72
|
Secreted gingipains from Porphyromonas gingivalis induce microglia migration through endosomal signaling by protease-activated receptor 2. Neurochem Int 2020; 140:104840. [DOI: 10.1016/j.neuint.2020.104840] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/20/2020] [Accepted: 08/16/2020] [Indexed: 01/08/2023]
|
73
|
Ocasio-Rivera M, Marin-Maldonado F, Trossi-Torres G, Ortiz-Rosado A, Rodríguez-Irizarry V, Rodriguez-Lopez E, Martínez S, Almodóvar S, Suarez-Martínez E. Targeting of protease activator receptor-2 (PAR-2) antagonist FSLLRY-NH2 as an asthma adjuvant therapy. Medicine (Baltimore) 2020; 99:e22351. [PMID: 33120736 PMCID: PMC7581070 DOI: 10.1097/md.0000000000022351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Asthma is a chronic inflammatory and multifactorial respiratory tract disease. It affects over 18 million adults and 6 million children in the USA with Puerto Ricans showing the highest prevalence (12%-19%). This airways illness can be triggered by an environmental stimulus such as grass pollen, fungi spores, cockroaches allergens, dust mites metabolic compounds, and importantly, by environmental proteases such as trypsin and tryptase. Because of the pivotal role of proteases in the onset of asthma pathophysiology, we focused this study on the serine Protease Activated Receptor-2 (PAR-2), a G-protein-coupled receptor widely expressed in cells across the respiratory tract. Herein, we measured the activation of PAR-2 on primary pulmonary bronchial/tracheal epithelial cells, human small airway epithelial cells, lung bronchial smooth muscle cells (with and without asthma). We tested human-derived eosinophils from 61 Puerto Rican participants (33 asthmatic and 28 non-asthmatic). As surrogate of PAR-2 activation or inhibition we used intracellular calcium mobilization assay. We hypothesized that following exposure of the PAR-2 agonist (AC264613), the studied human primary cell types will increase the mobilization of intracellular calcium levels. In contrast, we expected a decrease of the intracellular calcium levels upon exposure to a PAR-2 antagonist (FSLLRY-NH2). The Puerto Rican-derived eosinophils were analyzed for the proinflammatory markers MAPK/PI3K using flow cytometry (n = 8). As expected, the PAR-2 agonist significantly increased the activation of PAR-2 on the bronchial/tracheal epithelial cells, bronchial smooth muscle cells and human small airway epithelial cells (P = .01). The PAR-2 antagonist significantly decreased the intracellular calcium levels of these lung primary down to undetectable levels (P = .01). Remarkably, the asthmatic-derived eosinophils showed a striking 300% increase of intracellular calcium mobilization suggesting a severe response to the PAR-2 agonist stimuli in asthmatics. In contrast, there were no significant changes between groups after adding the PAR-2 antagonist. Our outcomes revealed that PAR-2 antagonist effectively inhibited the studied primary cells, expecting to decrease the immune response of eosinophils. Most importantly, our results reveal a promising role for the PAR-2 antagonist in targeting bronchial/tracheal epithelial cells, human small airway epithelial cells and bronchial smooth muscle cells with the potential to oblige an asthma adjuvant therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Edu Suarez-Martínez
- University of Puerto Rico-Ponce, Ponce Puerto Rico
- Ponce Health Science University, Ponce Puerto Rico
| |
Collapse
|
74
|
Hendrickx JO, van Gastel J, Leysen H, Martin B, Maudsley S. High-dimensionality Data Analysis of Pharmacological Systems Associated with Complex Diseases. Pharmacol Rev 2020; 72:191-217. [PMID: 31843941 DOI: 10.1124/pr.119.017921] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
It is widely accepted that molecular reductionist views of highly complex human physiologic activity, e.g., the aging process, as well as therapeutic drug efficacy are largely oversimplifications. Currently some of the most effective appreciation of biologic disease and drug response complexity is achieved using high-dimensionality (H-D) data streams from transcriptomic, proteomic, metabolomics, or epigenomic pipelines. Multiple H-D data sets are now common and freely accessible for complex diseases such as metabolic syndrome, cardiovascular disease, and neurodegenerative conditions such as Alzheimer's disease. Over the last decade our ability to interrogate these high-dimensionality data streams has been profoundly enhanced through the development and implementation of highly effective bioinformatic platforms. Employing these computational approaches to understand the complexity of age-related diseases provides a facile mechanism to then synergize this pathologic appreciation with a similar level of understanding of therapeutic-mediated signaling. For informative pathology and drug-based analytics that are able to generate meaningful therapeutic insight across diverse data streams, novel informatics processes such as latent semantic indexing and topological data analyses will likely be important. Elucidation of H-D molecular disease signatures from diverse data streams will likely generate and refine new therapeutic strategies that will be designed with a cognizance of a realistic appreciation of the complexity of human age-related disease and drug effects. We contend that informatic platforms should be synergistic with more advanced chemical/drug and phenotypic cellular/tissue-based analytical predictive models to assist in either de novo drug prioritization or effective repurposing for the intervention of aging-related diseases. SIGNIFICANCE STATEMENT: All diseases, as well as pharmacological mechanisms, are far more complex than previously thought a decade ago. With the advent of commonplace access to technologies that produce large volumes of high-dimensionality data (e.g., transcriptomics, proteomics, metabolomics), it is now imperative that effective tools to appreciate this highly nuanced data are developed. Being able to appreciate the subtleties of high-dimensionality data will allow molecular pharmacologists to develop the most effective multidimensional therapeutics with effectively engineered efficacy profiles.
Collapse
Affiliation(s)
- Jhana O Hendrickx
- Receptor Biology Laboratory, Department of Biomedical Research (J.O.H., J.v.G., H.L., S.M.) and Faculty of Pharmacy, Biomedical and Veterinary Sciences (J.O.H., J.v.G., H.L., B.M., S.M.), University of Antwerp, Antwerp, Belgium
| | - Jaana van Gastel
- Receptor Biology Laboratory, Department of Biomedical Research (J.O.H., J.v.G., H.L., S.M.) and Faculty of Pharmacy, Biomedical and Veterinary Sciences (J.O.H., J.v.G., H.L., B.M., S.M.), University of Antwerp, Antwerp, Belgium
| | - Hanne Leysen
- Receptor Biology Laboratory, Department of Biomedical Research (J.O.H., J.v.G., H.L., S.M.) and Faculty of Pharmacy, Biomedical and Veterinary Sciences (J.O.H., J.v.G., H.L., B.M., S.M.), University of Antwerp, Antwerp, Belgium
| | - Bronwen Martin
- Receptor Biology Laboratory, Department of Biomedical Research (J.O.H., J.v.G., H.L., S.M.) and Faculty of Pharmacy, Biomedical and Veterinary Sciences (J.O.H., J.v.G., H.L., B.M., S.M.), University of Antwerp, Antwerp, Belgium
| | - Stuart Maudsley
- Receptor Biology Laboratory, Department of Biomedical Research (J.O.H., J.v.G., H.L., S.M.) and Faculty of Pharmacy, Biomedical and Veterinary Sciences (J.O.H., J.v.G., H.L., B.M., S.M.), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
75
|
Bao Z, Zhou S, Zhou H. Sorting Nexin 27 as a potential target in G protein‑coupled receptor recycling for cancer therapy (Review). Oncol Rep 2020; 44:1779-1786. [PMID: 33000258 PMCID: PMC7551096 DOI: 10.3892/or.2020.7766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 08/10/2020] [Indexed: 12/12/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are the largest family of membrane receptors and activate several downstream signaling pathways involved in numerous physiological cellular processes. GPCRs are usually internalized and desensitized by intracellular signals. Numerous studies have shown that several GPCRs interact with sorting nexin 27 (SNX27), a cargo selector of the retromer complex, and are recycled from endosomes to the plasma membrane. Recycled GPCRs usually contain specific C-terminal postsynaptic density protein 95/Discs large protein/Zonula occludens 1 (PDZ) binding motifs, which are specifically recognized by SNX27, and return to the cell surface as functionally naïve receptors. Aberrant endosome-to-membrane recycling of GPCRs mediated by SNX27 may serve a critical role in cancer growth and development. Therefore, SNX27 may be a novel target for cancer therapies.
Collapse
Affiliation(s)
- Zixu Bao
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Shijun Zhou
- Department of Infectious Disease, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Haisheng Zhou
- Department of Biochemistry and Molecular Biology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
76
|
PAR2, Keratinocytes, and Cathepsin S Mediate the Sensory Effects of Ciguatoxins Responsible for Ciguatera Poisoning. J Invest Dermatol 2020; 141:648-658.e3. [PMID: 32800876 DOI: 10.1016/j.jid.2020.07.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 01/14/2023]
Abstract
Ciguatera fish poisoning is caused by the consumption of fish contaminated with ciguatoxins (CTXs). The most distressing symptoms are cutaneous sensory disturbances, including cold dysesthesia and itch. CTXs are neurotoxins known to activate voltage-gated sodium channels, but no specific treatment exists. Peptidergic neurons have been critically involved in ciguatera fish poisoning sensory disturbances. Protease-activated receptor-2 (PAR2) is an itch- and pain-related G protein‒coupled receptor whose activation leads to a calcium-dependent neuropeptide release. In this study, we studied the role of voltage-gated sodium channels, PAR2, and the PAR2 agonist cathepsin S in the cytosolic calcium increase and subsequent release of the neuropeptide substance P elicited by Pacific CTX-2 (P-CTX-2) in rat sensory neurons and human epidermal keratinocytes. In sensory neurons, the P-CTX-2‒evoked calcium response was driven by voltage-gated sodium channels and PAR2-dependent mechanisms. In keratinocytes, P-CTX-2 also induced voltage-gated sodium channels and PAR2-dependent marked calcium response. In the cocultured cells, P-CTX-2 significantly increased cathepsin S activity, and cathepsin S and PAR2 antagonists almost abolished P-CTX-2‒elicited substance P release. Keratinocytes synergistically favored the induced substance P release. Our results demonstrate that the sensory effects of CTXs involve the cathepsin S-PAR2 pathway and are potentiated by their direct action on nonexcitable keratinocytes through the same pathway.
Collapse
|
77
|
Folcuti C, Horescu C, Barcan E, Alexandru O, Tuta C, Vatu BI, Artene SA, Dricu A. β-arrestin 1 transfection induced cell death in high grade glioma in vitro. J Immunoassay Immunochem 2020; 41:1021-1032. [PMID: 32807003 DOI: 10.1080/15321819.2020.1808990] [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: 01/11/2023]
Abstract
The best known functions of β-arrestins (β-arr) are to regulate G protein-coupled receptors (GPCR) signaling through receptor desensitization and internalization. Many reports also suggest that β-arrs play important role in immune regulation and inflammatory responses, under physiological and pathological conditions. Recent studies have shown that β-arr 1 silencing halts proliferation and increases temozolomide (TMZ) response in glioblastoma (GBM) cells. The focus of this paper is to analyze the role of β-arr 1 overexpression in the 18 high grade glioma (HGG) cell line in terms of viability and their response to TMZ treatment. For this reason, the cell line was transfected with β-arr 1 and the effect was analyzed after 24 h, 48 h and 72 h in terms of proliferation and treatment response. We observed that β-arr 1 overexpression induced a time and dose dependant inhibition in the HGG cells. Unexpectedly, β-arr transfection resulted in a very mild increase in TMZ toxicity after 24 h, becoming non-statistically significant at 72 h. In conclusion, we showed that β-arr 1 overexpression inhibits cell proliferation in the 18 cell line but only has a very modest effect on treatment response with the alkylating agent TMZ.
Collapse
Affiliation(s)
- Catalin Folcuti
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Cristina Horescu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Edmond Barcan
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Oana Alexandru
- Department of Neurology, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Cristian Tuta
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Bogdan-Ionel Vatu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Stefan-Alexandru Artene
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| | - Anica Dricu
- Department of Biochemistry, University of Medicine and Pharmacy of Craiova , Craiova, Romania
| |
Collapse
|
78
|
Thibeault PE, Ramachandran R. Role of the Helix-8 and C-Terminal Tail in Regulating Proteinase Activated Receptor 2 Signaling. ACS Pharmacol Transl Sci 2020; 3:868-882. [PMID: 33073187 DOI: 10.1021/acsptsci.0c00039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Indexed: 12/11/2022]
Abstract
The C-terminal tail of G-protein-coupled receptors (GPCR) contain important regulatory sites that enable interaction with intracellular signaling effectors. Here we examine the relative contribution of the C-tail serine/threonine phosphorylation sites (Ser383-385, Ser387-Thr392) and the helix-8 palmitoylation site (Cys361) in signaling regulation downstream of the proteolytically activated GPCR, PAR2. We examined Gαq/11-coupled calcium signaling, β-arrestin-1/-2 recruitment, and MAPK activation (p44/42 phosphorylation) by wild-type and mutant receptors expressed in a CRISPR/Cas9 PAR2-knockout HEK-293 cell background with both peptide stimulation of the receptor (SLIGRL-NH2) as well as activation with its endogenous trypsin revealed a tethered ligand. We find that alanine substitution of the membrane proximal serine residues (Ser383-385Ala) had no effect on SLIGRL-NH2- or trypsin-stimulated β-arrestin recruitment. In contrast, alanine substitutions in the Ser387-Thr392 cluster resulted in a large (∼50%) decrease in β-arrestin-1/-2 recruitment triggered by the activating peptide, SLIGRL-NH2, but was without an effect on trypsin-activated β-arrestin-1/-2 recruitment. Additionally, we find that alanine substitution of the helix-8 cysteine residue (Cys361Ala) led to a large decrease in both Gαq/11 coupling and β-arrestin-1/-2 recruitment to PAR2. Furthermore, we show that Gαq/11 inhibition with YM254890, inhibited ERK phosphorylation by PAR2 agonists, while genetic deletion of β-arrestin-1/-2 by CRISPR/Cas9 enhanced MAPK activation. Knockout of β-arrestins also enhanced Gαq/11-mediated calcium signaling. In line with these findings, a C-tail serine/threonine mutant that has decreased β-arrestin recruitment also showed enhanced ERK activation. Thus, our studies point to multiple mechanisms that regulate β-arrestin interaction with PAR2 and highlight differences in regulation of tethered-ligand- and peptide-mediated activation of this receptor.
Collapse
Affiliation(s)
- Pierre E Thibeault
- Department of Physiology and Pharmacology, University of Western Ontario, 1151 Richmond Street, London, Ontario N6A5C1, Canada
| | - Rithwik Ramachandran
- Department of Physiology and Pharmacology, University of Western Ontario, 1151 Richmond Street, London, Ontario N6A5C1, Canada
| |
Collapse
|
79
|
Han X, Nieman MT. The domino effect triggered by the tethered ligand of the protease activated receptors. Thromb Res 2020; 196:87-98. [PMID: 32853981 DOI: 10.1016/j.thromres.2020.08.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/23/2020] [Accepted: 08/03/2020] [Indexed: 12/20/2022]
Abstract
Protease activated receptors (PARs) are G-protein coupled receptors (GPCRs) that have a unique activation mechanism. Unlike other GPCRs that can be activated by free ligands, under physiological conditions, PARs are activated by the tethered ligand, which is a part of their N-terminus that is unmasked by proteolysis. It has been 30 years since the first member of the family, PAR1, was identified. In this review, we will discuss this unique tethered ligand mediate receptor activation of PARs in detail: how they interact with the proteases, the complex structural rearrangement of the receptors upon activation, and the termination of the signaling. We also summarize the structural studies of the PARs and how single nucleotide polymorphisms impact the receptor reactivity. Finally, we review the current strategies for inhibiting PAR function with therapeutic targets for anti-thrombosis. The focus of this review is PAR1 and PAR4 as they are the thrombin signal mediators on human platelets and therapeutics targets. We also include the structural studies of PAR2 as it informs the mechanism of action for PARs in general.
Collapse
Affiliation(s)
- Xu Han
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA
| | - Marvin T Nieman
- Department of Pharmacology, Case Western Reserve University, Cleveland, OH, USA.
| |
Collapse
|
80
|
Endosomal signaling of delta opioid receptors is an endogenous mechanism and therapeutic target for relief from inflammatory pain. Proc Natl Acad Sci U S A 2020; 117:15281-15292. [PMID: 32546520 DOI: 10.1073/pnas.2000500117] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Whether G protein-coupled receptors signal from endosomes to control important pathophysiological processes and are therapeutic targets is uncertain. We report that opioids from the inflamed colon activate δ-opioid receptors (DOPr) in endosomes of nociceptors. Biopsy samples of inflamed colonic mucosa from patients and mice with colitis released opioids that activated DOPr on nociceptors to cause a sustained decrease in excitability. DOPr agonists inhibited mechanically sensitive colonic nociceptors. DOPr endocytosis and endosomal signaling by protein kinase C (PKC) and extracellular signal-regulated kinase (ERK) pathways mediated the sustained inhibitory actions of endogenous opioids and DOPr agonists. DOPr agonists stimulated the recruitment of Gαi/o and β-arrestin1/2 to endosomes. Analysis of compartmentalized signaling revealed a requirement of DOPr endocytosis for activation of PKC at the plasma membrane and in the cytosol and ERK in the nucleus. We explored a nanoparticle delivery strategy to evaluate whether endosomal DOPr might be a therapeutic target for pain. The DOPr agonist DADLE was coupled to a liposome shell for targeting DOPr-positive nociceptors and incorporated into a mesoporous silica core for release in the acidic and reducing endosomal environment. Nanoparticles activated DOPr at the plasma membrane, were preferentially endocytosed by DOPr-expressing cells, and were delivered to DOPr-positive early endosomes. Nanoparticles caused a long-lasting activation of DOPr in endosomes, which provided sustained inhibition of nociceptor excitability and relief from inflammatory pain. Conversely, nanoparticles containing a DOPr antagonist abolished the sustained inhibitory effects of DADLE. Thus, DOPr in endosomes is an endogenous mechanism and a therapeutic target for relief from chronic inflammatory pain.
Collapse
|
81
|
Liu S, Luttrell LM, Premont RT, Rockey DC. β-Arrestin2 is a critical component of the GPCR-eNOS signalosome. Proc Natl Acad Sci U S A 2020; 117:11483-11492. [PMID: 32404425 PMCID: PMC7261012 DOI: 10.1073/pnas.1922608117] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 03/31/2020] [Indexed: 12/14/2022] Open
Abstract
Endothelial cell nitric oxide (NO) synthase (eNOS), the enzyme responsible for synthesis of NO in endothelial cells, is regulated by complex posttranslational mechanisms. Sinusoidal portal hypertension, a disorder characterized by liver sinusoidal endothelial cell (SEC) injury with resultant reduced eNOS activity and NO production within the liver, has been associated with defects in eNOS protein-protein interactions and posttranslational modifications. We and others have previously identified novel eNOS interactors, including G protein-coupled receptor (GPCR) kinase interactor 1 (GIT1), which we found to play an unexpected stimulatory role in GPCR-mediated eNOS signaling. Here we report that β-arrestin 2 (β-Arr2), a canonical GPCR signaling partner, localizes in SECs with eNOS in a GIT1/eNOS/NO signaling module. Most importantly, we show that β-Arr2 stimulates eNOS activity, and that β-Arr2 expression is reduced and formation of the GIT1/eNOS/NO signaling module is interrupted during liver injury. In β-Arr2-deficient mice, bile duct ligation injury (BDL) led to significantly reduced eNOS activity and to a dramatic increase in portal hypertension compared to BDL in wild-type mice. Overexpression of β-Arr2 in injured or β-Arr2-deficient SECs rescued eNOS function by increasing eNOS complex formation and NO production. We also found that β-Arr2-mediated GIT1/eNOS complex formation is dependent on Erk1/2 and Src, two kinases known to interact with and be activated by β-Arr2 in response to GCPR activation. Our data emphasize that β-Arr2 is an integral component of the GIT1/eNOS/NO signaling pathway and have implications for the pathogenesis of sinusoidal portal hypertension.
Collapse
Affiliation(s)
- Songling Liu
- Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, SC 29425
| | - Louis M Luttrell
- Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, SC 29425
| | - Richard T Premont
- Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH 44106
- Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH 44106
| | - Don C Rockey
- Division of Gastroenterology and Hepatology, Medical University of South Carolina, Charleston, SC 29425;
| |
Collapse
|
82
|
Ghali GZ, Ghali MGZ. β adrenergic receptor modulated signaling in glioma models: promoting β adrenergic receptor-β arrestin scaffold-mediated activation of extracellular-regulated kinase 1/2 may prove to be a panacea in the treatment of intracranial and spinal malignancy and extra-neuraxial carcinoma. Mol Biol Rep 2020; 47:4631-4650. [PMID: 32303958 PMCID: PMC7165076 DOI: 10.1007/s11033-020-05427-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 04/03/2020] [Indexed: 12/03/2022]
Abstract
Neoplastically transformed astrocytes express functionally active cell surface β adrenergic receptors (βARs). Treatment of glioma models in vitro and in vivo with β adrenergic agonists variably amplifies or attenuates cellular proliferation. In the majority of in vivo models, β adrenergic agonists generally reduce cellular proliferation. However, treatment with β adrenergic agonists consistently reduces tumor cell invasive potential, angiogenesis, and metastasis. β adrenergic agonists induced decreases of invasive potential are chiefly mediated through reductions in the expression of matrix metalloproteinases types 2 and 9. Treatment with β adrenergic agonists also clearly reduce tumoral neoangiogenesis, which may represent a putatively useful mechanism to adjuvantly amplify the effects of bevacizumab. Bevacizumab is a monoclonal antibody targeting the vascular endothelial growth factor receptor. We may accordingly designate βagonists to represent an enhancer of bevacizumab. The antiangiogenic effects of β adrenergic agonists may thus effectively render an otherwise borderline effective therapy to generate significant enhancement in clinical outcomes. β adrenergic agonists upregulate expression of the major histocompatibility class II DR alpha gene, effectively potentiating the immunogenicity of tumor cells to tumor surveillance mechanisms. Authors have also demonstrated crossmodal modulation of signaling events downstream from the β adrenergic cell surface receptor and microtubular polymerization and depolymerization. Complex effects and desensitization mechanisms of the β adrenergic signaling may putatively represent promising therapeutic targets. Constant stimulation of the β adrenergic receptor induces its phosphorylation by β adrenergic receptor kinase (βARK), rendering it a suitable substrate for alternate binding by β arrestins 1 or 2. The binding of a β arrestin to βARK phosphorylated βAR promotes receptor mediated internalization and downregulation of cell surface receptor and contemporaneously generates a cell surface scaffold at the βAR. The scaffold mediated activation of extracellular regulated kinase 1/2, compared with protein kinase A mediated activation, preferentially favors cytosolic retention of ERK1/2 and blunting of nuclear translocation and ensuant pro-transcriptional activity. Thus, βAR desensitization and consequent scaffold assembly effectively retains the cytosolic homeostatic functions of ERK1/2 while inhibiting its pro-proliferative effects. We suggest these mechanisms specifically will prove quite promising in developing primary and adjuvant therapies mitigating glioma growth, angiogenesis, invasive potential, and angiogenesis. We suggest generating compounds and targeted mutations of the β adrenergic receptor favoring β arrestin binding and scaffold facilitated activation of ERK1/2 may hold potential promise and therapeutic benefit in adjuvantly treating most or all cancers. We hope our discussion will generate fruitful research endeavors seeking to exploit these mechanisms.
Collapse
Affiliation(s)
- George Zaki Ghali
- United States Environmental Protection Agency, Arlington, VA, USA.,Emeritus Professor, Department of Toxicology, Purdue University, West Lafayette, IN, USA
| | - Michael George Zaki Ghali
- Department of Neurological Surgery, University of California, San Francisco, 505 Parnassus Avenue, Box-0112, San Francisco, CA, 94143, USA. .,Department of Neurological Surgery, Karolinska Institutet, Nobels väg 6, Solna and Alfred Nobels Allé 8, Huddinge, SE-171 77, Stockholm, Sweden.
| |
Collapse
|
83
|
Acharya S, Kundu D, Choi HJ, Kim KM. Metabotropic signaling cascade involved in α4β2 nicotinic acetylcholine receptor-mediated PKCβII activation. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118721. [PMID: 32304729 DOI: 10.1016/j.bbamcr.2020.118721] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 04/04/2020] [Accepted: 04/10/2020] [Indexed: 01/27/2023]
Abstract
Nicotinic acetylcholine receptors (nAChRs) belong to the ionophore receptor family, which regulates plasma membrane conductance to Na+, K+, and Ca2+ ions. Some studies, however, have shown that nAChRs also employ second messengers for intracellular signaling. We previously showed that α4β2 nAChR mediates the translocation of protein kinase CβII (PKCβII) from the cytoplasm to the plasma membrane, which is a typical activation marker for PKCβII. In this study, we investigated the molecular mechanisms underlying PKCβII activation through α4β2 nAChR. α4β2 nAChR is the most abundant nAChR subtype and is implicated in various brain functions and diseases. Putative α4β2 nAChR signaling components were identified by knockdown or chemical inhibition of candidate proteins, and the signaling cascade was deduced by protein interactions in predicted cellular components. α4β2 nAChR-mediated PKCβII translocation was found to occur in an ionophore activity-independent manner. Nicotinic stimulation of α4β2 nAChR activated Src in a β-arrestin1 and 14-3-3η-dependent manner. Activated Src phosphorylated the tyrosine residue(s) on Syk molecules, which in turn interacted with phospholipase C γ1 to trigger the translocation of PKCβII to the cell membrane by elevating cellular diacylglycerol levels. The activated PKCβII in turn exerted a positive feedback effect on Src activation, suggesting that α4β2 nAChR signaling is amplified by a positive feedback loop. These findings provide novel information for unveiling the previously unclear metabotropic second messenger-based signal transduction pathway of nAChRs.
Collapse
Affiliation(s)
- Srijan Acharya
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea
| | - Dooti Kundu
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea
| | - Hyun Jin Choi
- College of Pharmacy, Cha University, Seongnam, Gyeonggi-do 13488, Republic of Korea
| | - Kyeong-Man Kim
- Department of Pharmacology, College of Pharmacy, Chonnam National University, Gwang-Ju 61186, Republic of Korea.
| |
Collapse
|
84
|
Miningou N, Blackwell KT. The road to ERK activation: Do neurons take alternate routes? Cell Signal 2020; 68:109541. [PMID: 31945453 PMCID: PMC7127974 DOI: 10.1016/j.cellsig.2020.109541] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Revised: 01/11/2020] [Accepted: 01/12/2020] [Indexed: 01/29/2023]
Abstract
The ERK cascade is a central signaling pathway that regulates a wide variety of cellular processes including proliferation, differentiation, learning and memory, development, and synaptic plasticity. A wide range of inputs travel from the membrane through different signaling pathway routes to reach activation of one set of output kinases, ERK1&2. The classical ERK activation pathway beings with growth factor activation of receptor tyrosine kinases. Numerous G-protein coupled receptors and ionotropic receptors also lead to ERK through increases in the second messengers calcium and cAMP. Though both types of pathways are present in diverse cell types, a key difference is that most stimuli to neurons, e.g. synaptic inputs, are transient, on the order of milliseconds to seconds, whereas many stimuli acting on non-neural tissue, e.g. growth factors, are longer duration. The ability to consolidate these inputs to regulate the activation of ERK in response to diverse signals raises the question of which factors influence the difference in ERK activation pathways. This review presents both experimental studies and computational models aimed at understanding the control of ERK activation and whether there are fundamental differences between neurons and other cells. Our main conclusion is that differences between cell types are quite subtle, often related to differences in expression pattern and quantity of some molecules such as Raf isoforms. In addition, the spatial location of ERK is critical, with regulation by scaffolding proteins producing differences due to colocalization of upstream molecules that may differ between neurons and other cells.
Collapse
Affiliation(s)
- Nadiatou Miningou
- Department of Chemistry and Biochemistry, George Mason University, Fairfax, VA 22030, United States of America
| | - Kim T Blackwell
- Interdisciplinary Program in Neuroscience and Bioengineering Department, George Mason University, Fairfax, VA 22030, United States of America.
| |
Collapse
|
85
|
PLAG Exerts Anti-Metastatic Effects by Interfering with Neutrophil Elastase/PAR2/EGFR Signaling in A549 Lung Cancer Orthotopic Model. Cancers (Basel) 2020; 12:cancers12030560. [PMID: 32121107 PMCID: PMC7139301 DOI: 10.3390/cancers12030560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Revised: 02/26/2020] [Accepted: 02/26/2020] [Indexed: 12/20/2022] Open
Abstract
The effectiveness of chemotherapy and radiotherapy to treat lung cancer is limited because of highly metastatic nature. Novel strategies and drugs to attenuate metastatic activity are urgently required. In this study, red fluorescence proteins (RFP)-labeled A549 human lung cancer cells were orthotopically implantation, where they developed primary tumors. Metastasis in brain and intestines were reduced by up to 80% by treatment with 100 mpk 1-palmitoyl-2-linoleoyl-3-acetyl-rac-glycerol (PLAG) compared with that in control mice. PLAG treatment also reduced the migration of the primary tumors. Interestingly, substantial neutrophil infiltration was observed in the tumors in control mice. The neutrophil contribution to A549 cell metastatic activity was examined in in vitro co-culture system. Metastatic activity could be achieved in the A549 cells through epidermal growth factor receptor (EGFR) transactivation mediated by protease activating receptor 2 (PAR2) receptor. Neutrophil elastase secreted from tumor-infiltrating neutrophils stimulated PAR2 and induced EGFR transactivation. However, this transactivation was inhibited by inducing PAR2 degradation following PLAG treatment and metastatic activity was effectively inhibited. PLAG attenuated cancer metastatic activity via modulated PAR2/EGFR transactivation by accelerating PAR2 degradation. These results suggest PLAG as potential therapeutic agent to combat tumor metastasis via regulating the activation signal pathway of PAR2 by tumor infiltrate-neutrophils.
Collapse
|
86
|
Inhibition of excessive kallikrein-8 improves neuroplasticity in Alzheimer's disease mouse model. Exp Neurol 2020; 324:113115. [DOI: 10.1016/j.expneurol.2019.113115] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 10/02/2019] [Accepted: 11/12/2019] [Indexed: 01/24/2023]
|
87
|
Lobingier BT, von Zastrow M. When trafficking and signaling mix: How subcellular location shapes G protein-coupled receptor activation of heterotrimeric G proteins. Traffic 2019; 20:130-136. [PMID: 30578610 DOI: 10.1111/tra.12634] [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: 12/04/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 12/14/2022]
Abstract
G protein-coupled receptors (GPCRs) physically connect extracellular information with intracellular signal propagation. Membrane trafficking plays a supportive role by "bookending" signaling events: movement through the secretory pathway delivers GPCRs to the cell surface where receptors can sample the extracellular environment, while endocytosis and endolysosomal membrane trafficking provide a versatile system to titrate cellular signaling potential and maintain homeostatic control. Recent evidence suggests that, in addition to these important effects, GPCR trafficking actively shapes the cellular signaling response by altering the location and timing of specific receptor-mediated signaling reactions. Here, we review key experimental evidence underlying this expanding view, focused on GPCR signaling mediated through activation of heterotrimeric G proteins located in the cytoplasm. We then discuss lingering and emerging questions regarding the interface between GPCR signaling and trafficking.
Collapse
Affiliation(s)
- Braden T Lobingier
- Department of Psychiatry and Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California
| | - Mark von Zastrow
- Department of Psychiatry and Department of Cellular and Molecular Pharmacology, University of California, San Francisco School of Medicine, San Francisco, California
| |
Collapse
|
88
|
Li F, Yang W, Jiang H, Guo C, Huang AJW, Hu H, Liu Q. TRPV1 activity and substance P release are required for corneal cold nociception. Nat Commun 2019; 10:5678. [PMID: 31831729 PMCID: PMC6908618 DOI: 10.1038/s41467-019-13536-0] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 10/30/2019] [Indexed: 01/07/2023] Open
Abstract
As a protective mechanism, the cornea is sensitive to noxious stimuli. Here, we show that in mice, a high proportion of corneal TRPM8+ cold-sensing fibers express the heat-sensitive TRPV1 channel. Despite its insensitivity to cold, TRPV1 enhances membrane potential changes and electrical firing of TRPM8+ neurons in response to cold stimulation. This elevated neuronal excitability leads to augmented ocular cold nociception in mice. In a model of dry eye disease, the expression of TRPV1 in TRPM8+ cold-sensing fibers is increased, and results in severe cold allodynia. Overexpression of TRPV1 in TRPM8+ sensory neurons leads to cold allodynia in both corneal and non-corneal tissues without affecting their thermal sensitivity. TRPV1-dependent neuronal sensitization facilitates the release of the neuropeptide substance P from TRPM8+ cold-sensing neurons to signal nociception in response to cold. Our study identifies a mechanism underlying corneal cold nociception and suggests a potential target for the treatment of ocular pain. The eye shows protective responses to noxious stimuli including cold. Here, the authors show that TRPV1, found co-expressed on TRPM8 + fibres in the cornea, is necessary for cold nociception in the eye.
Collapse
Affiliation(s)
- Fengxian Li
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA.,Department of Anesthesiology, Zhujiang Hospital of Southern Medical University, Guangdong, China
| | - Weishan Yang
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Haowu Jiang
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Changxiong Guo
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Andrew J W Huang
- Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Hongzhen Hu
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Qin Liu
- Department of Anesthesiology, Center for the Study of Itch and Sensory Disorders, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA. .,Department of Ophthalmology & Visual Sciences, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
89
|
Retamal JS, Ramírez-García PD, Shenoy PA, Poole DP, Veldhuis NA. Internalized GPCRs as Potential Therapeutic Targets for the Management of Pain. Front Mol Neurosci 2019; 12:273. [PMID: 31798411 PMCID: PMC6874167 DOI: 10.3389/fnmol.2019.00273] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 10/28/2019] [Indexed: 01/14/2023] Open
Abstract
Peripheral and central neurons in the pain pathway are well equipped to detect and respond to extracellular stimuli such as pro-inflammatory mediators and neurotransmitters through the cell surface expression of receptors that can mediate rapid intracellular signaling. Following injury or infection, activation of cell surface G protein-coupled receptors (GPCRs) initiates cell signaling processes that lead to the generation of action potentials in neurons or inflammatory responses such as cytokine secretion by immune cells. However, it is now appreciated that cell surface events alone may not be sufficient for all receptors to generate their complete signaling repertoire. Following an initial wave of signaling at the cell surface, active GPCRs can engage with endocytic proteins such as the adaptor protein β-arrestin (βArr) to promote clathrin-mediated internalization. Classically, βArr-mediated internalization of GPCRs was hypothesized to terminate signaling, yet for multiple GPCRs known to contribute to pain, it has been demonstrated that endocytosis can also promote a unique "second wave" of signaling from intracellular membranes, including those of endosomes and the Golgi, that is spatiotemporally distinct from initial cell-surface events. In the context of pain, understanding the cellular and molecular mechanisms that drive spatiotemporal signaling of GPCRs is invaluable for understanding how pain occurs and persists, and how current analgesics achieve efficacy or promote side-effects. This review article discusses the importance of receptor localization for signaling outcomes of pro- and anti-nociceptive GPCRs, and new analgesic opportunities emerging through the development of "location-biased" ligands that favor binding with intracellular GPCR populations.
Collapse
Affiliation(s)
- Jeffri S Retamal
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| | - Paulina D Ramírez-García
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| | - Priyank A Shenoy
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| | - Daniel P Poole
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia.,Department of Anatomy and Neuroscience, The University of Melbourne, Parkville, VIC, Australia
| | - Nicholas A Veldhuis
- Drug Discovery Biology Theme, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, Australia.,Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, VIC, Australia
| |
Collapse
|
90
|
Fu D, Li P, Sheng Q, Lv Z. β-arrestin-2 enhances intestinal epithelial apoptosis in necrotizing enterocolitis. Aging (Albany NY) 2019; 11:8294-8312. [PMID: 31612867 PMCID: PMC6814604 DOI: 10.18632/aging.102320] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 09/21/2019] [Indexed: 12/20/2022]
Abstract
Apoptosis among intestinal epithelial cells contributes to necrotizing enterocolitis (NEC), a severe intestinal disease that particularly affects premature infants. β-arrestin-2, an important regulator of G-protein-coupled receptors, is expressed in intestinal epithelial cells, where its activation promotes apoptosis. We found that β-arrestin-2 was overexpressed in both human and murine NEC samples. β-arrestin-2-deficient mice were protected from endoplasmic reticulum stress and NEC development. The endoplasmic reticulum-resident chaperone BiP was found to promote intestinal epithelial cell survival. Pretreatment of intestinal epithelial cells or mice with the BiP inhibitor HA15 increased cell apoptosis and promoted NEC development. β-arrestin-2 bound to BiP and promoted its polyubiquitination and degradation, thereby facilitating the release of the pro-apoptotic molecule BIK from BiP. Silencing β-arrestin-2 downregulated apoptosis by increasing BiP levels, which suppressed endoplasmic reticulum stress. This study suggests that β-arrestin-2 induces NEC development by inhibiting BiP, thereby triggering apoptosis in response to endoplasmic reticulum stress. Thus, novel therapeutic strategies to inhibit β-arrestin-2 may enhance the treatment of NEC.
Collapse
Affiliation(s)
- Dong Fu
- Department of General Surgery, Children's Hospital of Shanghai, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China
| | - Peng Li
- Department of General Surgery, Children's Hospital of Shanghai, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China
| | - Qingfeng Sheng
- Department of General Surgery, Children's Hospital of Shanghai, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China
| | - Zhibao Lv
- Department of General Surgery, Children's Hospital of Shanghai, School of Medicine, Shanghai Jiao Tong University, Shanghai 200000, China
| |
Collapse
|
91
|
Martini ML, Ray C, Yu X, Liu J, Pogorelov VM, Wetsel WC, Huang XP, McCorvy JD, Caron MG, Jin J. Designing Functionally Selective Noncatechol Dopamine D 1 Receptor Agonists with Potent In Vivo Antiparkinsonian Activity. ACS Chem Neurosci 2019; 10:4160-4182. [PMID: 31387346 DOI: 10.1021/acschemneuro.9b00410] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Dopamine receptors are important G protein-coupled receptors (GPCRs) with therapeutic opportunities for treating Parkinson's Disease (PD) motor and cognitive deficits. Biased D1 dopamine ligands that differentially activate G protein over β-arrestin recruitment pathways are valuable chemical tools for dissecting positive versus negative effects in drugs for PD. Here, we reveal an iterative approach toward modification of a D1-selective noncatechol scaffold critical for G protein-biased agonism. This approach provided enhanced understanding of the structural components critical for activity and signaling bias and led to the discovery of several novel compounds with useful pharmacological properties, including three highly GS-biased partial agonists. Administration of a potent, balanced, and brain-penetrant lead compound from this series results in robust antiparkinsonian effects in a rodent model of PD. This study suggests that the noncatechol ligands developed through this approach are valuable tools for probing D1 receptor signaling biology and biased agonism in models of neurologic disease.
Collapse
Affiliation(s)
- Michael L. Martini
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
- Medical Scientist Training Program, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Caroline Ray
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Xufen Yu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Jing Liu
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| | - Vladimir M. Pogorelov
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
- Departments of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, United States
- Department of Medicine and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - William C. Wetsel
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
- Departments of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, North Carolina 27710, United States
- Department of Medicine and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Xi-Ping Huang
- Department of Pharmacology and National Institute of Mental Health Psychoactive Drug Screening Program, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States
| | - John D. McCorvy
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, Wisconsin 53226, United States
| | - Marc G. Caron
- Department of Cell Biology, Duke University Medical Center, Durham, North Carolina 27710, United States
- Department of Medicine and Neurobiology, Duke University Medical Center, Durham, North Carolina 27710, United States
| | - Jian Jin
- Mount Sinai Center for Therapeutics Discovery, Departments of Pharmacological Sciences and Oncological Sciences, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York 10029, United States
| |
Collapse
|
92
|
Villegas-Comonfort S, Guzmán-Silva A, Romero-Ávila MT, Takei Y, Tsujimoto G, Hirasawa A, García-Sáinz JA. Receptor tyrosine kinase activation induces free fatty acid 4 receptor phosphorylation, β-arrestin interaction, and internalization. Eur J Pharmacol 2019; 855:267-275. [PMID: 31078517 DOI: 10.1016/j.ejphar.2019.05.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 04/12/2019] [Accepted: 05/06/2019] [Indexed: 12/30/2022]
Abstract
FFA4 (Free Fatty Acid receptor 4, previously known as GPR120) is a G protein-coupled receptor that acts as a sensor of long-chain fatty acids, modulates metabolism, and whose dysfunction participates in endocrine disturbances. FFA4 is known to be phosphorylated and internalized in response to agonists and protein kinase C activation. In this paper report the modulation of this fatty acid receptor by activation of receptor tyrosine kinases. Cell-activation with growth factors (insulin, epidermal growth factor, insulin-like growth factor-I, and platelet-derived growth factor) increases FFA4 phosphorylation in a time- and concentration-dependent fashion. This effect was blocked by inhibitors of protein kinase C and phosphoinositide 3-kinase, suggesting the involvement of these kinases in it. FFA4 phosphorylation did not alter agonist-induced FFA4 calcium signaling, but was associated with decreased ERK 1/2 phosphorylation. In addition, insulin, insulin-like growth factor-I, epidermal growth factor, and to a lesser extent, platelet-derived growth factor, induce receptor internalization. This action of insulin, insulin-like growth factor I, and epidermal growth factor was blocked by inhibitors of protein kinase C and phosphoinositide 3-kinase. Additionally, cell treatment with these growth factors induced FFA4-β-arrestin coimmunoprecipitation. Our results evidenced cross-talk between receptor tyrosine kinases and FFA4 and suggest roles of protein kinase C and phosphoinositide 3-kinase in such a functional interaction.
Collapse
Affiliation(s)
- Sócrates Villegas-Comonfort
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México, 04510, Mexico
| | - Alejandro Guzmán-Silva
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México, 04510, Mexico
| | - M Teresa Romero-Ávila
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México, 04510, Mexico
| | - Yoshinori Takei
- Graduate School of Pharmaceutical Sciences, Kyoto University: Sakyo-ku, Kyoto, 606-8501, Japan
| | - Gozoh Tsujimoto
- Graduate School of Pharmaceutical Sciences, Kyoto University: Sakyo-ku, Kyoto, 606-8501, Japan
| | - Akira Hirasawa
- Graduate School of Pharmaceutical Sciences, Kyoto University: Sakyo-ku, Kyoto, 606-8501, Japan
| | - J Adolfo García-Sáinz
- Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Ap. Postal 70-248, Ciudad de México, 04510, Mexico.
| |
Collapse
|
93
|
Philip JL, Xu X, Han M, Akhter SA, Razzaque MA. Regulation of cardiac fibroblast-mediated maladaptive ventricular remodeling by β-arrestins. PLoS One 2019; 14:e0219011. [PMID: 31269046 PMCID: PMC6609028 DOI: 10.1371/journal.pone.0219011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 06/13/2019] [Indexed: 01/17/2023] Open
Abstract
Cardiac fibroblasts (CF) play a critical role in post-infarction remodeling which can ultimately lead to pathological fibrosis and heart failure. Recent evidence demonstrates that remote (non-infarct) territory fibrosis is a major mechanism for ventricular dysfunction and arrhythmogenesis. β-arrestins are important signaling molecules involved in β-adrenergic receptor (β-AR) desensitization and can also mediate signaling in a G protein independent fashion. Recent work has provided evidence that β-arrestin signaling in the heart may be beneficial, however, these studies have primarily focused on cardiac myocytes and their role in adult CF biology has not been well studied. In this study, we show that β-arrestins can regulate CF biology and contribute to pathological fibrosis. Adult male rats underwent LAD ligation to induce infarction and were studied by echocardiography. There was a significant decline in LV function at 2–12 weeks post-MI with increased infarct and remote territory fibrosis by histology consistent with maladaptive remodeling. Collagen synthesis was upregulated 2.9-fold in CF isolated at 8 and 12 weeks post-MI and β-arrestin expression was significantly increased. β-adrenergic signaling was uncoupled in the post-MI CF and β-agonist-mediated inhibition of collagen synthesis was lost. Knockdown of β-arrestin1 or 2 in the post-MI CF inhibited transformation to myofibroblasts as well as basal and TGF-β-stimulated collagen synthesis. These data suggest that β-arrestins can regulate CF biology and that targeted inhibition of these signaling molecules may represent a novel approach to prevent post-infarction pathological fibrosis and the transition to HF.
Collapse
Affiliation(s)
- Jennifer L. Philip
- Department of Surgery, Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Department of Surgery, Section of Cardiac and Thoracic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Xianyao Xu
- Department of Surgery, Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Department of Surgery, Section of Cardiac and Thoracic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Mei Han
- Department of Surgery, Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Department of Surgery, Section of Cardiac and Thoracic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
| | - Shahab A. Akhter
- Department of Surgery, Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Department of Surgery, Section of Cardiac and Thoracic Surgery, The University of Chicago Medical Center, Chicago, Illinois, United States of America
- Department of Cardiovascular Sciences, East Carolina Heart Institute at East Carolina University, Greenville, North Carolina, United States of America
| | - Md Abdur Razzaque
- Department of Surgery, Division of Cardiothoracic Surgery, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
- Department of Surgery and Cardiovascular Center of Excellence, Louisiana State University, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
94
|
Zaballos MA, Acuña-Ruiz A, Morante M, Crespo P, Santisteban P. Regulators of the RAS-ERK pathway as therapeutic targets in thyroid cancer. Endocr Relat Cancer 2019; 26:R319-R344. [PMID: 30978703 DOI: 10.1530/erc-19-0098] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/30/2022]
Abstract
Thyroid cancer is mostly an ERK-driven carcinoma, as up to 70% of thyroid carcinomas are caused by mutations that activate the RAS/ERK mitogenic signaling pathway. The incidence of thyroid cancer has been steadily increasing for the last four decades; yet, there is still no effective treatment for advanced thyroid carcinomas. Current research efforts are focused on impairing ERK signaling with small-molecule inhibitors, mainly at the level of BRAF and MEK. However, despite initial promising results in animal models, the clinical success of these inhibitors has been limited by the emergence of tumor resistance and relapse. The RAS/ERK pathway is an extremely complex signaling cascade with multiple points of control, offering many potential therapeutic targets: from the modulatory proteins regulating the activation state of RAS proteins to the scaffolding proteins of the pathway that provide spatial specificity to the signals, and finally, the negative feedbacks and phosphatases responsible for inactivating the pathway. The aim of this review is to give an overview of the biology of RAS/ERK regulators in human cancer highlighting relevant information on thyroid cancer and future areas of research.
Collapse
Affiliation(s)
- Miguel A Zaballos
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Adrián Acuña-Ruiz
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Morante
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Cantabria, Santander, Spain
| | - Piero Crespo
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), Consejo Superior de Investigaciones Científicas (CSIC), Universidad de Cantabria, Santander, Spain
| | - Pilar Santisteban
- Instituto de Investigaciones Biomédicas 'Alberto Sols', Consejo Superior de Investigaciones Científicas (CSIC), Universidad Autónoma de Madrid (UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
95
|
Zhao P, Pattison LA, Jensen DD, Jimenez-Vargas NN, Latorre R, Lieu T, Jaramillo JO, Lopez-Lopez C, Poole DP, Vanner SJ, Schmidt BL, Bunnett NW. Protein kinase D and Gβγ mediate sustained nociceptive signaling by biased agonists of protease-activated receptor-2. J Biol Chem 2019; 294:10649-10662. [PMID: 31142616 DOI: 10.1074/jbc.ra118.006935] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 05/23/2019] [Indexed: 12/20/2022] Open
Abstract
Proteases sustain hyperexcitability and pain by cleaving protease-activated receptor-2 (PAR2) on nociceptors through distinct mechanisms. Whereas trypsin induces PAR2 coupling to Gαq, Gαs, and β-arrestins, cathepsin-S (CS) and neutrophil elastase (NE) cleave PAR2 at distinct sites and activate it by biased mechanisms that induce coupling to Gαs, but not to Gαq or β-arrestins. Because proteases activate PAR2 by irreversible cleavage, and activated PAR2 is degraded in lysosomes, sustained extracellular protease-mediated signaling requires mobilization of intact PAR2 from the Golgi apparatus or de novo synthesis of new receptors by incompletely understood mechanisms. We found here that trypsin, CS, and NE stimulate PAR2-dependent activation of protein kinase D (PKD) in the Golgi of HEK293 cells, in which PKD regulates protein trafficking. The proteases stimulated translocation of the PKD activator Gβγ to the Golgi, coinciding with PAR2 mobilization from the Golgi. Proteases also induced translocation of a photoconverted PAR2-Kaede fusion protein from the Golgi to the plasma membrane of KNRK cells. After incubation of HEK293 cells and dorsal root ganglia neurons with CS, NE, or trypsin, PAR2 responsiveness initially declined, consistent with PAR2 cleavage and desensitization, and then gradually recovered. Inhibitors of PKD, Gβγ, and protein translation inhibited recovery of PAR2 responsiveness. PKD and Gβγ inhibitors also attenuated protease-evoked mechanical allodynia in mice. We conclude that proteases that activate PAR2 by canonical and biased mechanisms stimulate PKD in the Golgi; PAR2 mobilization and de novo synthesis repopulate the cell surface with intact receptors and sustain nociceptive signaling by extracellular proteases.
Collapse
Affiliation(s)
- Peishen Zhao
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Luke A Pattison
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Dane D Jensen
- the Departments of Surgery and Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - Nestor N Jimenez-Vargas
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Rocco Latorre
- the Departments of Surgery and Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032
| | - TinaMarie Lieu
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Josue O Jaramillo
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Cintya Lopez-Lopez
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Daniel P Poole
- From the Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria 3052, Australia
| | - Stephen J Vanner
- the Gastrointestinal Diseases Research Unit, Division of Gastroenterology, Queen's University, Kingston, Ontario K7L 3N6, Canada, and
| | - Brian L Schmidt
- the Bluestone Center for Clinical Research, New York University College of Dentistry, New York, New York 10010
| | - Nigel W Bunnett
- the Departments of Surgery and Pharmacology, Columbia University Vagelos College of Physicians and Surgeons, Columbia University, New York, New York 10032,
| |
Collapse
|
96
|
Li Y, Yang T, Yao Q, Li S, Fang E, Li Y, Liu C, Li W. Metformin prevents colonic barrier dysfunction by inhibiting mast cell activation in maternal separation-induced IBS-like rats. Neurogastroenterol Motil 2019; 31:e13556. [PMID: 30740845 DOI: 10.1111/nmo.13556] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 11/23/2018] [Accepted: 12/27/2018] [Indexed: 02/08/2023]
Abstract
BACKGROUND Intestinal barrier dysfunction is a key etiologic factor of irritable bowel syndrome (IBS). Metformin improves intestinal barrier function, although the underlying mechanism has yet to be fully explained. This study evaluates the protective effect of metformin on colonic barrier integrity and explores the underlying cellular mechanisms. METHODS IBS-like rats were induced by maternal separation. Metformin was administered daily by gavage at 08:30, and rat pups were then separated from their mother. Visceral hyperalgesia and depression-like behaviors were evaluated by colorectal distension, sucrose preference tests, and forced swimming tests. Intestinal integrity was analyzed using sugar probes and transmission electron microscopy. Inflammatory factors and the levels of corticotropin-releasing factor were assessed by PCR and ELISA. The number of mast cells was evaluated by toluidine blue staining. Protein expression and localization were determined using Western blot and immunochemistry. KEY RESULTS Metformin pretreatment (a) reduced visceral hypersensitivity to colorectal distension, immobility time and enhanced sucrose consumption; (b) decreased urine lactulose/mannitol ratio and sucralose output; (c) inhibited the dilation of tight junction and prevented claudin-4 translocation; (d) inhibited mast cell activation and downregulated the expression of IL-6, IL-18, tryptase, PAR-2, and ERK activation; (e) inhibited claudin-4 phosphorylation at serine sites and interactions between clau-4 and ZO-1. CONCLUSIONS & INFERENCES Metformin may block mast cell activation to reduce PAR-2 expression and subsequently inhibit ERK activation and clau-4 phosphorylation at serine sites to normalize the interaction of clau-4 and ZO-1 and clau-4 distribution. Metformin may be clinically beneficial for patients with IBS or IBS-like symptoms.
Collapse
Affiliation(s)
- Yong Li
- Laboratory of Neuronal Network and Systems Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.,Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China
| | - Tingting Yang
- Laboratory of Neuronal Network and Systems Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qing Yao
- Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China
| | - Songsong Li
- Xianning Institute for Drug Control, Xianning, China
| | - En Fang
- Xianning Institute for Drug Control, Xianning, China
| | - Yankun Li
- College of Pharmacy, Hubei University of Science and Technology, Xianning, China
| | - Chao Liu
- Hubei Key Laboratory of Cardiovascular, Cerebrovascular, and Metabolic Disorders, Hubei University of Science and Technology, Xianning, China
| | - Weimin Li
- Laboratory of Neuronal Network and Systems Biology, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
97
|
Canals M, Poole DP, Veldhuis NA, Schmidt BL, Bunnett NW. G-Protein-Coupled Receptors Are Dynamic Regulators of Digestion and Targets for Digestive Diseases. Gastroenterology 2019; 156:1600-1616. [PMID: 30771352 PMCID: PMC6508858 DOI: 10.1053/j.gastro.2019.01.266] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Revised: 12/15/2018] [Accepted: 01/08/2019] [Indexed: 01/11/2023]
Abstract
G-protein-coupled receptors (GPCRs) are the largest family of transmembrane signaling proteins. In the gastrointestinal tract, GPCRs expressed by epithelial cells sense contents of the lumen, and GPCRs expressed by epithelial cells, myocytes, neurons, and immune cells participate in communication among cells. GPCRs control digestion, mediate digestive diseases, and coordinate repair and growth. GPCRs are the target of more than one third of therapeutic drugs, including many drugs used to treat digestive diseases. Recent advances in structural, chemical, and cell biology research have shown that GPCRs are not static binary switches that operate from the plasma membrane to control a defined set of intracellular signals. Rather, GPCRs are dynamic signaling proteins that adopt distinct conformations and subcellular distributions when associated with different ligands and intracellular effectors. An understanding of the dynamic nature of GPCRs has provided insights into the mechanism of activation and signaling of GPCRs and has shown opportunities for drug discovery. We review the allosteric modulation, biased agonism, oligomerization, and compartmentalized signaling of GPCRs that control digestion and digestive diseases. We highlight the implications of these concepts for the development of selective and effective drugs to treat diseases of the gastrointestinal tract.
Collapse
Affiliation(s)
- Meritxell Canals
- Centre for Membrane Proteins and Receptors (COMPARE), School of Life Sciences, University of Nottingham, Nottingham, United Kingdom
| | - Daniel P. Poole
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, Victoria, Australia
| | - Nicholas A. Veldhuis
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia
| | - Brian L. Schmidt
- Bluestone Center for Clinical Research, New York University College of Dentistry, New York, New York
| | - Nigel W. Bunnett
- Monash Institute of Pharmaceutical Sciences and Australian Research Council Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Parkville, Victoria, Australia,Department of Pharmacology and Therapeutics, University of Melbourne, Parkville, Victoria, Australia,Columbia University College of Physicians and Surgeons, Columbia University, New York, New York
| |
Collapse
|
98
|
Heuberger DM, Schuepbach RA. Protease-activated receptors (PARs): mechanisms of action and potential therapeutic modulators in PAR-driven inflammatory diseases. Thromb J 2019; 17:4. [PMID: 30976204 PMCID: PMC6440139 DOI: 10.1186/s12959-019-0194-8] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 03/08/2019] [Indexed: 12/29/2022] Open
Abstract
Inflammatory diseases have become increasingly prevalent with industrialization. To address this, numerous anti-inflammatory agents and molecular targets have been considered in clinical trials. Among molecular targets, protease-activated receptors (PARs) are abundantly recognized for their roles in the development of chronic inflammatory diseases. In particular, several inflammatory effects are directly mediated by the sensing of proteolytic activity by PARs. PARs belong to the seven transmembrane domain G protein-coupled receptor family, but are unique in their lack of physiologically soluble ligands. In contrast with classical receptors, PARs are activated by N-terminal proteolytic cleavage. Upon removal of specific N-terminal peptides, the resulting N-termini serve as tethered activation ligands that interact with the extracellular loop 2 domain and initiate receptor signaling. In the classical pathway, activated receptors mediate signaling by recruiting G proteins. However, activation of PARs alternatively lead to the transactivation of and signaling through receptors such as co-localized PARs, ion channels, and toll-like receptors. In this review we consider PARs and their modulators as potential therapeutic agents, and summarize the current understanding of PAR functions from clinical and in vitro studies of PAR-related inflammation.
Collapse
Affiliation(s)
- Dorothea M Heuberger
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland.,Surgical Research Division, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Reto A Schuepbach
- Institute of Intensive Care Medicine, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
99
|
Martini ML, Liu J, Ray C, Yu X, Huang XP, Urs A, Urs N, McCorvy JD, Caron MG, Roth BL, Jin J. Defining Structure-Functional Selectivity Relationships (SFSR) for a Class of Non-Catechol Dopamine D 1 Receptor Agonists. J Med Chem 2019; 62:3753-3772. [PMID: 30875219 DOI: 10.1021/acs.jmedchem.9b00351] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
G protein-coupled receptors (GPCRs) are capable of downstream signaling through distinct noncanonical pathways such as β-arrestins in addition to the canonical G protein-dependent pathways. GPCR ligands that differentially activate the downstream signaling pathways are termed functionally selective or biased ligands. A class of novel non-catechol G protein-biased agonists of the dopamine D1 receptor (D1R) was recently disclosed. We conducted the first comprehensive structure-functional selectivity relationship study measuring GS and β-arrestin2 recruitment activities focused on four regions of this scaffold, resulting in over 50 analogs with diverse functional selectivity profiles. Some compounds became potent full agonists of β-arrestin2 recruitment, while others displayed enhanced GS bias compared to the starting compound. Pharmacokinetic testing of an analog with an altered functional selectivity profile demonstrated excellent blood-brain barrier penetration. This study provides novel tools for studying ligand bias at D1R and paves the way for developing the next generation of biased D1R ligands.
Collapse
Affiliation(s)
| | | | | | | | - Xi-Ping Huang
- Department of Pharmacology and National Institute of Mental Health Psychoactive Drug Screening Program, School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | - Aarti Urs
- Department of Pharmacology and Therapeutics, College of Medicine , University of Florida , Gainesville , Florida 32610 , United States
| | - Nikhil Urs
- Department of Pharmacology and Therapeutics, College of Medicine , University of Florida , Gainesville , Florida 32610 , United States
| | - John D McCorvy
- Department of Pharmacology and National Institute of Mental Health Psychoactive Drug Screening Program, School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States.,Department of Cell Biology, Neurobiology and Anatomy , Medical College of Wisconsin , Milwaukee , Wisconsin 53226 , United States
| | | | - Bryan L Roth
- Department of Pharmacology and National Institute of Mental Health Psychoactive Drug Screening Program, School of Medicine , University of North Carolina at Chapel Hill , Chapel Hill , North Carolina 27599 , United States
| | | |
Collapse
|
100
|
|