1
|
Yin L, Ni K, Mao T, Tian S, Liu C, Chen J, Zhou M, Li H, Hu Q. Attributes novel drug candidate: Constitutive GPCR signal bias mediated by purinergic receptors. Pharmacol Ther 2025; 267:108802. [PMID: 39862926 DOI: 10.1016/j.pharmthera.2025.108802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 12/05/2024] [Accepted: 01/15/2025] [Indexed: 01/27/2025]
Abstract
G protein-coupled receptors (GPCRs) can transmit signals via G protein-dependent or independent pathways due to the conformational changes of receptors and ligands, which is called biased signaling. This concept posits that ligands can selectively activate a specific signaling pathway after receptor activation, facilitating downstream signaling along a preferred pathway. Biased agonism enables the development of ligands that prioritize therapeutic signaling pathways while mitigating on-target undesired effects. As a class of GPCRs located on the surface of cell membranes, the discovery and clinical implementation of adenosine and P2Y receptors purinergic signaling modulators have progressed dramatically. However, many preclinical drug candidates targeting purinergic receptors have failed in clinical trials due to limited efficacy and/or severe on-target undesired effects. To overcome the key barriers typically encountered when transitioning ligands into the clinic, the renewed impetus has focused on the modulation of purinergic receptor function by exogenous agonists/antagonists and allosteric modulators to exploit biased agonism. This article provides a brief overview of the research progress on the mechanism of purinergic biased signal transduction from the conformational changes of purinergic GPCRs and biased ligands primarily, and highlights therapeutically relevant biased agonism at purinergic receptors.
Collapse
Affiliation(s)
- Li Yin
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Kexin Ni
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Tianqi Mao
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Sheng Tian
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China.
| | - Chunxiao Liu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| | - Jiayao Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Mengze Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Huanqiu Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Qinghua Hu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China; School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
2
|
Shafaghat Z, Ghomi AHK, Khorramdelazad H, Safari E. Purinergic signaling: decoding its role in COVID-19 pathogenesis and promising treatment strategies. Inflammopharmacology 2023; 31:3005-3020. [PMID: 37805959 DOI: 10.1007/s10787-023-01344-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023]
Abstract
The pathogenesis of coronavirus disease (COVID-19), caused by severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2), is complex and involves dysregulated immune responses, inflammation, and coagulopathy. Purinergic signaling, mediated by extracellular nucleotides and nucleosides, has emerged as a significant player in the pathogenesis of COVID-19. Extracellular adenosine triphosphate (ATP), released from damaged or infected cells, is a danger signal triggering immune responses. It activates immune cells, releasing pro-inflammatory cytokines, contributing to the cytokine storm observed in severe COVID-19 cases. ATP also promotes platelet activation and thrombus formation, contributing to the hypercoagulability seen in COVID-19 patients. On the other hand, adenosine, an immunosuppressive nucleoside, can impair anti-viral immune responses and promote tissue damage through its anti-inflammatory effects. Modulating purinergic receptors represents a promising therapeutic strategy for COVID-19. Understanding the role of purinergic signaling in COVID-19 pathogenesis and developing targeted therapeutic approaches can potentially improve patient outcomes. This review focuses on the part of purinergic signaling in COVID-19 pathogenesis and highlights potential therapeutic approaches targeting purinergic receptors.
Collapse
Affiliation(s)
- Zahra Shafaghat
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| | - Elaheh Safari
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
P2Y purinergic signaling in prostate cancer: Emerging insights into pathophysiology and therapy. Biochim Biophys Acta Rev Cancer 2022; 1877:188732. [DOI: 10.1016/j.bbcan.2022.188732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 04/18/2022] [Accepted: 04/21/2022] [Indexed: 12/15/2022]
|
4
|
Treppiedi D, Marra G, Di Muro G, Catalano R, Mangili F, Esposito E, Calebiro D, Arosio M, Peverelli E, Mantovani G. Dimerization of GPCRs: Novel insight into the role of FLNA and SSAs regulating SST 2 and SST 5 homo- and hetero-dimer formation. Front Endocrinol (Lausanne) 2022; 13:892668. [PMID: 35992099 PMCID: PMC9389162 DOI: 10.3389/fendo.2022.892668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
The process of GPCR dimerization can have profound effects on GPCR activation, signaling, and intracellular trafficking. Somatostatin receptors (SSTs) are class A GPCRs abundantly expressed in pituitary tumors where they represent the main pharmacological targets of somatostatin analogs (SSAs), thanks to their antisecretory and antiproliferative actions. The cytoskeletal protein filamin A (FLNA) directly interacts with both somatostatin receptor type 2 (SST2) and 5 (SST5) and regulates their expression and signaling in pituitary tumoral cells. So far, the existence and physiological relevance of SSTs homo- and hetero-dimerization in the pituitary have not been explored. Moreover, whether octreotide or pasireotide may play modulatory effects and whether FLNA may participate to this level of receptor organization have remained elusive. Here, we used a proximity ligation assay (PLA)-based approach for the in situ visualization and quantification of SST2/SST5 dimerization in rat GH3 as well as in human melanoma cells either expressing (A7) or lacking (M2) FLNA. First, we observed the formation of endogenous SST5 homo-dimers in GH3, A7, and M2 cells. Using the PLA approach combined with epitope tagging, we detected homo-dimers of human SST2 in GH3, A7, and M2 cells transiently co-expressing HA- and SNAP-tagged SST2. SST2 and SST5 can also form endogenous hetero-dimers in these cells. Interestingly, FLNA absence reduced the basal number of hetero-dimers (-36.8 ± 6.3% reduction of PLA events in M2, P < 0.05 vs. A7), and octreotide but not pasireotide promoted hetero-dimerization in both A7 and M2 (+20.0 ± 11.8% and +44.1 ± 16.3% increase of PLA events in A7 and M2, respectively, P < 0.05 vs. basal). Finally, immunofluorescence data showed that SST2 and SST5 recruitment at the plasma membrane and internalization are similarly induced by octreotide and pasireotide in GH3 and A7 cells. On the contrary, in M2 cells, octreotide failed to internalize both receptors whereas pasireotide promoted robust receptor internalization at shorter times than in A7 cells. In conclusion, we demonstrated that in GH3 cells SST2 and SST5 can form both homo- and hetero-dimers and that FLNA plays a role in the formation of SST2/SST5 hetero-dimers. Moreover, we showed that FLNA regulates SST2 and SST5 intracellular trafficking induced by octreotide and pasireotide.
Collapse
Affiliation(s)
- Donatella Treppiedi
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Giusy Marra
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Genesio Di Muro
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- University Sapienza of Rome, Rome, Italy
| | - Rosa Catalano
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Federica Mangili
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Emanuela Esposito
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Davide Calebiro
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, United Kingdom
- Centre of Membrane Proteins and Receptors, Universities of Birmingham and Nottingham, Birmingham, United Kingdom
| | - Maura Arosio
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Endocrinology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Erika Peverelli
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- *Correspondence: Erika Peverelli,
| | - Giovanna Mantovani
- Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
- Endocrinology Unit, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
5
|
Kosmowska B, Wardas J. The Pathophysiology and Treatment of Essential Tremor: The Role of Adenosine and Dopamine Receptors in Animal Models. Biomolecules 2021; 11:1813. [PMID: 34944457 PMCID: PMC8698799 DOI: 10.3390/biom11121813] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/25/2021] [Accepted: 11/30/2021] [Indexed: 12/13/2022] Open
Abstract
Essential tremor (ET) is one of the most common neurological disorders that often affects people in the prime of their lives, leading to a significant reduction in their quality of life, gradually making them unable to independently perform the simplest activities. Here we show that current ET pharmacotherapy often does not sufficiently alleviate disease symptoms and is completely ineffective in more than 30% of patients. At present, deep brain stimulation of the motor thalamus is the most effective ET treatment. However, like any brain surgery, it can cause many undesirable side effects; thus, it is only performed in patients with an advanced disease who are not responsive to drugs. Therefore, it seems extremely important to look for new strategies for treating ET. The purpose of this review is to summarize the current knowledge on the pathomechanism of ET based on studies in animal models of the disease, as well as to present and discuss the results of research available to date on various substances affecting dopamine (mainly D3) or adenosine A1 receptors, which, due to their ability to modulate harmaline-induced tremor, may provide the basis for the development of new potential therapies for ET in the future.
Collapse
Affiliation(s)
| | - Jadwiga Wardas
- Department of Neuropsychopharmacology, Maj Institute of Pharmacology Polish Academy of Sciences, 31-343 Kraków, Poland;
| |
Collapse
|
6
|
Guo X, Li Q, Pi S, Xia Y, Mao L. G protein-coupled purinergic P2Y receptor oligomerization: Pharmacological changes and dynamic regulation. Biochem Pharmacol 2021; 192:114689. [PMID: 34274353 DOI: 10.1016/j.bcp.2021.114689] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/12/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022]
Abstract
P2Y receptors (P2YRs) are a δ group of rhodopsin-like G protein-coupled receptors (GPCRs) with many essential functions in physiology and pathology, such as platelet aggregation, immune responses, neuroprotective effects, inflammation, and cellular proliferation. Thus, they are among the most researched therapeutic targets used for the clinical treatment of diseases (e.g., the antithrombotic drug clopidogrel and the dry eye treatment drug diquafosol). GPCRs transmit signals as dimers to increase the diversity of signalling pathways and pharmacological activities. Many studies have frequently confirmed dimerization between P2YRs and other GPCRs due to their functions in cardiovascular and cerebrovascular processes in vivo and in vitro. Recently, some P2YR dimers that dynamically balance physiological functions in the body were shown to be involved in effective signal transduction and exert pathological responses. In this review, we summarize the types, pharmacological changes, and active regulators of P2YR-related dimerization, and delineate new functions and pharmacological activities of P2YR-related dimers, which may be a novel direction to improve the effectiveness of medications.
Collapse
Affiliation(s)
- Xiaoqing Guo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qin Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shulan Pi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yuanpeng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| | - Ling Mao
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
7
|
Nguyen KDQ, Vigers M, Sefah E, Seppälä S, Hoover JP, Schonenbach NS, Mertz B, O'Malley MA, Han S. Homo-oligomerization of the human adenosine A 2A receptor is driven by the intrinsically disordered C-terminus. eLife 2021; 10:e66662. [PMID: 34269678 PMCID: PMC8328514 DOI: 10.7554/elife.66662] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 07/15/2021] [Indexed: 11/27/2022] Open
Abstract
G protein-coupled receptors (GPCRs) have long been shown to exist as oligomers with functional properties distinct from those of the monomeric counterparts, but the driving factors of oligomerization remain relatively unexplored. Herein, we focus on the human adenosine A2A receptor (A2AR), a model GPCR that forms oligomers both in vitro and in vivo. Combining experimental and computational approaches, we discover that the intrinsically disordered C-terminus of A2AR drives receptor homo-oligomerization. The formation of A2AR oligomers declines progressively with the shortening of the C-terminus. Multiple interaction types are responsible for A2AR oligomerization, including disulfide linkages, hydrogen bonds, electrostatic interactions, and hydrophobic interactions. These interactions are enhanced by depletion interactions, giving rise to a tunable network of bonds that allow A2AR oligomers to adopt multiple interfaces. This study uncovers the disordered C-terminus as a prominent driving factor for the oligomerization of a GPCR, offering important insight into the effect of C-terminus modification on receptor oligomerization of A2AR and other GPCRs reconstituted in vitro for biophysical studies.
Collapse
Affiliation(s)
- Khanh Dinh Quoc Nguyen
- Department of Chemistry and Biochemistry, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Michael Vigers
- Department of Chemical Engineering, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Eric Sefah
- C. Eugene Bennett Department of Chemistry, West Virginia UniversityMorgantownUnited States
| | - Susanna Seppälä
- Department of Chemical Engineering, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Jennifer Paige Hoover
- Department of Chemistry and Biochemistry, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Nicole Star Schonenbach
- Department of Chemical Engineering, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Blake Mertz
- C. Eugene Bennett Department of Chemistry, West Virginia UniversityMorgantownUnited States
| | - Michelle Ann O'Malley
- Department of Chemical Engineering, University of California, Santa BarbaraSanta BarbaraUnited States
| | - Songi Han
- Department of Chemistry and Biochemistry, University of California, Santa BarbaraSanta BarbaraUnited States
- Department of Chemical Engineering, University of California, Santa BarbaraSanta BarbaraUnited States
| |
Collapse
|
8
|
Yang X, Chen Y, Wang H, Fu X, Kural KC, Cao H, Li Y. Schizophrenia Plays a Negative Role in the Pathological Development of Myocardial Infarction at Multiple Biological Levels. Front Genet 2021; 12:607690. [PMID: 34149793 PMCID: PMC8211423 DOI: 10.3389/fgene.2021.607690] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 05/06/2021] [Indexed: 02/05/2023] Open
Abstract
It has shown that schizophrenia (SCZ) is associated with a higher chance of myocardial infarction (MI) and increased mortality. However, the underlying mechanism is largely unknown. Here, we first constructed a literature-based genetic pathway linking SCZ and MI, and then we tested the expression levels of the genes involved in the pathway by a meta-analysis using nine gene expression datasets of MI. In addition, a literature-based data mining process was conducted to explore the connection between SCZ at different levels: small molecules, complex molecules, and functional classes. The genetic pathway revealed nine genes connecting SCZ and MI. Specifically, SCZ activates two promoters of MI (IL6 and CRP) and deactivates seven inhibitors of MI (ADIPOQ, SOD2, TXN, NGF, ADORA1, NOS1, and CTNNB1), suggesting that no protective role of SCZ in MI was detected. Meta-analysis showed that one promoter of MI (CRP) presented no significant increase, and six out of seven genetic inhibitors of MI demonstrated minor to moderately increased expression. Therefore, the elevation of CRP and inhibition of the six inhibitors of MI by SCZ could be critical pathways to promote MI. Nine other regulators of MI were influenced by SCZ, including two gene families (inflammatory cytokine and IL1 family), five small molecules (lipid peroxide, superoxide, ATP, ascorbic acid, melatonin, arachidonic acid), and two complexes (CaM kinase 2 and IL23). Our results suggested that SCZ promotes the development and progression of MI at different levels, including genes, small molecules, complex molecules, and functional classes.
Collapse
Affiliation(s)
- Xiaorong Yang
- Department of Outpatient, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Yao Chen
- Department of Outpatient, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Huiyao Wang
- Mental Health Center of West China Hospital, Sichuan University, Chengdu, China
| | - Xia Fu
- Department of Outpatient, West China Hospital, Sichuan University/West China School of Nursing, Sichuan University, Chengdu, China
| | - Kamil Can Kural
- School of Systems Biology, George Mason University (GMU), Fairfax, VA, United States
| | - Hongbao Cao
- School of Systems Biology, George Mason University (GMU), Fairfax, VA, United States.,Department of Psychiatry, First Hospital/First Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Ying Li
- The Center of Gerontology and Geriatrics, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Kennedy C. That was then, this is now: the development of our knowledge and understanding of P2 receptor subtypes. Purinergic Signal 2021; 17:9-23. [PMID: 33527235 PMCID: PMC7954963 DOI: 10.1007/s11302-021-09763-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 01/06/2021] [Indexed: 11/17/2022] Open
Abstract
P2 receptors are present in virtually all tissues and cell types in the human body, and they mediate the physiological and pharmacological actions of extracellular purine and pyrimidine nucleotides. They were first characterised and named by Geoff Burnstock in 1978, then subdivided into P2X and P2Y purinoceptors in 1985 on the basis of pharmacological criteria in functional studies on native receptors. Molecular cloning of receptors in the 1990s revealed P2X receptors to comprise seven different subunits that interact to produce functional homo- and heterotrimeric ligand-gated cation channels. A family of eight P2Y G protein-coupled receptors were also cloned, which can form homo- and heterodimers. Deep insight into the molecular mechanisms of agonist and antagonist action has been provided by more recent determination of the tertiary and quaternary structures of several P2X and P2Y receptor subtypes. Agonists and antagonists that are highly selective for individual subtypes are now available and some are in clinical use. This has all come about because of the intelligence, insight and drive of the force of nature that was Geoff Burnstock.
Collapse
Affiliation(s)
- Charles Kennedy
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, John Arbuthnott Building, 161 Cathedral St, Glasgow, G4 0RE, Scotland, UK.
| |
Collapse
|
10
|
Procopio MC, Lauro R, Nasso C, Carerj S, Squadrito F, Bitto A, Di Bella G, Micari A, Irrera N, Costa F. Role of Adenosine and Purinergic Receptors in Myocardial Infarction: Focus on Different Signal Transduction Pathways. Biomedicines 2021; 9:biomedicines9020204. [PMID: 33670488 PMCID: PMC7922652 DOI: 10.3390/biomedicines9020204] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Accepted: 02/15/2021] [Indexed: 12/24/2022] Open
Abstract
Myocardial infarction (MI) is a dramatic event often caused by atherosclerotic plaque erosion or rupture and subsequent thrombotic occlusion of a coronary vessel. The low supply of oxygen and nutrients in the infarcted area may result in cardiomyocytes necrosis, replacement of intact myocardium with non-contractile fibrous tissue and left ventricular (LV) function impairment if blood flow is not quickly restored. In this review, we summarized the possible correlation between adenosine system, purinergic system and Wnt/β-catenin pathway and their role in the pathogenesis of cardiac damage following MI. In this context, several pathways are involved and, in particular, the adenosine receptors system shows different interactions between its members and purinergic receptors: their modulation might be effective not only for a normal functional recovery but also for the treatment of heart diseases, thus avoiding fibrosis, reducing infarcted area and limiting scaring. Similarly, it has been shown that Wnt/β catenin pathway is activated following myocardial injury and its unbalanced activation might promote cardiac fibrosis and, consequently, LV systolic function impairment. In this regard, the therapeutic benefits of Wnt inhibitors use were highlighted, thus demonstrating that Wnt/β-catenin pathway might be considered as a therapeutic target to prevent adverse LV remodeling and heart failure following MI.
Collapse
Affiliation(s)
- Maria Cristina Procopio
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Rita Lauro
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Chiara Nasso
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Scipione Carerj
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Francesco Squadrito
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Alessandra Bitto
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Gianluca Di Bella
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| | - Antonio Micari
- Department of Biomedical and Dental Sciences and Morphological and Functional Imaging, University of Messina, A.O.U. Policlinic “G. Martino”, 98165 Messina, Italy;
| | - Natasha Irrera
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
- Correspondence: ; Tel.: +39-090-221-3093; Fax: +39-090-221-23-81
| | - Francesco Costa
- Department of Clinical and Experimental Medicine, University of Messina, 98165 Messina, Italy; (M.C.P.); (R.L.); (C.N.); (S.C.); (F.S.); (A.B.); (G.D.B.); (F.C.)
| |
Collapse
|
11
|
Molecular pharmacology of P2Y receptor subtypes. Biochem Pharmacol 2020; 187:114361. [PMID: 33309519 DOI: 10.1016/j.bcp.2020.114361] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 12/06/2020] [Accepted: 12/07/2020] [Indexed: 02/06/2023]
Abstract
Professor Geoffrey Burnstock proposed the concept of purinergic signaling via P1 and P2 receptors. P2Y receptors are G-protein-coupled receptors (GPCRs) for extracellular adenine and uracil nucleotides. Eight mammalian P2Y receptor subtypes have been identified. They are divided into two subgroups (P2Y1, P2Y2, P2Y4, P2Y6, and P2Y11) and (P2Y12, P2Y13, and P2Y14). P2Y receptors are found in almost all cells and mediate responses in physiology and pathophysiology including pain and inflammation. The antagonism of platelet P2Y12 receptors by cangrelor, ticagrelor or active metabolites of the thienopyridine compounds ticlopidine, clopidogrel and prasugrel reduces the ADP-induced platelet aggregation in patients with thrombotic complications of vascular diseases. The nucleotide agonist diquafosol acting at P2Y2 receptors is used for the treatment of the dry eye syndrome. Structural information obtained by crystallography of the human P2Y1 and P2Y12 receptor proteins, site-directed mutagenesis and molecular modeling will facilitate the rational design of novel selective drugs.
Collapse
|
12
|
Ralevic V. History of Geoff Burnstock's research on P2 receptors. Biochem Pharmacol 2020; 187:114358. [PMID: 33279495 DOI: 10.1016/j.bcp.2020.114358] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 11/30/2022]
Abstract
Geoffrey Burnstock is a purinergic signalling legend who's discoveries and conceptualisation created and shaped the field. His scientific achievements were extraordinary and sustained. They included his demonstration that ATP can act as a neurotransmitter and hence extracellular signalling molecule, which he championed despite considerable initial opposition to his proposal that ATP acts outside of its role as an energy source inside cells. He led on purine receptor classification: initially of the P1 and P2 receptor families, then the P2X and P2Y receptor families, and then subtypes of P2X and P2Y receptors. This was achieved across several decades as he conceptualised and made sense of the emerging and growing evidence that there were multiple receptor subtypes for ATP and other nucleotides. He made discoveries about short term and long term/trophic purinergic signalling. He was a leader in the field for over 50 years. He inspired many and was a great colleague and mentor. I had the privilege of spending over 10 years (from 1985) with Geoff at the Department of Anatomy and Developmental Biology, University College London. This review is a personal perspective of some of Geoff's research on P2 receptors carried out during that time. It is a tribute to Geoff who I regarded with enormous respect and admiration.
Collapse
Affiliation(s)
- Vera Ralevic
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, NG7 2UH, United Kingdom.
| |
Collapse
|
13
|
Ralevic V. Purinergic signalling in the cardiovascular system-a tribute to Geoffrey Burnstock. Purinergic Signal 2020; 17:63-69. [PMID: 33151503 PMCID: PMC7954917 DOI: 10.1007/s11302-020-09734-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 09/13/2020] [Indexed: 01/02/2023] Open
Abstract
Geoffrey Burnstock made groundbreaking discoveries on the physiological roles of purinergic receptors and led on P2 purinergic receptor classification. His knowledge, vision and leadership inspired and influenced the international scientific community. I had the privilege of spending over 10 years (from 1985) with Geoff at the Department of Anatomy and Developmental Biology, initially as a PhD student and then as a postdoctoral research fellow. I regarded him with enormous admiration and affection. This review on purinergic signalling in the cardiovascular system is a tribute to Geoff. It includes some personal recollections of Geoff.
Collapse
Affiliation(s)
- Vera Ralevic
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham, NG7 2UH, UK.
| |
Collapse
|
14
|
Neumann A, Müller CE, Namasivayam V. P2Y
1
‐like nucleotide receptors—Structures, molecular modeling, mutagenesis, and oligomerization. WILEY INTERDISCIPLINARY REVIEWS-COMPUTATIONAL MOLECULAR SCIENCE 2020. [DOI: 10.1002/wcms.1464] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Alexander Neumann
- Department of Pharmaceutical and Medicinal Chemistry, PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Sciences Bonn (PSB) University of Bonn Bonn Germany
- Research Training Group 1873, University of Bonn Bonn Germany
| | - Christa E. Müller
- Department of Pharmaceutical and Medicinal Chemistry, PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Sciences Bonn (PSB) University of Bonn Bonn Germany
- Research Training Group 1873, University of Bonn Bonn Germany
| | - Vigneshwaran Namasivayam
- Department of Pharmaceutical and Medicinal Chemistry, PharmaCenter Bonn, Pharmaceutical Institute, Pharmaceutical Sciences Bonn (PSB) University of Bonn Bonn Germany
| |
Collapse
|
15
|
Distinct P2Y Receptors Mediate Extension and Retraction of Microglial Processes in Epileptic and Peritumoral Human Tissue. J Neurosci 2020; 40:1373-1388. [PMID: 31896671 DOI: 10.1523/jneurosci.0218-19.2019] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/12/2022] Open
Abstract
Microglia exhibit multiple, phenotype-dependent motility patterns often triggered by purinergic stimuli. However, little data exist on motility of human microglia in pathological situations. Here we examine motility of microglia stained with a fluorescent lectin in tissue slices from female and male epileptic patients diagnosed with mesial temporal lobe epilepsy or cortical glioma (peritumoral cortex). Microglial shape varied from ramified to amoeboid cells predominantly in regions of high neuronal loss or closer to a tumor. Live imaging revealed unstimulated or purine-induced microglial motilities, including surveillance movements, membrane ruffling, and process extension or retraction. At different concentrations, ADP triggered opposing motilities. Low doses triggered process extension. It was suppressed by P2Y12 receptor antagonists, which also reduced process length and surveillance movements. Higher purine doses caused process retraction and membrane ruffling, which were blocked by joint application of P2Y1 and P2Y13 receptor antagonists. Purinergic effects on motility were similar for all microglia tested. Both amoeboid and ramified cells from mesial temporal lobe epilepsy or peritumoral cortex tissue expressed P2Y12 receptors. A minority of microglia expressed the adenosine A2A receptor, which has been linked with process withdrawal of rodent cells. Laser-mediated tissue damage let us test the functional significance of these effects. Moderate damage induced microglial process extension, which was blocked by P2Y12 receptor antagonists. Overall, the purine-induced motility of human microglia in epileptic tissue is similar to that of rodent microglia in that the P2Y12 receptor initiates process extension. It differs in that retraction is triggered by joint activation of P2Y1/P2Y13 receptors.SIGNIFICANCE STATEMENT Microglial cells are brain-resident immune cells with multiple functions in healthy or diseased brains. These diverse functions are associated with distinct phenotypes, including different microglial shapes. In the rodent, purinergic signaling is associated with changes in cell shape, such as process extension toward tissue damage. However, there are little data on living human microglia, especially in diseased states. We developed a reliable technique to stain microglia from epileptic and glioma patients to examine responses to purines. Low-intensity purinergic stimuli induced process extension, as in rodents. In contrast, high-intensity stimuli triggered a process withdrawal mediated by both P2Y1 and P2Y13 receptors. P2Y1/P2Y13 receptor activation has not previously been linked to microglial morphological changes.
Collapse
|
16
|
Vallon V, Unwin R, Inscho EW, Leipziger J, Kishore BK. Extracellular Nucleotides and P2 Receptors in Renal Function. Physiol Rev 2019; 100:211-269. [PMID: 31437091 DOI: 10.1152/physrev.00038.2018] [Citation(s) in RCA: 64] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The understanding of the nucleotide/P2 receptor system in the regulation of renal hemodynamics and transport function has grown exponentially over the last 20 yr. This review attempts to integrate the available data while also identifying areas of missing information. First, the determinants of nucleotide concentrations in the interstitial and tubular fluids of the kidney are described, including mechanisms of cellular release of nucleotides and their extracellular breakdown. Then the renal cell membrane expression of P2X and P2Y receptors is discussed in the context of their effects on renal vascular and tubular functions. Attention is paid to effects on the cortical vasculature and intraglomerular structures, autoregulation of renal blood flow, tubuloglomerular feedback, and the control of medullary blood flow. The role of the nucleotide/P2 receptor system in the autocrine/paracrine regulation of sodium and fluid transport in the tubular and collecting duct system is outlined together with its role in integrative sodium and fluid homeostasis and blood pressure control. The final section summarizes the rapidly growing evidence indicating a prominent role of the extracellular nucleotide/P2 receptor system in the pathophysiology of the kidney and aims to identify potential therapeutic opportunities, including hypertension, lithium-induced nephropathy, polycystic kidney disease, and kidney inflammation. We are only beginning to unravel the distinct physiological and pathophysiological influences of the extracellular nucleotide/P2 receptor system and the associated therapeutic perspectives.
Collapse
Affiliation(s)
- Volker Vallon
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Robert Unwin
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Edward W Inscho
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Jens Leipziger
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| | - Bellamkonda K Kishore
- Departments of Medicine and Pharmacology, University of California San Diego & VA San Diego Healthcare System, San Diego, California; Centre for Nephrology, Division of Medicine, University College London, London, United Kingdom; IMED ECD CVRM R&D, AstraZeneca, Gothenburg, Sweden; Department of Medicine, Division of Nephrology, The University of Alabama at Birmingham, Birmingham, Alabama; Department of Biomedicine/Physiology, Aarhus University, Aarhus, Denmark; Departments of Internal Medicine and Nutrition and Integrative Physiology, and Center on Aging, University of Utah Health & Nephrology Research, VA Salt Lake City Healthcare System, Salt Lake City, Utah
| |
Collapse
|
17
|
von Kügelgen I. Pharmacology of P2Y receptors. Brain Res Bull 2019; 151:12-24. [PMID: 30922852 DOI: 10.1016/j.brainresbull.2019.03.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 03/04/2019] [Accepted: 03/17/2019] [Indexed: 01/17/2023]
Abstract
P2Y receptors are G-protein-coupled receptors (GPCRs) for extracellular nucleotides. There are eight mammalian P2Y receptor subtypes divided into two subgroups (P2Y1, P2Y2, P2Y4, P2Y6, and P2Y11) and (P2Y12, P2Y13, and P2Y14). The P2Y receptors are expressed in various cell types and play important roles in physiology and pathophysiology including inflammatory responses and neuropathic pain. The antagonism of P2Y12 receptors is used in pharmacotherapy for the prevention and therapy of cardiovascular events. The nucleoside analogue ticagrelor and active metabolites of the thienopyridine compounds ticlopidine, clopidogrel and prasugrel inhibit platelet P2Y12 receptors and reduce thereby platelet aggregation. The P2Y2 receptor agonist diquafosol is used for the treatment of the dry eye syndrome. The P2Y receptor subtypes differ in their amino acid sequences, their pharmacological profiles and their signaling transduction pathways. Recently, selective receptor ligands have been developed for all subtypes. The published crystal structures of the human P2Y1 and P2Y12 receptors as well as receptor models will facilitate the development of novel drugs for pharmacotherapy.
Collapse
Affiliation(s)
- Ivar von Kügelgen
- Department of Pharmacology and Toxicology, Pharma Center, University of Bonn, D-53127, Bonn, Germany.
| |
Collapse
|
18
|
Zhou Z, Matsumoto T, Jankowski V, Pernow J, Mustafa SJ, Duncker DJ, Merkus D. Uridine adenosine tetraphosphate and purinergic signaling in cardiovascular system: An update. Pharmacol Res 2019; 141:32-45. [PMID: 30553823 PMCID: PMC6685433 DOI: 10.1016/j.phrs.2018.12.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/26/2018] [Accepted: 12/12/2018] [Indexed: 02/07/2023]
Abstract
Uridine adenosine tetraphosphate (Up4A), biosynthesized by activation of vascular endothelial growth factor receptor (VEGFR) 2, was initially identified as a potent endothelium-derived vasoconstrictor in perfused rat kidney. Subsequently, the effect of Up4A on vascular tone regulation was intensively investigated in arteries isolated from different vascular beds in rodents including rat pulmonary arteries, aortas, mesenteric and renal arteries as well as mouse aortas, in which Up4A produces vascular contraction. In contrast, Up4A produces vascular relaxation in porcine coronary small arteries and rat aortas. Intravenous infusion of Up4A into conscious rats or mice decreases blood pressure, and intravenous bolus injection of Up4A into anesthetized mice increases coronary blood flow, indicating an overall vasodilator influence in vivo. Although Up4A is the first dinucleotide described that contains both purine and pyrimidine moieties, its cardiovascular effects are exerted mainly through activation of purinergic receptors. These effects not only encompass regulation of vascular tone, but also endothelial angiogenesis, smooth muscle cell proliferation and migration, and vascular calcification. Furthermore, this review discusses a potential role for Up4A in cardiovascular pathophysiology, as plasma levels of Up4A are elevated in juvenile hypertensive patients and Up4A-mediated vascular purinergic signaling changes in cardiovascular disease such as hypertension, diabetes, atherosclerosis and myocardial infarction. Better understanding the vascular effect of the novel dinucleotide Up4A and the purinergic signaling mechanisms mediating its effects will enhance its potential as target for treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Zhichao Zhou
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden.
| | - Takayuki Matsumoto
- Department of Physiology and Morphology, Institute of Medicinal Chemistry, Hoshi University, Tokyo, Japan
| | - Vera Jankowski
- RWTH-Aachen, Institute for Molecular Cardiovascular Research, Aachen, Germany
| | - John Pernow
- Division of Cardiology, Department of Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - S Jamal Mustafa
- Department of Physiology, Pharmacology & Neuroscience, Center for Cardiovascular and Respiratory Sciences, Clinical and Translational Science Institute, West Virginia University, Morgantown, WV, United States
| | - Dirk J Duncker
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Daphne Merkus
- Division of Experimental Cardiology, Department of Cardiology, Thoraxcenter, Erasmus University Medical Center, Rotterdam, the Netherlands
| |
Collapse
|
19
|
Felce JH, MacRae A, Davis SJ. Constraints on GPCR Heterodimerization Revealed by the Type-4 Induced-Association BRET Assay. Biophys J 2019; 116:31-41. [PMID: 30558888 PMCID: PMC6341220 DOI: 10.1016/j.bpj.2018.09.034] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 09/14/2018] [Accepted: 09/21/2018] [Indexed: 01/24/2023] Open
Abstract
G-protein-coupled receptors (GPCRs) comprise the largest and most pharmacologically important family of cell-surface receptors encoded by the human genome. In many instances, the distinct signaling behavior of certain GPCRs has been explained in terms of the formation of heteromers with, for example, distinct signaling properties and allosteric cross-regulation. Confirmation of this has, however, been limited by the paucity of reliable methods for probing heteromeric GPCR interactions in situ. The most widely used assays for GPCR stoichiometry, based on resonance energy transfer, are unsuited to reporting heteromeric interactions. Here, we describe a targeted bioluminescence resonance energy transfer (BRET) assay, called type-4 BRET, which detects both homo- and heteromeric interactions using induced multimerization of protomers within such complexes, at constant expression. Using type-4 BRET assays, we investigate heterodimerization among known GPCR homodimers: the CXC chemokine receptor 4 and sphingosine-1-phosphate receptors. We observe that CXC chemokine receptor 4 and sphingosine-1-phosphate receptors can form heterodimers with GPCRs from their immediate subfamilies but not with more distantly related receptors. We also show that heterodimerization appears to disrupt homodimeric interactions, suggesting the sharing of interfaces. Broadly, these observations indicate that heterodimerization results from the divergence of homodimeric receptors and will therefore likely be restricted to closely related homodimeric GPCRs.
Collapse
Affiliation(s)
- James H Felce
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom; Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, United Kingdom.
| | - Alasdair MacRae
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom; Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, United Kingdom
| | - Simon J Davis
- MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, United Kingdom; Radcliffe Department of Medicine, University of Oxford, Oxford, OX3 9DU, United Kingdom
| |
Collapse
|
20
|
Haque ME, Kim IS, Jakaria M, Akther M, Choi DK. Importance of GPCR-Mediated Microglial Activation in Alzheimer's Disease. Front Cell Neurosci 2018; 12:258. [PMID: 30186116 PMCID: PMC6110855 DOI: 10.3389/fncel.2018.00258] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/30/2018] [Indexed: 12/11/2022] Open
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder associated with impairment of cognition, memory deficits and behavioral abnormalities. Accumulation of amyloid beta (Aβ) is a characteristic hallmark of AD. Microglia express several GPCRs, which, upon activation by modulators, mediate microglial activation and polarization phenotype. This GPCR-mediated microglial activation has both protective and detrimental effects. Microglial GPCRs are involved in amyloid precursor protein (APP) cleavage and Aβ generation. In addition, microglial GPCRs are featured in the regulation of Aβ degradation and clearance through microglial phagocytosis and chemotaxis. Moreover, in response to Aβ binding on microglial Aβ receptors, they can trigger multiple inflammatory pathways. However, there is still a lack of insight into the mechanistic link between GPCR-mediated microglial activation and its pathological consequences in AD. Currently, the available drugs for the treatment of AD are mostly symptomatic and dominated by acetylcholinesterase inhibitors (AchEI). The selection of a specific microglial GPCR that is highly expressed in the AD brain and capable of modulating AD progression through Aβ generation, degradation and clearance will be a potential source of therapeutic intervention. Here, we have highlighted the expression and distribution of various GPCRs connected to microglial activation in the AD brain and their potential to serve as therapeutic targets of AD.
Collapse
Affiliation(s)
- Md Ezazul Haque
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, South Korea
| | - In-Su Kim
- Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease, Konkuk University, Chungju, South Korea
| | - Md Jakaria
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, South Korea
| | - Mahbuba Akther
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, South Korea
| | - Dong-Kug Choi
- Department of Applied Life Science, Graduate School, Konkuk University, Chungju, South Korea.,Department of Integrated Bioscience and Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease, Konkuk University, Chungju, South Korea
| |
Collapse
|
21
|
Dobrachinski F, Gerbatin RR, Sartori G, Golombieski RM, Antoniazzi A, Nogueira CW, Royes LF, Fighera MR, Porciúncula LO, Cunha RA, Soares FAA. Guanosine Attenuates Behavioral Deficits After Traumatic Brain Injury by Modulation of Adenosinergic Receptors. Mol Neurobiol 2018; 56:3145-3158. [PMID: 30105669 DOI: 10.1007/s12035-018-1296-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/02/2018] [Indexed: 12/16/2022]
Abstract
Traumatic brain injury (TBI) is a leading cause of disability worldwide, triggering chronic neurodegeneration underlying cognitive and mood disorder still without therapeutic prospects. Based on our previous observations that guanosine (GUO) attenuates short-term neurochemical alterations caused by TBI, this study investigated the effects of chronical GUO treatment in behavioral, molecular, and morphological disturbances 21 days after trauma. Rats subject to TBI displayed mood (anxiety-like) and memory dysfunction. This was accompanied by a decreased expression of both synaptic (synaptophysin) and plasticity proteins (BDNF and CREB), a loss of cresyl violet-stained neurons, and increased astrogliosis and microgliosis in the hippocampus. Notably, chronic GUO treatment (7.5 mg/kg i.p. daily starting 1 h after TBI) prevented all these TBI-induced long-term behavioral, neurochemical, and morphological modifications. This neuroprotective effect of GUO was abrogated in the presence of the adenosine A1 receptor antagonist DPCPX (1 mg/kg) but unaltered by the adenosine A2A receptor antagonist SCH58261 (0.05 mg/kg). These findings show that a chronic GUO treatment prevents the long-term mood and memory dysfunction triggered by TBI, which involves adenosinergic receptors.
Collapse
Affiliation(s)
- Fernando Dobrachinski
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, UFSM, Santa Maria, RS, 97105-900, Brazil
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Rogério R Gerbatin
- Laboratory of Exercise Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Gláubia Sartori
- Laboratory of Synthesis, Reactivity and Pharmacological Evaluating and Toxicology of Organochalcogens, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Ronaldo M Golombieski
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, UFSM, Santa Maria, RS, 97105-900, Brazil
| | - Alfredo Antoniazzi
- Laboratory of Biotechnology and Animal Reproduction - BioRep Veterinary Hospital, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Cristina W Nogueira
- Laboratory of Synthesis, Reactivity and Pharmacological Evaluating and Toxicology of Organochalcogens, Department of Biochemistry and Molecular Biology, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Luiz F Royes
- Laboratory of Exercise Biochemistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Michele R Fighera
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, UFSM, Santa Maria, RS, 97105-900, Brazil
- Department of Neuropsychiatry, Health Sciences Center, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Lisiane O Porciúncula
- Laboratory of Studies on the Purinergic System, Department of Biochemistry / ICBS, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Rodrigo A Cunha
- CNC - Center for Neurosciences and Cell Biology, University of Coimbra, 3004-504, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, 3004-504, Coimbra, Portugal
| | - Félix A A Soares
- Department of Biochemistry and Molecular Biology, Center for Natural and Exact Sciences, Federal University of Santa Maria, UFSM, Santa Maria, RS, 97105-900, Brazil.
| |
Collapse
|
22
|
Porzionato A, Stocco E, Guidolin D, Agnati L, Macchi V, De Caro R. Receptor-Receptor Interactions of G Protein-Coupled Receptors in the Carotid Body: A Working Hypothesis. Front Physiol 2018; 9:697. [PMID: 29930516 PMCID: PMC6000251 DOI: 10.3389/fphys.2018.00697] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Accepted: 05/18/2018] [Indexed: 12/15/2022] Open
Abstract
In the carotid body (CB), a wide series of neurotransmitters and neuromodulators have been identified. They are mainly produced and released by type I cells and act on many different ionotropic and metabotropic receptors located in afferent nerve fibers, type I and II cells. Most metabotropic receptors are G protein-coupled receptors (GPCRs). In other transfected or native cells, GPCRs have been demonstrated to establish physical receptor–receptor interactions (RRIs) with formation of homo/hetero-complexes (dimers or receptor mosaics) in a dynamic monomer/oligomer equilibrium. RRIs modulate ligand binding, signaling, and internalization of GPCR protomers and they are considered of relevance for physiology, pharmacology, and pathology of the nervous system. We hypothesize that RRI may also occur in the different structural elements of the CB (type I cells, type II cells, and afferent fibers), with potential implications in chemoreception, neuromodulation, and tissue plasticity. This ‘working hypothesis’ is supported by literature data reporting the contemporary expression, in type I cells, type II cells, or afferent terminals, of GPCRs which are able to physically interact with each other to form homo/hetero-complexes. Functional data about cross-talks in the CB between different neurotransmitters/neuromodulators also support the hypothesis. On the basis of the above findings, the most significant homo/hetero-complexes which could be postulated in the CB include receptors for dopamine, adenosine, ATP, opioids, histamine, serotonin, endothelin, galanin, GABA, cannabinoids, angiotensin, neurotensin, and melatonin. From a methodological point of view, future studies should demonstrate the colocalization in close proximity (less than 10 nm) of the above receptors, through biophysical (i.e., bioluminescence/fluorescence resonance energy transfer, protein-fragment complementation assay, total internal reflection fluorescence microscopy, fluorescence correlation spectroscopy and photoactivated localization microscopy, X-ray crystallography) or biochemical (co-immunoprecipitation, in situ proximity ligation assay) methods. Moreover, functional approaches will be able to show if ligand binding to one receptor produces changes in the biochemical characteristics (ligand recognition, decoding, and trafficking processes) of the other(s). Plasticity aspects would be also of interest, as development and environmental stimuli (chronic continuous or intermittent hypoxia) produce changes in the expression of certain receptors which could potentially invest the dynamic monomer/oligomer equilibrium of homo/hetero-complexes and the correlated functional implications.
Collapse
Affiliation(s)
| | - Elena Stocco
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Diego Guidolin
- Department of Neuroscience, University of Padua, Padua, Italy
| | - Luigi Agnati
- Department of Diagnostic, Clinical Medicine and Public Health, University of Modena and Reggio Emilia, Modena, Italy
| | - Veronica Macchi
- Department of Neuroscience, University of Padua, Padua, Italy
| | | |
Collapse
|
23
|
Abstract
P2Y receptors (P2YRs) are a family of G protein-coupled receptors activated by extracellular nucleotides. Physiological P2YR agonists include purine and pyrimidine nucleoside di- and triphosphates, such as ATP, ADP, UTP, UDP, nucleotide sugars, and dinucleotides. Eight subtypes exist, P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, P2Y13, and P2Y14, which represent current or potential future drug targets. Here we provide a comprehensive overview of ligands for the subgroup of the P2YR family that is activated by uracil nucleotides: P2Y2 (UTP, also ATP and dinucleotides), P2Y4 (UTP), P2Y6 (UDP), and P2Y14 (UDP, UDP-glucose, UDP-galactose). The physiological agonists are metabolically unstable due to their fast hydrolysis by ectonucleotidases. A number of agonists with increased potency, subtype-selectivity and/or enzymatic stability have been developed in recent years. Useful P2Y2R agonists include MRS2698 (6-01, highly selective) and PSB-1114 (6-05, increased metabolic stability). A potent and selective P2Y2R antagonist is AR-C118925 (10-01). For studies of the P2Y4R, MRS4062 (3-15) may be used as a selective agonist, while PSB-16133 (10-06) is a selective antagonist. Several potent P2Y6R agonists have been developed including 5-methoxyuridine 5'-O-((Rp)α-boranodiphosphate) (6-12), PSB-0474 (3-11), and MRS2693 (3-26). The isocyanate MRS2578 (10-08) is used as a selective P2Y6R antagonist, although its reactivity and low water-solubility are limiting. With MRS2905 (6-08), a potent and metabolically stable P2Y14R agonist is available, while PPTN (10-14) represents a potent and selective P2Y14R antagonist. The radioligand [3H]UDP can be used to label P2Y14Rs. In addition, several fluorescent probes have been developed. Uracil nucleotide-activated P2YRs show great potential as drug targets, especially in inflammation, cancer, cardiovascular and neurodegenerative diseases.
Collapse
|
24
|
Wang W, Qiao Y, Li Z. New Insights into Modes of GPCR Activation. Trends Pharmacol Sci 2018; 39:367-386. [DOI: 10.1016/j.tips.2018.01.001] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/03/2018] [Accepted: 01/08/2018] [Indexed: 12/22/2022]
|
25
|
Abstract
Platelet P2Y1 receptor signalling via RhoGTPases is necessary for platelet-dependent leukocyte recruitment, where no platelet aggregation is observed. We investigated signalling cascades involved in distinct P2Y1-dependent platelet activities in vitro, using specific inhibitors for phospholipase C (PLC) (U73122, to inhibit the canonical pathway), and RhoGTPases: Rac1 (NSC23766) and RhoA (ROCK inhibitor GSK429286). Human platelet rich plasma (for platelet aggregation) or isolated washed platelets (for chemotaxis assays) was treated with U73122, GSK429286 or NSC23766 prior to stimulation with adenosine diphosphate (ADP) or the P2Y1 specific agonist MRS2365. Aggregation, chemotaxis (towards f-MLP), or platelet-induced human neutrophil chemotaxis (PINC) towards macrophage derived chemokine (MDC) was assessed. Molecular docking of ADP and MRS2365 to P2Y1 was analysed using AutoDock Smina followed by GOLD molecular docking in the Accelrys Discovery Studio software. Inhibition of PLC, but not Rac1 or RhoA, suppressed platelet aggregation induced by ADP and MRS2365. In contrast, platelet chemotaxis and PINC, were significantly attenuated by inhibition of platelet Rac1 or RhoA, but not PLC. MRS2365, compared to ADP had a less pronounced effect on P2Y1-induced aggregation, but a similar efficacy to stimulate platelet chemotaxis and PINC, which might be explained by differences in molecular interaction of ADP compared to MRS2365 with the P2Y1 receptor. Platelet P2Y1 receptor activation during inflammation signals through alternate pathways involving Rho GTPases in contrast to canonical P2Y1 receptor induced PLC signalling. This might be explained by selective molecular interactions of ligands within the orthosteric site of the P2Y1 receptor.
Collapse
|
26
|
Nishimura A, Sunggip C, Oda S, Numaga-Tomita T, Tsuda M, Nishida M. Purinergic P2Y receptors: Molecular diversity and implications for treatment of cardiovascular diseases. Pharmacol Ther 2017. [DOI: 10.1016/j.pharmthera.2017.06.010] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
27
|
N -glycan-dependent cell-surface expression of the P2Y 2 receptor and N -glycan-independent distribution to lipid rafts. Biochem Biophys Res Commun 2017; 485:427-431. [DOI: 10.1016/j.bbrc.2017.02.061] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 02/10/2017] [Indexed: 01/23/2023]
|
28
|
Kosmowska B, Ossowska K, Głowacka U, Wardas J. Tremorolytic effect of 5'-chloro-5'-deoxy-(±)-ENBA, a potent and selective adenosine A1 receptor agonist, evaluated in the harmaline-induced model in rats. CNS Neurosci Ther 2017; 23:438-446. [PMID: 28371468 DOI: 10.1111/cns.12692] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/24/2017] [Accepted: 03/01/2017] [Indexed: 01/19/2023] Open
Abstract
AIM The aim of this study was to examine the role of adenosine A1 receptors in the harmaline-induced tremor in rats using 5'-chloro-5'-deoxy-(±)-ENBA (5'Cl5'd-(±)-ENBA), a brain-penetrant, potent, and selective adenosine A1 receptor agonist. METHODS Harmaline was injected at a dose of 15 mg/kg ip and tremor was measured automatically in force-plate actimeters by an increased averaged power in the frequency band of 9-15 Hz (AP2) and by tremor index (a difference in power between AP2 and averaged power in the frequency band of 0-8 Hz). The zif-268 mRNA expression was additionally analyzed by in situ hybridization in several brain structures. RESULTS 5'Cl5'd-(±)-ENBA (0.05-0.5 mg/kg ip) dose dependently reduced the harmaline-induced tremor and this effect was reversed by 8-cyclopentyl-1,3-dipropylxanthine (DPCPX), a selective antagonist of adenosine A1 receptors (1 mg/kg ip). Harmaline increased the zif-268 mRNA expression in the inferior olive, cerebellar cortex, ventroanterior/ventrolateral thalamic nuclei, and motor cortex. 5'Cl5'd-(±)-ENBA reversed these increases in all the above structures. DPCPX reduced the effect of 5'Cl5'd-(±)-ENBA on zif-268 mRNA in the motor cortex. CONCLUSION This study suggests that adenosine A1 receptors may be a potential target for the treatment of essential tremor.
Collapse
Affiliation(s)
- Barbara Kosmowska
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Krystyna Ossowska
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Urszula Głowacka
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| | - Jadwiga Wardas
- Department of Neuro-Psychopharmacology, Institute of Pharmacology, Polish Academy of Sciences, Kraków, Poland
| |
Collapse
|
29
|
Farran B. An update on the physiological and therapeutic relevance of GPCR oligomers. Pharmacol Res 2017; 117:303-327. [PMID: 28087443 DOI: 10.1016/j.phrs.2017.01.008] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 01/06/2017] [Accepted: 01/09/2017] [Indexed: 01/17/2023]
Abstract
The traditional view on GPCRs held that they function as single monomeric units composed of identical subunits. This notion was overturned by the discovery that GPCRs can form homo- and hetero-oligomers, some of which are obligatory, and can further assemble into receptor mosaics consisting of three or more protomers. Oligomerisation exerts significant impacts on receptor function and physiology, offering a platform for the diversification of receptor signalling, pharmacology, regulation, crosstalk, internalization and trafficking. Given their involvement in the modulation of crucial physiological processes, heteromers could constitute important therapeutic targets for a wide range of diseases, including schizophrenia, Parkinson's disease, substance abuse or obesity. This review aims at depicting the current developments in GPCR oligomerisation research, documenting various class A, B and C GPCR heteromers detected in vitro and in vivo using biochemical and biophysical approaches, as well as recently identified higher-order oligomeric complexes. It explores the current understanding of dimerization dynamics and the possible interaction interfaces that drive oligomerisation. Most importantly, it provides an inventory of the wide range of physiological processes and pathophysiological conditions to which GPCR oligomers contribute, surveying some of the oligomers that constitute potential drug targets. Finally, it delineates the efforts to develop novel classes of ligands that specifically target and tether to receptor oligomers instead of a single monomeric entity, thus ameliorating their ability to modulate GPCR function.
Collapse
Affiliation(s)
- Batoul Farran
- Department of Structural and Molecular Biology, University College London, Gower Street, London, WC1E 6BT, United Kingdom.
| |
Collapse
|
30
|
Nishimura A, Nishida M. [Purinergic signaling in cardiovascular system]. Nihon Yakurigaku Zasshi 2017; 149:84-90. [PMID: 28154303 DOI: 10.1254/fpj.149.84] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
|
31
|
Barańska J, Czajkowski R, Pomorski P. P2Y 1 Receptors - Properties and Functional Activities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017. [PMID: 28639247 DOI: 10.1007/5584_2017_57] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this chapter we try to show a comprehensive image of current knowledge of structure, activity and physiological role of the P2Y1 purinergic receptor. The structure, distribution and changes in the expression of this receptor are summarized, as well as the mechanism of its signaling activity by the intracellular calcium mobilization. We try to show the connection between the components of its G protein activation and cellular or physiological effects, starting from changes in protein phosphorylation patterns and ending with such remote effects as receptor-mediated apoptosis. The special emphasis is put on the role of the P2Y1 receptor in cancer cells and neuronal plasticity. We concentrate on the P2Y1 receptor, it is though impossible to completely abstract from other aspects of nucleotide signaling and cross-talk with other nucleotide receptors is here discussed. Especially, the balance between P2Y1 and P2Y12 receptors, sharing the same ligand but signaling through different pathways, is presented.
Collapse
Affiliation(s)
- Jolanta Barańska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., PL 02-093, Warsaw, Poland
| | - Rafał Czajkowski
- Laboratory of Spatial Memory, Department of Molecular and Cellular Neurobiology, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., PL 02-093, Warsaw, Poland
| | - Paweł Pomorski
- Laboratory of Molecular Basis of Cell Motility, Department of Cell Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur St., PL 02-093, Warsaw, Poland.
| |
Collapse
|
32
|
A1 Adenosine Receptor Activation Modulates Central Nervous System Development and Repair. Mol Neurobiol 2016; 54:8128-8139. [DOI: 10.1007/s12035-016-0292-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 11/08/2016] [Indexed: 01/22/2023]
|
33
|
Puchałowicz K, Baranowska-Bosiacka I, Dziedziejko V, Chlubek D. Purinergic signaling and the functioning of the nervous system cells. Cell Mol Biol Lett 2016; 20:867-918. [PMID: 26618572 DOI: 10.1515/cmble-2015-0050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 10/29/2015] [Indexed: 12/19/2022] Open
Abstract
Purinergic signaling in the nervous system has been the focus of a considerable number of studies since the 1970s. The P2X and P2Y receptors are involved in the initiation of purinergic signaling. They are very abundant in the central and peripheral nervous systems, where they are expressed on the surface of neurons and glial cells--microglia, astrocytes, oligodendrocytes and Schwann cells and the precursors of the latter two. Their ligands--extracellular nucleotides--are released in the physiological state by astrocytes and neurons forming synaptic connections, and are essential for the proper functioning of nervous system cells. Purinergic signaling plays a crucial role in neuromodulation, neurotransmission, myelination in the CNS and PNS, intercellular communication, the regulation of ramified microglia activity, the induction of the response to damaging agents, the modulation of synaptic activity and other glial cells by astrocytes, and the induction of astrogliosis. Understanding these mechanisms and the fact that P2 receptors and their ligands are involved in the pathogenesis of diseases of the nervous system may help in the design of drugs with different and more effective mechanisms of action.
Collapse
|
34
|
Suzuki T. [Hetero-oligomerization and Functional Interaction between Purinergic Receptors Expressed in Platelets to Regulate Platelet Shape Change]. YAKUGAKU ZASSHI 2016; 135:1335-40. [PMID: 26632148 DOI: 10.1248/yakushi.15-00178] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Adenosine and its precursors, ATP and ADP, exert various physiological effects via binding to purinergic receptors. We previously used co-immunoprecipitation, bioluminescence resonance energy transfer (BRET) and immunoelectron microscopy to demonstrate the hetero-oligomerization of purinergic receptor subtypes. Furthermore, pharmacological studies found significant changes in receptor-mediated signaling in human embryonic kidney (HEK) 293T cells co-transfected with these receptors. These findings suggest that heterodimers of purinergic receptors may have distinct pharmacological profiles, possibly due to dimerization-induced conformational changes, further suggesting that hetero-dimerization may be employed to "fine-tune" purinergic receptor signaling. Adenosine A(2A) receptor (A(2A)R), P2Y1 receptor (P2Y1R) and P2Y12 receptor (P2Y12R) are predominantly expressed on human platelets. ADP activates human platelets by stimulating both P2Y1R and P2Y12R, which act sequentially and in concert to achieve complete platelet aggregation. In contrast, adenosine stimulates Gs-coupled A(2A)R, followed by activativation of adenylate cyclase, leading to an increase in intracellular cAMP levels, which potently inhibits platelet activation. We examined the hetero-oligomerization and functional interactions of A(2A)R, P2Y1R, and P2Y12R. In HEK293T cells triply expressing all three receptors, hetero-oligomerization was observed among the three receptors. Additionally, P2Y1R agonist-evoked Ca(2+) signaling was significantly inhibited by co-treatment with an A(2A)R antagonist in HEK293T cells. In human platelets, we identified endogenous A(2A)R/P2Y1R and A(2A)R/P2Y12R heterodimers. We also observed functional Ca(2+)-signaling-related cross-talk similar to those found in HEK293T cells, and found that they appeared to affect platelet shape. These results collectively suggest that intermolecular signal transduction and specific conformational changes occur among components of the hetero-oligomers formed by these three receptors.
Collapse
Affiliation(s)
- Tokiko Suzuki
- Department of Cellular Signaling, Graduate School of Pharmaceutical Sciences, Tohoku University
| |
Collapse
|
35
|
Ribeiro-Filho AC, Buri MV, Barros CC, Dreyfuss JL, Nader HB, Justo GZ, Craveiro RB, Pesquero JB, Miranda A, Ferreira AT, Paredes-Gamero EJ. Functional and molecular evidence for heteromeric association of P2Y1 receptor with P2Y2 and P2Y4 receptors in mouse granulocytes. BMC Pharmacol Toxicol 2016; 17:29. [PMID: 27384918 PMCID: PMC4936188 DOI: 10.1186/s40360-016-0072-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 06/24/2016] [Indexed: 12/27/2022] Open
Abstract
BACKGROUND All hematopoietic cells express P2 receptors, however pharmacological characteristics such as expression and affinity in granulocytes are unknown. METHODS Pharmacological characteristics of P2 receptors were evaluated by Ca(2+) measurements using Fura-2 fluorophore. P2 receptors expression were analyzed by flow cytometry and RT-PCR. P2 interaction were shown by coimmunoprecipitation, western blotting and FRET. RESULTS Granulocytes were responsive to P2Y agonists, whereas P2X agonists were ineffective. Ca(2+) increase, elicited by ADP and UTP was dependent on intracellular stocks and sensitive to G-coupled receptor inhibition. Moreover, MRS2179, a specific antagonist of the P2Y1 receptor, abolished ADP response. Interestingly, ADP and UTP exhibited full heterologous desensitization, suggesting that these agonists interact with the same receptor. The heteromeric association between P2Y1 receptor and the P2Y2 and P2Y4 receptors was shown by immunoprecipitation and FRET analysis. CONCLUSION Clear evidence of heteromeric association of P2Y receptors was found during the evaluation of P2 receptors present in mice granulocytes, which could impact in the classical pharmacology of P2Y receptors in granulocytes.
Collapse
Affiliation(s)
- Antonio Carlos Ribeiro-Filho
- Centro Interdisciplinar de Investigação Bioquı́mica, Universidade de Mogi das Cruzes, Av. Dr Cândido Xavier de Almeida Souza, 200, Mogi das Cruzes, São Paulo, Brazil
| | - Marcus Vinicius Buri
- Departamento de Bioquímica, Universidade Federal de São Paulo, Rua Pedro de Toledo, 669 - 9° andar - Prédio de Pesquisa II, R. Três de Maio 100, São Paulo, São Paulo, Brazil
| | - Carlos Castilho Barros
- Departamento de Nutrição, Universidade Federal de Pelotas, R. Gomes Carneiro, n°1, 96010-610, Pelotas, Rio Grande do Sul, Brazil
| | - Juliana Luporini Dreyfuss
- Departamento de Bioquímica, Universidade Federal de São Paulo, Rua Pedro de Toledo, 669 - 9° andar - Prédio de Pesquisa II, R. Três de Maio 100, São Paulo, São Paulo, Brazil
| | - Helena Bonciani Nader
- Departamento de Bioquímica, Universidade Federal de São Paulo, Rua Pedro de Toledo, 669 - 9° andar - Prédio de Pesquisa II, R. Três de Maio 100, São Paulo, São Paulo, Brazil
| | - Giselle Zenker Justo
- Departamento de Bioquímica, Universidade Federal de São Paulo, Rua Pedro de Toledo, 669 - 9° andar - Prédio de Pesquisa II, R. Três de Maio 100, São Paulo, São Paulo, Brazil
| | - Rogério Bastos Craveiro
- Departamento de Biofísica, Universidade Federal de São Paulo, R. Botucatu 862, São Paulo, São Paulo, Brazil
| | - João Bosco Pesquero
- Departamento de Biofísica, Universidade Federal de São Paulo, R. Botucatu 862, São Paulo, São Paulo, Brazil
| | - Antonio Miranda
- Departamento de Biofísica, Universidade Federal de São Paulo, R. Botucatu 862, São Paulo, São Paulo, Brazil
| | - Alice Teixeira Ferreira
- Departamento de Biofísica, Universidade Federal de São Paulo, R. Botucatu 862, São Paulo, São Paulo, Brazil
| | - Edgar Julian Paredes-Gamero
- Centro Interdisciplinar de Investigação Bioquı́mica, Universidade de Mogi das Cruzes, Av. Dr Cândido Xavier de Almeida Souza, 200, Mogi das Cruzes, São Paulo, Brazil. .,Departamento de Bioquímica, Universidade Federal de São Paulo, Rua Pedro de Toledo, 669 - 9° andar - Prédio de Pesquisa II, R. Três de Maio 100, São Paulo, São Paulo, Brazil.
| |
Collapse
|
36
|
Nemoto W, Yamanishi Y, Limviphuvadh V, Saito A, Toh H. GGIP: Structure and sequence-based GPCR-GPCR interaction pair predictor. Proteins 2016; 84:1224-33. [PMID: 27191053 DOI: 10.1002/prot.25071] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2016] [Revised: 04/26/2016] [Accepted: 05/09/2016] [Indexed: 01/20/2023]
Abstract
G Protein-Coupled Receptors (GPCRs) are important pharmaceutical targets. More than 30% of currently marketed pharmaceutical medicines target GPCRs. Numerous studies have reported that GPCRs function not only as monomers but also as homo- or hetero-dimers or higher-order molecular complexes. Many GPCRs exert a wide variety of molecular functions by forming specific combinations of GPCR subtypes. In addition, some GPCRs are reportedly associated with diseases. GPCR oligomerization is now recognized as an important event in various biological phenomena, and many researchers are investigating this subject. We have developed a support vector machine (SVM)-based method to predict interacting pairs for GPCR oligomerization, by integrating the structure and sequence information of GPCRs. The performance of our method was evaluated by the Receiver Operating Characteristic (ROC) curve. The corresponding area under the curve was 0.938. As far as we know, this is the only prediction method for interacting pairs among GPCRs. Our method could accelerate the analyses of these interactions, and contribute to the elucidation of the global structures of the GPCR networks in membranes. Proteins 2016; 84:1224-1233. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Wataru Nemoto
- Division of Life Science and Engineering, School of Science and Engineering, Tokyo Denki University (TDU), Ishizaka, Hatoyama-Machi, Hiki-Gun, Saitama, 350-0394, Japan.,Computational Biology Research Center (CBRC), Advanced Industrial Science and Technology (AIST), AIST Tokyo Waterfront Bio-IT Research Building, 2-4-7 Aomi, Koto-Ku, Tokyo, 135-0064, Japan
| | - Yoshihiro Yamanishi
- Medical Institute of Bioregulation (MiB), Kyushu University, 3-1-1 Maidashi, Higashi-Ku, Fukuoka, 812-8582, Japan.,Institute for Advanced Study, Kyushu University, 6-10-1, Hakozaki, Higashi-ku, Fukuoka, 812-8581, Japan
| | - Vachiranee Limviphuvadh
- Bioinformatics Institute (BII), Agency for Science, Technology and Research (A*STAR), 30 Biopolis Street, #07-01 Matrix, 138671, Singapore
| | - Akira Saito
- Division of Life Science and Engineering, School of Science and Engineering, Tokyo Denki University (TDU), Ishizaka, Hatoyama-Machi, Hiki-Gun, Saitama, 350-0394, Japan
| | - Hiroyuki Toh
- Computational Biology Research Center (CBRC), Advanced Industrial Science and Technology (AIST), AIST Tokyo Waterfront Bio-IT Research Building, 2-4-7 Aomi, Koto-Ku, Tokyo, 135-0064, Japan.,Department of Biomedical Chemistry, School of Science and Technology, Kwansei Gakuin University, 2-1 Gakuen, Sanda-Shi, Hyogo, 669-1337, Japan
| |
Collapse
|
37
|
Hajjawi MOR, Patel JJ, Corcelli M, Arnett TR, Orriss IR. Lack of effect of adenosine on the function of rodent osteoblasts and osteoclasts in vitro. Purinergic Signal 2016; 12:247-58. [PMID: 26861849 DOI: 10.1007/s11302-016-9499-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/01/2016] [Indexed: 01/06/2023] Open
Abstract
Extracellular ATP, signalling through P2 receptors, exerts well-documented effects on bone cells, inhibiting mineral deposition by osteoblasts and stimulating the formation and resorptive activity of osteoclasts. The aims of this study were to determine the potential osteotropic effects of adenosine, the hydrolysis product of ATP, on primary bone cells in vitro. We determined the effect of exogenous adenosine on (1) the growth, alkaline phosphatase (TNAP) activity and bone-forming ability of osteoblasts derived from the calvariae of neonatal rats and mice and the marrow of juvenile rats and (2) the formation and resorptive activity of osteoclasts from juvenile mouse marrow. Reverse transcription polymerase chain reaction (RT-PCR) analysis showed marked differences in the expression of P1 receptors in osteoblasts from different sources. Whilst mRNA for the A1 and A2B receptors was expressed by all primary osteoblasts, A2A receptor expression was limited to rat bone marrow and mouse calvarial osteoblasts and the A3 receptor to rat bone marrow osteoblasts. We found that adenosine had no detectable effects on cell growth, TNAP activity or bone formation by rodent osteoblasts in vitro. The analogue 2-chloroadenosine, which is hydrolysed more slowly than adenosine, had no effects on rat or mouse calvarial osteoblasts but increased TNAP activity and bone formation by rat bone marrow osteoblasts by 30-50 % at a concentration of 1 μM. Osteoclasts were found to express the A2A, A2B and A3 receptors; however, neither adenosine (≤100 μM) nor 2-chloroadenosine (≤10 μM) had any effect on the formation or resorptive activity of mouse osteoclasts in vitro. These results suggest that adenosine, unlike ATP, is not a major signalling molecule in the bone.
Collapse
Affiliation(s)
- Mark O R Hajjawi
- Department of Cell and Developmental Biology, University College London, Anatomy Building, Gower Street, WC1E 6BT, London, UK
| | - Jessal J Patel
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Michelangelo Corcelli
- Department of Cell and Developmental Biology, University College London, Anatomy Building, Gower Street, WC1E 6BT, London, UK
| | - Timothy R Arnett
- Department of Cell and Developmental Biology, University College London, Anatomy Building, Gower Street, WC1E 6BT, London, UK.
| | - Isabel R Orriss
- Department of Cell and Developmental Biology, University College London, Anatomy Building, Gower Street, WC1E 6BT, London, UK.,Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| |
Collapse
|
38
|
von Kügelgen I, Hoffmann K. Pharmacology and structure of P2Y receptors. Neuropharmacology 2015; 104:50-61. [PMID: 26519900 DOI: 10.1016/j.neuropharm.2015.10.030] [Citation(s) in RCA: 172] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 10/22/2015] [Accepted: 10/23/2015] [Indexed: 01/30/2023]
Abstract
P2Y receptors are G-protein-coupled receptors (GPCRs) for extracellular nucleotides. There are eight mammalian P2Y receptor subtypes (P2Y1, P2Y2, P2Y4, P2Y6, P2Y11, P2Y12, P2Y13, and P2Y14). P2Y receptors are widely expressed and play important roles in physiology and pathophysiology. One important example is the ADP-induced platelet aggregation mediated by P2Y1 and P2Y12 receptors. Active metabolites of the thienopyridine compounds ticlopidine, clopidogrel and prasugrel as well as the nucleoside analogue ticagrelor block P2Y12 receptors and thereby platelet aggregation. These drugs are used for the prevention and therapy of cardiovascular events. Moreover, P2Y receptors play important roles in the nervous system. Adenine nucleotides modulate neuronal activity and neuronal fibre outgrowth by activation of P2Y1 receptors and control migration of microglia by P2Y12 receptors. UDP stimulates microglial phagocytosis through activation of P2Y6 receptors. There is evidence for a role for P2Y2 receptors in Alzheimer's disease pathology. The P2Y receptor subtypes are highly diverse in both their amino acid sequences and their pharmacological profiles. Selective receptor ligands have been developed for the pharmacological characterization of the receptor subtypes. The recently published three-dimensional crystal structures of the human P2Y1 and P2Y12 receptors will facilitate the development of therapeutic agents that selectively target P2Y receptors. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Ivar von Kügelgen
- Department of Pharmacology and Toxicology, Pharma Center, University of Bonn, D-53127 Bonn, Germany.
| | - Kristina Hoffmann
- Department of Pharmacology and Toxicology, Pharma Center, University of Bonn, D-53127 Bonn, Germany
| |
Collapse
|
39
|
Navarro G, Borroto-Escuela DO, Fuxe K, Franco R. Purinergic signaling in Parkinson's disease. Relevance for treatment. Neuropharmacology 2015. [PMID: 26211977 DOI: 10.1016/j.neuropharm.2015.07.024] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Purinergic signaling modulates dopaminergic neurotransmission in health and disease. Classically adenosine A1 and A2A receptors have been considered key for the fine tune control of dopamine actions in the striatum, the main CNS motor control center. The main adenosine signaling mechanism is via the cAMP pathway but the future will tell whether calcium signaling is relevant in adenosinergic control of striatal function. Very relevant is the recent approval in Japan of the adenosine A2A receptor antagonist, istradefylline, for use in Parkinson's disease patients. Purine nucleotides are also regulators of striatal dopamine neurotransmission via P2 purinergic receptors. In parallel to the alpha-synuclein hypothesis of Parkinson's disease etiology, purinergic P2X1 receptors have been identified as mediators of accumulation of the Lewy-body enriched protein alpha-synuclein. Of note is the expression in striatum of purinergic-receptor-containing heteromers that are potential targets of anti-Parkinson's disease therapies and should be taken into account in drug discovery programs. This article is part of the Special Issue entitled 'Purines in Neurodegeneration and Neuroregeneration'.
Collapse
Affiliation(s)
- Gemma Navarro
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| | - Dasiel O Borroto-Escuela
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden; Department of Earth, Life and Environmental Sciences, Section of Physiology, Campus Scientifico Enrico Mattei, University of Urbino, Urbino, Italy.
| | - Kjell Fuxe
- Department of Neuroscience, Karolinska Institutet, Stockholm, Sweden.
| | - Rafael Franco
- Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, Barcelona, Spain; Centro de Investigación en Red, Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
40
|
Abstract
There are nineteen different receptor proteins for adenosine, adenine and uridine nucleotides, and nucleotide sugars, belonging to three families of G protein-coupled adenosine and P2Y receptors, and ionotropic P2X receptors. The majority are functionally expressed in blood vessels, as purinergic receptors in perivascular nerves, smooth muscle and endothelial cells, and roles in regulation of vascular contractility, immune function and growth have been identified. The endogenous ligands for purine receptors, ATP, ADP, UTP, UDP and adenosine, can be released from different cell types within the vasculature, as well as from circulating blood cells, including erythrocytes and platelets. Many purine receptors can be activated by two or more of the endogenous ligands. Further complexity arises because of interconversion between ligands, notably adenosine formation from the metabolism of ATP, leading to complex integrated responses through activation of different subtypes of purine receptors. The enzymes responsible for this conversion, ectonucleotidases, are present on the surface of smooth muscle and endothelial cells, and may be coreleased with neurotransmitters from nerves. What selectivity there is for the actions of purines/pyrimidines comes from differential expression of their receptors within the vasculature. P2X1 receptors mediate the vasocontractile actions of ATP released as a neurotransmitter with noradrenaline (NA) from sympathetic perivascular nerves, and are located on the vascular smooth muscle adjacent to the nerve varicosities, the sites of neurotransmitter release. The relative contribution of ATP and NA as functional cotransmitters varies with species, type and size of blood vessel, neuronal firing pattern, the tone/pressure of the blood vessel, and in ageing and disease. ATP is also a neurotransmitter in non-adrenergic non-cholinergic perivascular nerves and mediates vasorelaxation via smooth muscle P2Y-like receptors. ATP and adenosine can act as neuromodulators, with the most robust evidence being for prejunctional inhibition of neurotransmission via A1 adenosine receptors, but also prejunctional excitation and inhibition of neurotransmission via P2X and P2Y receptors, respectively. P2Y2, P2Y4 and P2Y6 receptors expressed on the vascular smooth muscle are coupled to vasocontraction, and may have a role in pathophysiological conditions, when purines are released from damaged cells, or when there is damage to the protective barrier that is the endothelium. Adenosine is released during hypoxia to increase blood flow via vasodilator A2A and A2B receptors expressed on the endothelium and smooth muscle. ATP is released from endothelial cells during hypoxia and shear stress and can act at P2Y and P2X4 receptors expressed on the endothelium to increase local blood flow. Activation of endothelial purine receptors leads to the release of nitric oxide, hyperpolarising factors and prostacyclin, which inhibits platelet aggregation and thus ensures patent blood flow. Vascular purine receptors also regulate endothelial and smooth muscle growth, and inflammation, and thus are involved in the underlying processes of a number of cardiovascular diseases.
Collapse
Affiliation(s)
- Vera Ralevic
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom.
| | - William R Dunn
- School of Life Sciences, Queen's Medical Centre, University of Nottingham, Nottingham NG7 2UH, United Kingdom
| |
Collapse
|
41
|
Zhou Z, Sun C, Tilley SL, Mustafa SJ. Mechanisms underlying uridine adenosine tetraphosphate-induced vascular contraction in mouse aorta: Role of thromboxane and purinergic receptors. Vascul Pharmacol 2015; 73:78-85. [PMID: 25921923 DOI: 10.1016/j.vph.2015.04.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 04/13/2015] [Accepted: 04/20/2015] [Indexed: 01/12/2023]
Abstract
Uridine adenosine tetraphosphate (Up4A), a novel endothelium-derived vasoactive agent, is proposed to play a role in cardiovascular disorders and induces aortic contraction through activation of cyclooxygenases (COXs). We and others demonstrated that activation of A1 or A3 adenosine receptors (ARs) results in vascular contraction via thromboxane (TX) A2 production. However, the mechanisms of Up4A-induced vascular contraction in mouse aorta are not understood. We hypothesize that Up4A-induced aortic contraction is through COX-derived TXA2 production, which requires activation of A1 and/or A3AR. Concentration responses to Up4A were conducted in isolated aorta. The TXB2 production, a metabolite of TXA2, was also measured. Up4A (10(-9)-10(-5)M) produced a concentration-dependent contraction >70%, which was markedly attenuated by COX and COX1 but not by COX2 inhibition. Notably, Up4A-induced aortic contraction was blunted by both TX synthase inhibitor ozagrel and TXA2 receptor (TP) antagonist SQ29548. Surprisingly, A3AR deletion had no effect on Up4A-induced contraction. Moreover, A1AR deletion or antagonism as well as A1/A3AR deletion potentiated Up4A-induced aortic contraction, suggesting a vasodilator influence of A1AR. In contrast, non-selective purinergic P2 receptor antagonist PPADS significantly blunted Up4A-induced aortic contraction to a similar extent as selective P2X1R antagonist MRS2159, the latter of which was further reduced by addition of ozagrel. Endothelial denudation almost fully attenuated Up4A-induced contraction. Furthermore, Up4A (3μM) increased TXB2 formation, which was inhibited by either MRS2159 or ozagrel. In conclusion, Up4A-induced aortic contraction depends on activation of TX synthase and TP, which partially requires the activation of P2X1R but not A1 or A3AR through an endothelium-dependent mechanism.
Collapse
Affiliation(s)
- Zhichao Zhou
- Department of Physiology and Pharmacology, Center for Cardiovascular and Respiratory Sciences, Clinical and Translational Science Institute, West Virginia University, Morgantown, WV, USA
| | - Changyan Sun
- Department of Physiology and Pharmacology, Center for Cardiovascular and Respiratory Sciences, Clinical and Translational Science Institute, West Virginia University, Morgantown, WV, USA
| | - Stephen L Tilley
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
| | - S Jamal Mustafa
- Department of Physiology and Pharmacology, Center for Cardiovascular and Respiratory Sciences, Clinical and Translational Science Institute, West Virginia University, Morgantown, WV, USA.
| |
Collapse
|
42
|
Yashima S, Shimazaki A, Mitoma J, Nakagawa T, Abe M, Yamada H, Higashi H. Close association of B2 bradykinin receptors with P2Y2 ATP receptors. J Biochem 2015; 158:155-63. [PMID: 25713410 DOI: 10.1093/jb/mvv022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 02/10/2015] [Indexed: 11/12/2022] Open
Abstract
Two G-protein-coupled receptors (GPCRs) that couple with Gαq/11, B2 bradykinin (BK) receptor (B2R) and ATP/UTP receptor P2Y2 (P2Y2R), are ubiquitously expressed and responsible for vascular tone, inflammation, and pain. We analysed the cellular signalling of P2Y2Rs in cells that express B2Rs. B2R desensitization induced by BK or B2R internalization-inducing glycans cross-desensitized the P2Y2R response to ATP/UTP. Fluorescence resonance energy transfer from P2Y2R-AcGFP to B2R-DsRed was detected in the cells and on the cell surfaces, showing the close association of these GPCRs. BK- and ATP-induced cross-internalization of P2Y2R and B2R, respectively, was shown in a β-galactosidase complementation assay using P2Y2R or B2R fused to the H31R substituted α donor peptide of a β-galactosidase reporter enzyme (P2Y2R-α or B2R-α) with coexpression of the FYVE domain of endofin, an early endosome protein, fused to the M15 acceptor deletion mutant of β-galactosidase (the ω peptide, FYVE-ω). Arrestin recruitment to the GPCRs by cross-activation was also shown with the similar way. Coimmunoprecipitation showed that B2R and P2Y2R were closely associated in the cotransfected cells. These results indicate that B2R couples with P2Y2R and that these GPCRs act together to fine-tune cellular responsiveness. The collaboration between these receptors may permit rapid onset and turning off of biological events.
Collapse
Affiliation(s)
- Sayo Yashima
- Division of Glyco-Signal Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| | - Ayaka Shimazaki
- Division of Glyco-Signal Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| | - Junya Mitoma
- Division of Glyco-Signal Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| | - Tetsuto Nakagawa
- Division of Glyco-Signal Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| | - Maya Abe
- Division of Glyco-Signal Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| | - Hiroyuki Yamada
- Division of Glyco-Signal Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| | - Hideyoshi Higashi
- Division of Glyco-Signal Research, Institute of Molecular Biomembrane and Glycobiology, Tohoku Pharmaceutical University, Sendai, Miyagi 981-8558, Japan
| |
Collapse
|
43
|
Fragakis N, Antoniadis AP, Saviano M, Vassilikos V, Pappone C. The use of adenosine and adenosine triphosphate testing in the diagnosis, risk stratification and management of patients with syncope: current evidence and future perspectives. Int J Cardiol 2015; 183:267-73. [PMID: 25725201 DOI: 10.1016/j.ijcard.2015.01.089] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 12/08/2014] [Accepted: 01/03/2015] [Indexed: 10/24/2022]
Abstract
Syncope is a significant source of cardiovascular-related morbidity yet the etiology is frequently obscure and the identification of patients at highest risk is challenging. Adenosine (AD) and adenosine triphosphate (ATP) administrations have been suggested as potentially useful non-invasive tools in the diagnostic workup of patients with neurally-mediated or bradycardia-related syncope. It has been postulated that both compounds by modulating the autonomic innervation in the heart and exerting negative chronotropic and dromotropic effects in the conduction system, may unmask the mechanism of syncope. However, the clinical implications derived from the efficacy of both tests in the investigation of syncope remain unclear mainly due to inconclusive and occasionally contradictory results of published studies. This review article summarizes recent and past information in the use of ATP and AD in the investigation of syncope with emphasis on clinical trials. We present the current level of evidence for the use of these agents in clinical practice, identify areas where further research is warranted and highlight the future perspectives of these agents as complements to an accurate risk-stratification of patients with syncope.
Collapse
Affiliation(s)
- Nikolaos Fragakis
- 3rd Cardiology Department, Hippokration Hospital, Aristotle University Medical School, Thessaloniki, Greece.
| | - Antonios P Antoniadis
- 3rd Cardiology Department, Hippokration Hospital, Aristotle University Medical School, Thessaloniki, Greece
| | - Massimo Saviano
- Department of Arrhythmology, Maria Cecilia Hospital, Cotignola, Italy
| | - Vassilios Vassilikos
- 3rd Cardiology Department, Hippokration Hospital, Aristotle University Medical School, Thessaloniki, Greece
| | - Carlo Pappone
- Department of Arrhythmology, Maria Cecilia Hospital, Cotignola, Italy
| |
Collapse
|
44
|
Tehseen M, Liao C, Dacres H, Dumancic M, Trowell S, Anderson A. Oligomerisation of C. elegans olfactory receptors, ODR-10 and STR-112, in yeast. PLoS One 2014; 9:e108680. [PMID: 25254556 PMCID: PMC4177895 DOI: 10.1371/journal.pone.0108680] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Accepted: 09/02/2014] [Indexed: 01/08/2023] Open
Abstract
It is widely accepted that vertebrate G-Protein Coupled Receptors (GPCRs) associate with each other as homo- or hetero-dimers or higher-order oligomers. The C. elegans genome encodes hundreds of olfactory GPCRs, which may be expressed in fewer than a dozen chemosensory neurons, suggesting an opportunity for oligomerisation. Here we show, using three independent lines of evidence: co-immunoprecipitation, bioluminescence resonance energy transfer and a yeast two-hybrid assay that nematode olfactory receptors (ORs) oligomerise when heterologously expressed in yeast. Specifically, the nematode receptor ODR-10 is able to homo-oligomerise and can also form heteromers with the related nematode receptor STR-112. ODR-10 also oligomerised with the rat I7 OR but did not oligomerise with the human somatostatin receptor 5, a neuropeptide receptor. In this study, the question of functional relevance was not addressed and remains to be investigated.
Collapse
Affiliation(s)
- Muhammad Tehseen
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, Canberra, ACT, Australia
- Center for Desert Agriculture, Division of Biological and Environmental Sciences and Engineering, King Abdullah University of Science and Technology, Thuwal, Kingdom of Saudi Arabia
| | - Chunyan Liao
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, Canberra, ACT, Australia
| | - Helen Dacres
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, Canberra, ACT, Australia
| | - Mira Dumancic
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, Canberra, ACT, Australia
| | - Stephen Trowell
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, Canberra, ACT, Australia
| | - Alisha Anderson
- CSIRO Food Futures National Research Flagship & CSIRO Ecosystem Sciences, Australia, Canberra, ACT, Australia
- * E-mail:
| |
Collapse
|
45
|
Figueroa VA, Retamal MA, Cea LA, Salas JD, Vargas AA, Verdugo CA, Jara O, Martínez AD, Sáez JC. Extracellular gentamicin reduces the activity of connexin hemichannels and interferes with purinergic Ca(2+) signaling in HeLa cells. Front Cell Neurosci 2014; 8:265. [PMID: 25237294 PMCID: PMC4154469 DOI: 10.3389/fncel.2014.00265] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 08/18/2014] [Indexed: 01/15/2023] Open
Abstract
Gap junction channels (GJCs) and hemichannels (HCs) are composed of protein subunits termed connexins (Cxs) and are permeable to ions and small molecules. In most organs, GJCs communicate the cytoplasm of adjacent cells, while HCs communicate the intra and extracellular compartments. In this way, both channel types coordinate physiological responses of cell communities. Cx mutations explain several genetic diseases, including about 50% of autosomal recessive non-syndromic hearing loss. However, the possible involvement of Cxs in the etiology of acquired hearing loss remains virtually unknown. Factors that induce post-lingual hearing loss are diverse, exposure to gentamicin an aminoglycoside antibiotic, being the most common. Gentamicin has been proposed to block GJCs, but its effect on HCs remains unknown. In this work, the effect of gentamicin on the functional state of HCs was studied and its effect on GJCs was reevaluated in HeLa cells stably transfected with Cxs. We focused on Cx26 because it is the main Cx expressed in the cochlea of mammals where it participates in purinergic signaling pathways. We found that gentamicin applied extracellularly reduces the activity of HCs, while dye transfer across GJCs was not affected. HCs were also blocked by streptomycin, another aminoglycoside antibiotic. Gentamicin also reduced the adenosine triphosphate release and the HC-dependent oscillations of cytosolic free-Ca2+ signal. Moreover, gentamicin drastically reduced the Cx26 HC-mediated membrane currents in Xenopus laevis oocytes. Therefore, the extracellular gentamicin-induced inhibition of Cx HCs may adversely affect autocrine and paracrine signaling, including the purinergic one, which might partially explain its ototoxic effects.
Collapse
Affiliation(s)
- Vania A Figueroa
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile ; Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo Santiago, Chile
| | - Mauricio A Retamal
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo Santiago, Chile
| | - Luis A Cea
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile
| | - José D Salas
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo Santiago, Chile
| | - Aníbal A Vargas
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile
| | - Christian A Verdugo
- Centro de Fisiología Celular e Integrativa, Facultad de Medicina, Clínica Alemana Universidad del Desarrollo Santiago, Chile
| | - Oscar Jara
- Instituto Milenio, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Agustín D Martínez
- Instituto Milenio, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| | - Juan C Sáez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile Santiago, Chile ; Instituto Milenio, Centro Interdisciplinario de Neurociencia de Valparaíso, Universidad de Valparaíso Valparaíso, Chile
| |
Collapse
|
46
|
Tovo-Rodrigues L, Roux A, Hutz MH, Rohde LA, Woods AS. Functional characterization of G-protein-coupled receptors: a bioinformatics approach. Neuroscience 2014; 277:764-79. [PMID: 24997265 DOI: 10.1016/j.neuroscience.2014.06.049] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Revised: 05/22/2014] [Accepted: 06/18/2014] [Indexed: 12/18/2022]
Abstract
Complex molecular and cellular mechanisms regulate G protein-coupled receptors (GPCRs). It is suggested that proteins intrinsically disordered regions (IDRs) are to play a role in GPCR's intra and extracellular regions plasticity, due to their potential for post-translational modification and interaction with other proteins. These regions are defined as lacking a stable three-dimensional (3D) structure. They are rich in hydrophilic and charged, amino acids and are capable to assume different conformations which allow them to interact with multiple partners. In this study we analyzed 75 GPCR involved in synaptic transmission using computational tools for sequence-based prediction of IDRs within a protein. We also evaluated putative ligand-binding motifs using receptor sequences. The disorder analysis indicated that neurotransmitter GPCRs have a significant amount of disorder in their N-terminus, third intracellular loop (3IL) and C-terminus. About 31%, 39% and 53% of human GPCR involved in synaptic transmission are disordered in these regions. Thirty-three percent of receptors show at least one predicted PEST motif, this being statistically greater than the estimate for the rest of human GPCRs. About 90% of the receptors had at least one putative site for dimerization in their 3IL or C-terminus. ELM instances sampled in these domains were 14-3-3, SH3, SH2 and PDZ motifs. In conclusion, the increased flexibility observed in GPCRs, added to the enrichment of linear motifs, PEST and heteromerization sites, may be critical for the nervous system's functional plasticity.
Collapse
Affiliation(s)
- L Tovo-Rodrigues
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil; Structural Biology Unit, Integrative Neuroscience Branch, NIDA IRP, NIH, MD, United States
| | - A Roux
- Structural Biology Unit, Integrative Neuroscience Branch, NIDA IRP, NIH, MD, United States
| | - M H Hutz
- Department of Genetics, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - L A Rohde
- Child and Adolescent Psychiatric Division, Department of Psychiatry, Federal University of Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - A S Woods
- Structural Biology Unit, Integrative Neuroscience Branch, NIDA IRP, NIH, MD, United States.
| |
Collapse
|
47
|
Sheth S, Brito R, Mukherjea D, Rybak LP, Ramkumar V. Adenosine receptors: expression, function and regulation. Int J Mol Sci 2014; 15:2024-52. [PMID: 24477263 PMCID: PMC3958836 DOI: 10.3390/ijms15022024] [Citation(s) in RCA: 286] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Revised: 01/15/2014] [Accepted: 01/15/2014] [Indexed: 02/06/2023] Open
Abstract
Adenosine receptors (ARs) comprise a group of G protein-coupled receptors (GPCR) which mediate the physiological actions of adenosine. To date, four AR subtypes have been cloned and identified in different tissues. These receptors have distinct localization, signal transduction pathways and different means of regulation upon exposure to agonists. This review will describe the biochemical characteristics and signaling cascade associated with each receptor and provide insight into how these receptors are regulated in response to agonists. A key property of some of these receptors is their ability to serve as sensors of cellular oxidative stress, which is transmitted by transcription factors, such as nuclear factor (NF)-κB, to regulate the expression of ARs. Recent observations of oligomerization of these receptors into homo- and heterodimers will be discussed. In addition, the importance of these receptors in the regulation of normal and pathological processes such as sleep, the development of cancers and in protection against hearing loss will be examined.
Collapse
Affiliation(s)
- Sandeep Sheth
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Rafael Brito
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Debashree Mukherjea
- Department of Surgery (Otolaryngology), Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Leonard P Rybak
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| | - Vickram Ramkumar
- Department of Pharmacology and Neuroscience, Southern Illinois University School of Medicine, Springfield, IL 62702, USA.
| |
Collapse
|
48
|
Cardiovascular adenosine receptors: Expression, actions and interactions. Pharmacol Ther 2013; 140:92-111. [DOI: 10.1016/j.pharmthera.2013.06.002] [Citation(s) in RCA: 163] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 05/28/2013] [Indexed: 12/26/2022]
|
49
|
Park SJ, Lee KP, Kang S, Chung HY, Bae YS, Okajima F, Im DS. Lysophosphatidylethanolamine utilizes LPA(1) and CD97 in MDA-MB-231 breast cancer cells. Cell Signal 2013; 25:2147-54. [PMID: 23838008 DOI: 10.1016/j.cellsig.2013.07.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2013] [Accepted: 07/01/2013] [Indexed: 12/22/2022]
Abstract
Lysophosphatidylethanolamine (LPE) is a lyso-type metabolite of phosphatidylethanolamine (a plasma membrane component), and its intracellular Ca(2+) ([Ca(2+)]i) increasing actions may be mediated through G-protein-coupled receptor (GPCR). However, GPCRs for lysophosphatidic acid (LPA), a structurally similar representative lipid mediator, have not been implicated in LPE-mediated activities in SK-OV3 or OVCAR-3 ovarian cancer cells or in receptor over-expression systems. In the present study, LPE-induced [Ca(2+)]i increase was observed in MDA-MB-231 cells but not in other breast cancer cell lines. In addition, LPE- and LPA-induced responses showed homologous and heterologous desensitization. Furthermore, VPC32183 and Ki16425 (antagonists of LPA1 and LPA3) inhibited LPE-induced [Ca(2+)]i increases, and knockdown of LPA1 by transfection with LPA1 siRNA completely inhibited LPE-induced [Ca(2+)]i increases. Furthermore, the involvement of CD97 (an adhesion GPCR) in the action of LPA1 in MDA-MB-231 cells was demonstrated by siRNA transfection. Pertussis toxin (a specific inhibitor of Gi/o proteins), edelfosine (an inhibitor of phospholipase C), or 2-APB (an inhibitor of IP3 receptor) completely inhibited LPE-induced [Ca(2+)]i increases, whereas HA130, an inhibitor of autotaxin/lysophospholipase D, did not. Therefore, LPE is supposed to act on LPA1-CD97/Gi/o proteins/phospholipase C/IP3/Ca(2+) rise in MDA-MB-231 breast cancer cells.
Collapse
Affiliation(s)
- Soo-Jin Park
- Molecular Inflammation Research Center for Aging Intervention (MRCA), College of Pharmacy, Pusan National University, Busan 609-735, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
50
|
Weisman GA, Woods LT, Erb L, Seye CI. P2Y receptors in the mammalian nervous system: pharmacology, ligands and therapeutic potential. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2013; 11:722-38. [PMID: 22963441 DOI: 10.2174/187152712803581047] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 06/14/2012] [Accepted: 06/14/2012] [Indexed: 11/22/2022]
Abstract
P2Y receptors for extracellular nucleotides are coupled to activation of a variety of G proteins and stimulate diverse intracellular signaling pathways that regulate functions of cell types that comprise the central nervous system (CNS). There are 8 different subtypes of P2Y receptor expressed in cells of the CNS that are activated by a select group of nucleotide agonists. Here, the agonist selectivity of these 8 P2Y receptor subtypes is reviewed with an emphasis on synthetic agonists with high potency and resistance to degradation by extracellular nucleotidases that have potential applications as therapeutic agents. In addition, the recent identification of a wide variety of subtype-selective antagonists is discussed, since these compounds are critical for discerning cellular responses mediated by activation of individual P2Y receptor subtypes. The functional expression of P2Y receptor subtypes in cells that comprise the CNS is also reviewed and the role of each subtype in the regulation of physiological and pathophysiological responses is considered. Other topics include the role of P2Y receptors in the regulation of blood-brain barrier integrity and potential interactions between different P2Y receptor subtypes that likely impact tissue responses to extracellular nucleotides in the CNS. Overall, current research suggests that P2Y receptors in the CNS regulate repair mechanisms that are triggered by tissue damage, inflammation and disease and thus P2Y receptors represent promising targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Gary A Weisman
- Department of Biochemistry, 540E Life Sciences Center, 1201 Rollins Road, University of Missouri, Columbia, MO 65211-7310, USA.
| | | | | | | |
Collapse
|